effects of lead on human microglia and neuronal … · microglia interaction, as well as during cns...

223
Effects of Lead on Human Microglia and Neuronal Precursor Cells In Vitro Samar Etemad, B.Sc., M.Sc. This thesis is presented for the degree of Doctor of Philosophy The University of Western Australia School of Anatomy, Physiology and Human Biology 2013

Upload: tranque

Post on 27-Sep-2018

220 views

Category:

Documents


3 download

TRANSCRIPT

Page 1: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

Effects of Lead on Human Microglia

and Neuronal Precursor Cells

In Vitro

Samar Etemad, B.Sc., M.Sc.

This thesis is presented for the degree of

Doctor of Philosophy

The University of Western Australia

School of Anatomy, Physiology and Human Biology

2013

Page 2: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

To my parents, Ladan and Reza who taught me

to work hard and dream big with their continous

love and support.

To my fiancé, Houman, whose endless love, support,

understanding and encouragement provided me the

strength and motivation I needed to achieve my goals

during the PhD

Page 3: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[ii]

Abstract

Lead is an environmental persistent pollutant with potent neurotoxic effects. In vitro and

in vivo studies have reported that lead induces neuronal death, astrocyte activation,

cognitive deficits, as well as learning and memory impairments. Involvement of the

immune system and inflammation has also been reported in lead toxicity. In humans, lead

concentrations are difficult to measure and assess, especially for the brain. However, 10

M of lead is expected to be the highest concentration that is reached in the central

nervous system (CNS). Australia is one of the main lead mining and processing countries

with reported lead toxicity in cities close to the mining sites and ports.

Microglia are the resident innate immune cells of the brain. As the macrophages of the

CNS, microglia share common characteristics with the monocyte-macrophage lineage

such as expression of particular surface markers and chemokine/cytokine receptors,

antigen presentation and phagocytic activity. Microglia activation in response to lead

exposure has been reported in animal studies. This activation is usually followed by

secretion of pro-inflammatory cytokines and chemokines as mediators of the innate

immunity. In the CNS, chemokines and their receptors are important in the neuron-

microglia interaction, as well as during CNS development by mediating neurogenesis,

migration and differentiation of neuronal progenitor cells.

Despite being the innate immune effectors of the CNS, the role of human microglia in

lead-induced toxicity and inflammation is still unknown. Furthermore, at the

commencement of this study, there was no standardized simple model available to

investigate the biology of human microglia.

To investigate the effects of lead on human microglia, first, a new human in vitro

microglia model (M-MG) using blood-derived monocyte was developed and established

(Samar Etemad et al., Journal of Neuroscience Methods 2012, 30, 79-89). In the next

step, the effects of lead exposure on human microglia, were studied to examine in

different aspects such as morphology, phenotype and function. In addition, the human

microglia cell line HMC3 and human neuronal precursors cell line HCN2 were used as

established human microglia and neuronal precursor cell lines in this study.

The human M-MG was clearly different in morphology, phenotype and function from

monocytes and macrophages, as well as from monocyte derived dendritic cells (MDC),

but shared many properties with the established HMC3 microglia cells. The human M-

MG acquired a ramified morphology with primary and secondary processes, comparable

Page 4: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[iii]

to HMC3. They expressed very low levels of CD45, CD14 and HLA-DR, CD11b and

CD11c; but a distinct pattern of chemokine receptors, including CCR1, CCR2, CCR3,

CCR4, CCR5, CXCR1, CXCR3, CX3CR1. Similar to HMC3, under non-activated

conditions, M-MG secreted IL-8 and IL-6. In comparison with MDC, M-MG displayed

lower T-lymphocyte stimulatory capacity, as well as lower phagocytosis activity.

To investigate the effects of lead on M-MG, cells were exposed to lead acetate at

different concentrations (1 to 100 µM, 24 hours to 5 days). Then, cells were

characterized morphologically, phenotypically (chemokine receptors expression) and

functionally using flow cytometry. Interestingly, lead exposure at lower concentrations

(<25 M) was not cytotoxic and had no effect on morphology or viability of the cells.

Most of the chemokine receptors were expressed by M-MG and HMC3 microglia under

non-stimulatory conditions. General down-regulation in most of the pro-inflammatory

chemokine receptors (CCR1, CCR2, CCR3, CCR5, CXCR1, CXCR5) was observed in

both cell types. On the other hand, IL-8 was the only cytokine that was up-regulated by

lead. In that respect, it is important to understand that IL-8 plays a relevant role in neuro-

development, neuro-regeneration and neuronal turn-over.

The effects of lead exposure on expression of CX3CR1 and TLR4 were also investigated

in M-MG and HMC3 cells due to their key role in inflammation and development.

CX3CR1 expression was dimorphic in microglia derived from different individuals,

having either CX3CR1low

or CX3CR1intermediate

expressing cells, supporting the presence

of functionally relevant genetic polymorphisms in humans. The human HMC3 microglia

cell line belongs to the CX3CRlow

expressing population. Lead had a differential effect on

changes of CX3CR1 and TLR4 expression. Lead significantly increased CX3CR1

expression in CX3CRlow

cells, whereas it significantly decreased CX3CR1 expression in

CX3CR1intermediate

cells. TLR4 was expressed at low levels in all microglia populations,

but TLR4 expression was increased by lead in the CX3CR1intermediate

microglia cells.

Given the important role of the chemokine-chemokine receptor axis in neuron-microglia

interactions, the effects of lead were studied on the human neuronal precursor cell line

HCN2 under optimal conditions (10 µM, 24 hours). Interestingly, lead was not cytotoxic

and had no effects on the morphology or viability of the human neuronal cells. However,

functional changes were observed, including down regulation of the pro-inflammatory

chemokine receptors. Expression of CX3CR1 and CXCR1 significantly increased in

HCN2, as well as their reciprocal chemokines, fractalkine and IL-8. Finally, IL-6

secretion decreased in lead-treated HCN2.

Page 5: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[iv]

In conclusion, the presented data show that human monocyte-derived microglia represent

an appropriate and feasible in vitro model to investigate the role of microglia in

neurotoxicity. Furthermore, the research presented in this thesis suggests that lead has

functional effects on human microglia, but with no significant direct cytotoxic effects.

Lead alters the expression patterns of chemokines and chemokine receptors in both,

human microglia and neuronal precursor cells. Therefore, lead exposed microglia may

interfere with neuronal turnover, recruitment and differentiation, enhancing direct lead

effects on neuronal precursors.

Keywords: human microglia, lead, in vitro model, monocyte-derived microglia, IL-8,

chemokine receptor, neuronal precursor cells, CX3CR1

Page 6: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[v]

Table of contents

Abstract..................................................................................................................... ii

Table of contents ....................................................................................................... v

List of Figures ............................................................................................................ x

List of Tables ............................................................................................................. xi

Statement of Candidate Contribution ....................................................................... xii

Publications and Proceedings ................................................................................... xiii

Conference presentations ......................................................................................... xiv

Acknowledgements................................................................................................... xv

Abbreviations ......................................................................................................... xvii

Chapter 1

1. Literature Reviews ............................................................................................... 1

1.1. Lead .................................................................................................................. 1

1.1.1. Lead neurotoxicity ................................................................................................................. 7

1.1.2. Lead entry into the brain ........................................................................................................ 8

1.1.3. Mechanisms involved in lead-induced neurotoxicity: role of neuroinflammation ................. 8

1.1.4. Lead toxicity in animal models ............................................................................................ 10

1.1.5. Lead and the immune system ............................................................................................... 12

1.2. Microglia ........................................................................................................ 13

1.2.1. Origin ................................................................................................................................... 13

1.2.2. Turnover ............................................................................................................................... 14

1.2.3. Distribution .......................................................................................................................... 14

1.2.4. Morphology .......................................................................................................................... 14

1.2.5. Microglia function: immune effectors of the CNS .............................................................. 16

1.2.6. Role of microglia: neuroprotection versus neurodegeneration ............................................. 18

1.2.7. Cytokines and neuroinflammation ....................................................................................... 18

1.3. Lead and microglia ......................................................................................... 19

1.4. Role of chemokines and chemokine receptors in CNS development ............. 20

1.5. Role of cytokines, chemokines and chemokine receptors in neuro

inflammation ..................................................................................................................... 22

Chapter 2

2. Research aims and outlines ................................................................................ 26

2.1. Aims ............................................................................................................... 26

2.2. Research outline and methodology ................................................................ 28

Page 7: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[vi]

Chapter 3

3. Effect of lead on human cortical neuronal precursor cells .................................... 31

3.1. About the paper .............................................................................................. 31

3.2. Abstracts ......................................................................................................... 32

3.3. Introduction .................................................................................................... 33

3.4. Material and methods ..................................................................................... 38

3.4.1. Characterization of HCN2 .................................................................................................... 38

3.4.2. Viability assay ...................................................................................................................... 39

3.4.3. Surface marker expression ................................................................................................... 39

3.4.4. Cytokine production measurement ....................................................................................... 40

3.4.5. Statistics ............................................................................................................................... 40

3.5. Results ............................................................................................................ 41

3.5.1. Characterization of human neuronal cells HCN2 ................................................................. 41

3.5.2. Lead effects on HCN2 morphology and viability ................................................................. 43

3.5.3. Effect of lead exposure on expression of chemokine receptors in HCN2 ............................ 44

3.5.4. Effect of lead on cytokine secretion in HCN2 cells .............................................................. 47

3.6. Discussion ...................................................................................................... 48

3.7. Conclusion ...................................................................................................... 53

Chapter 4

4. A novel in vitro human microglia model: characterization of human monocyte-

derived microglia .......................................................................................................... 55

4.1. Introduction to chapter ................................................................................... 55

4.2. Abstract .......................................................................................................... 57

4.3. Introduction .................................................................................................... 57

4.4. Materials and methods .................................................................................... 61

4.4.1. Cell isolation ........................................................................................................................ 61

4.4.2. Cell culture ........................................................................................................................... 62

4.4.3. Morphological studies .......................................................................................................... 62

4.4.4. Flow cytometry .................................................................................................................... 63

4.4.5. Phagocytosis assay ............................................................................................................... 63

4.4.6. Mixed leukocyte reaction (MLR) ......................................................................................... 64

4.4.7. Statistics ............................................................................................................................... 64

4.5. Results ............................................................................................................ 64

4.5.1. Morphological and phenotypic changes of monocyte-derived microglia in culture ............. 64

4.5.2. Expression pattern of specific surface markers .................................................................... 68

4.5.3. Phagocytic capacity .............................................................................................................. 71

4.5.4. T-lymphocyte stimulatory capacity of MGM ....................................................................... 73

4.6. Discussion ...................................................................................................... 74

4.7. Conclusion ...................................................................................................... 76

Page 8: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[vii]

Chapter 5

5. Lead modulates chemokine receptors pattern and induce interleukin-8 in human

microglia ...................................................................................................................... 78

5.1. Introduction to chapter ................................................................................... 78

5.2. Abstract .......................................................................................................... 79

5.3. Introduction .................................................................................................... 80

5.4. Material and methods ..................................................................................... 82

5.4.1. Cell culture ........................................................................................................................... 82

5.4.2. Lead exposure ...................................................................................................................... 83

5.4.3. Morphological studies .......................................................................................................... 83

5.4.4. Viability assays .................................................................................................................... 84

5.4.5. Detection of up-take and intracellular accumulation of lead ............................................... 84

5.4.6. Reactive oxygen species ....................................................................................................... 85

5.4.7. Expression of surface markers.............................................................................................. 85

5.4.8. Cytokine production ............................................................................................................. 86

5.4.9. Statistics ............................................................................................................................... 86

5.5. Results ............................................................................................................ 86

5.5.1. Detection of lead uptake and intracellular accumulation by human microglia ..................... 86

5.5.2. Effect of various lead concentration on human microglia cell morphology and activation .. 88

5.5.3. Effect of various concentration of lead on human microglia viability .................................. 90

5.5.4. Effects of lead exposure on reactive oxygen species production and lipid peroxidation in

human microglia ................................................................................................................... 91

5.5.5. Effect of lead exposure on expression of chemokine receptors and surface molecules in

human microglia ................................................................................................................... 91

5.5.6. Effect of lead on cytokine secretion patterns in human microglia ........................................ 95

5.6. Discussion ...................................................................................................... 97

5.7. Conclusion .................................................................................................... 102

Chapter 6

6. Lead alters the chemokine receptors expression pattern of the human microglia

cell line (HMC3) .......................................................................................................... 104

6.1. Introduction to the chapter .......................................................................... 104

6.2. Abstract ........................................................................................................ 105

6.3. Introduction .................................................................................................. 106

6.4. Material and methods ................................................................................... 109

6.4.1. Cell culture ......................................................................................................................... 109

6.4.2. Lead exposure .................................................................................................................... 109

6.4.3. Morphological studies ........................................................................................................ 109

6.4.4. Detection of up-take and intracellular accumulation of lead .............................................. 110

6.4.5. Viability assays .................................................................................................................. 110

Page 9: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[viii]

6.4.6. Expression of surface markers............................................................................................ 111

6.4.7. Mixed leukocyte reaction (MLR) ....................................................................................... 111

6.4.8. Phagocytosis assay ............................................................................................................. 112

6.4.9. Cytokine measurement ....................................................................................................... 112

6.4.10. Statistics ............................................................................................................................. 112

6.5. Results .......................................................................................................... 113

6.5.1. Intracellular detection of lead uptake in human microglia cell line HMC3 ........................ 113

6.5.2. Effect of low level lead exposure on the morphology and viability of HMC3 ................... 114

6.5.3. Effect of of lead exposure on expression of chemokine receptors and surface molecules on

HMC3 ....................................................................................................................... ..... 116

6.5.4. Effects of lead on phagocytic activity of HMC .................................................................. 120

6.5.5. Influence of lead on microglia T-cell stimulatory capacity ................................................ 121

6.5.6. Effect of lead on cytokine secretion in HMC3 ................................................................... 121

6.6. Discussion .................................................................................................... 124

6.7. Conclusion .................................................................................................... 128

Chapter 7

7. Differential effects of lead on CX3CR1 and TLR4 expression in human microglia in

vitro 130

7.1. Introduction to chapter ................................................................................. 130

7.2. Abstract ........................................................................................................ 131

7.3. Introduction .................................................................................................. 132

7.4. Material and Methods ................................................................................... 135

7.4.1. Cell culture and experimental setting ................................................................................. 135

7.4.2. Flow cytometry .................................................................................................................. 136

7.4.3. Statistical analysis .............................................................................................................. 136

7.5. Results .......................................................................................................... 137

7.5.1. Differential expression of CX3CR1 in human microglia in vitro ....................................... 137

7.5.2. Differential effects of lead on CX3CR1 expression in human microglia ........................... 138

7.5.3. Effect of immune modulators on CX3CR1 expression in HMC3 cells .............................. 140

7.5.4. Effect of lead on TLR4 expression human microglia ......................................................... 140

7.6. Discussion .................................................................................................... 142

7.7. .Conclusion ................................................................................................... 144

Chapter 8

8. General discussion and future directions .......................................................... 146

8.1. Summary of key findings ............................................................................. 146

8.2. Neuronal remodeling and lead toxicity ........................................................ 151

8.3. Why microglia are important in lead-neurotoxicity? ................................... 152

8.4. Microglia-neuron interaction: A possible target of lead toxicity ................. 153

8.5. Role of IL-8 / CXCR1 signaling in microglia-neurons interaction: ............. 154

Page 10: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[ix]

8.6. Importance of fractalkine/CX3CR1 in microglia-neuron interaction ........... 155

8.7. Immune modulation by lead and IL-6 in the CNS: between neuroprotection

and neuroinflammation ................................................................................................... 157

8.8. Research implication and future direction .................................................... 158

8.9. General conclusion ....................................................................................... 159

Chapter 9

Bibliography .......................................................................................................... 161

Chapter 10

Appendix ............................................................................................................... 186

Page 11: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[x]

List of Figures

Figure 1.1: Lead mining and processing centers in Australia. .......................................... 2

Figure 1.2: Effects of lead at different concentration on children vs. adults. ................... 3

Figure 1.3: The main sources of lead poisoning. .............................................................. 5

Figure 1.4: Lead toxicity targets in the human body......................................................... 6

Figure 1.5: Lead accumulation, storage and release in astrocytes. ................................... 9

Figure 1.6: Lead interacts with different sites of actions in neurons. ............................. 10

Figure 1.7: Microglia is the immune effectors cells in the brain. ................................... 15

Figure 1.8: Chemo taxis or phagocytosis-involved receptors in microglia and

correlation of the inflammatory or anti-inflammatory response. ..................................... 17

Figure 1.9: Constitutive vs. inducible chemokine ........................................................... 20

Figure 2.1: Research outline ........................................................................................... 29

Figure 3.1: Expression of neuronal markers by HCN2. .................................................. 42

Figure 3.2: Effect of various concentrations of lead on viability of HCN2 using the

MTS assay. ....................................................................................................................... 43

Figure 3.3: Effect of lead exposure on HCN2 morphology ............................................ 44

Figure 3.4: Effect of lead exposure on expression pattern of neural marker. ................. 45

Figure 3.5: Effect of lead on expression patterns of chemokine receptor in HCN2. ...... 46

Figure 3.6: Effect of lead on cytokine secretion in HCN2. ............................................. 47

Figure 4.1: Phase contrast microscopy of monocyte-derived microglia (M-MG) and

human microglia cell line HMC3 ..................................................................................... 65

Figure 4.2: Expression of Iba-1 in HMC3 and M-MG ................................................... 66

Figure 4.3: Differential expression of CD68 in freshly isolated blood monocytes ,

HMC3 and M-MG............................................................................................................ 67

Figure 4.4: Expression of surface markers CD45 (a), CD14 (b), HLA-DR (c), CCR2

(d) and CCR4 (e) measured with flow cytometry. ........................................................... 69

Figure 4.5: Expression of chemokine receptors CCR1 (a), CCR5 (b), CXCR1(c),

CXCR3 (d) and CX3CR1 (e) measured with flow cytometry. ...................................... 70

Figure 4.6: Phagocytic capacity of M-DC (Black line), M-MG (Dotted line) and

HMC3 (dashed line) towards TM Rhodamine-labelled S. aureus measured with flow

cytometry. ........................................................................................................................ 71

Figure 4.7: Up-take of TM Rhodamine-labelled S. aureus by M-MG using confocal

microscopy. ...................................................................................................................... 72

Figure 4.8: A selected Z-stack image taken with confocal microscopy showing the

up-take of TM Rhodamine-labeled S. aureus by M-MG. ................................................ 72

Page 12: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[xi]

Figure 4.9: Mixed leukocyte reactions (MLR) ............................................................... 73

Figure 5.1: Lead uptake by human microglia. ................................................................ 87

Figure 5.2: Effect of lead exposure on morphology of microglia. .................................. 89

Figure 5.3: Viability of human microglia after lead exposure. ....................................... 90

Figure 5.4: Effect of lead on ROS production and membrane peroxidation in human

microglia. ......................................................................................................................... 92

Figure 5.5: Effect of lead exposure (10 µM, 24 hours) on chemokine receptors and

surface marker expression patterns in human microglia as measured with flow

cytometry. ........................................................................................................................ 93

Figure 5.6: Comparison between expression pattern of surface markers in the absence

or presence of lead in human microglia ........................................................................... 94

Figure 5.7: Effect of lead on cytokine secretion in human microglia. ............................ 96

Figure 6.1: Lead uptake by human microglia cell line HMC3......................................113

Figure 6.2: Effect of lead on morphology and Viability of HMC3. .............................. 115

Figure 6.3: Effect of lead exposure (10 µM, 24hrs) on chemokine receptor expression

patterns in human microglia HMC3. .............................................................................. 117

Figure 6.4: Lead alters the expression of surface markers in HMC3 ............................ 118

Figure 6.5: Lead showed distinct immunomodulatory effects on the surface molecule

expression in HMC3. ..................................................................................................... 119

Figure 6.6: Effects of lead on phagocytic activity of HMC3 ........................................ 120

Figure 6.7: Influence of lead on microglia T-cell stimulatory capacity. ....................... 122

Figure 6.8: Effect of lead on cytokine secretion in HMC3 ........................................... 123

Figure 7.1: Expression levels of CX3CR1 in human M-MG and HMC3 cells. ............ 138

Figure 7. 2: Effect of lead on expression of CX3CR1 in human M-MG and HMC3 .. 139

Figure 7. 3: Comparison of various stimuli on CX3CR1 expression in HMC3 cells. .. 140

Figure 7. 4: Effect of lead on expression of TLR4 in human M-MG and HMC3 cells. 141

Figure 8.1: Neuromodulatory effects of lead on neurounal precursors and microglia . 160

List of Tables

Table 1.1: Chemokine and chemokine receptors in CNS.............................................. 24

Table 2.1: Testing hypotheses of microglia involvement in lead-induced

neurotoxicity. ................................................................................................................. 27

Table 2.2: The main aims of this thesis. ........................................................................ 27

Table 2.3: Main methods applied in this study and the corresponding chapters. .......... 28

Page 13: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[xii]

Statement of Candidate Contribution

All the experiments detailed in this thesis have been conducted at the School of

Anatomy, Physiology and Human Biology; University of Western Australia under the

supervision of Prof. Luis Filgueira, Dr. Marc Ruitenberg and A/Prof. Silvana Gaudieri. I

hereby declare that all the works presented here is entirely my own, except where the

contribution of others have been acknowledged.

Samar Etemad

Dec. 2013

Page 14: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[xiii]

Publications and Proceedings

PUBLICATIONS

Etemad S., Ruitenberg M.J. and Filgueira L. (2013). Differential effects of lead (Pb)

on CX3CR1 and TLR4 expression in human microglia in vitro. Journal of

Neuroinflammation (under review).

Etemad S., Ruitenberg M.J. and Filgueira L. (2013). Lead (Pb) modulates chemokine

receptor patterns and induces interleukin-8 in human microglia. Journal of

Neurotoxicology (under review).

Etemad S., Zamin, R.M., Ruitenberg M.J. and Filgueira L. (2012). A novel in vitro

human microglia model:Characterozation of human monocyte-derived microglia.

Journal of Neuroscience Methods 209,79-89 (Appendix E).

Etemad S., Ruitenberg M.J. and Filgueira L.(2012), In vitro generated microglia:

from blood precursors. Journal of Immunology; 118:111.14 (Meeting Abstract).

Page 15: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[xiv]

Conference presentations

ORAL PRESENTATIONS

S. Etemad, M. Ruitenberg and L. Filgueira (2013), Lead (Pb) modulates human microglia

inflammatory responses. Australian Society of Neuroscience (ANS), Melbourne,

Australia, February 3-6.

S. Etemad, M. Ruitenberg and L. Filgueira (2012), Effect of low-level lead exposure on

chemokine receptor expression in human microglia in vitro .School of Anatomy,

Physiology and Human Biology (APHB) Student Expo, Perth, Australia, July 21.

S. Etemad, M. Ruitenberg and L. Filgueira (2011), Effect of Lead on human microglia in

vitro. APHB Student Expo, Perth, Australia, July 15

S. Etemad, M. Ruitenberg and L. Filgueira (2010) A new in vitro model for human

microglia. ASMR (Australian Society for Medical Researches) meeting, Curtin

University, June 8.

S. Etemad, M. Ruitenberg and L. Filgueira (2010), A New In vitro Model for Human

Microglia. School of Anatomy, Physiology and Human Biology Student Expo, Perth,

Australia, July 15

S. Etemad, M. Ruitenberg and L. Filgueira (2009). Role of human microglia in lead

neurotoxicity. School of Anatomy, Physiology and Human Biology Student Expo, Perth,

Australia, July 16.

POSTER PRESENTATION

S. Etemad, M. Ruitenberg and L. Filgueira (2012), Effect of low-level lead exposure on

chemokine receptor expression using a novel human in vitro microglia model. Combined

Biological Sciences Meeting (CBSM), Perth, Australia, August 24. (Appendix A)

S. Etemad, M. Ruitenberg and L. Filgueira (2012), In vitro generated microglia: from

blood precursors. Immunology 2012 (99th American Association of Immunology),

Boston, MA, USA, May 4-9 . (Appendix B)

S. Etemad, M. Ruitenberg and L. Filgueira (2012), In vitro generated microglia derived

from blood precursors. Experimental Biology (American Association of Anatomists),

SanDiego, CA, USA, April 21-25. (Appendix C)

S. Etemad, M. Ruitenberg and L. Filgueira (2010), A new in vitro human microglia

model. 41st Annual Scientific Meeting of the ASI (Australian Society of Immunology),

Perth, Australia, December 5-9. (Appendix D).

Page 16: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[xv]

Acknowledgements

I would like to express my sincere appreciation for my main supervisor; Prof. Luis

Filgueira. I specially like to thank him for giving me the opportunity to come to Australia

and experience new life here. Luis had provided me his enduring guidance and support

during each and every stage of this project. Luis, you have helped me to become a better

scientist and you have taught me to believe in my ideas. Thank you also for the

opportunity to meet many top researchers in the field of Anatomy, Human Biology and

Immunology.

I also wish to extend my deepest gratitude to my supervisor Dr. Silavana Gaudieri. It has

been a great privilege for me to work under her supervision in particular over the last

year of PhD in absence of Luis.

A very special thank you to my co-supervisor, Dr. Marc Ruitenberg for his invaluable

advice and feedback on my research and for always being so supportive of my work

I particularly want to say thanks to my dear friends and PhD colleagues of School: Su-

ann Koh, Helen Dooley, Belinda Burns, Fritha Milne, Rebecca Dawson and Linda

Blomster who all helped me in numerous ways during various stages of my PhD Thanks

for your continuous support and encouragement. You made memorable moments during

my PhD and I will never forget.

Special thanks to my colleagues, Praseetha Prabhakaran, Pooja Deshpande and the staff

and postgraduate students in School of Anatomy, Physiology, and Human Biology

(APHB) for their encouragement. I would like to extend my thanks to Rasheeda Mohd

Zamin and acknowledge her assistance in confocal imaging.

I would like acknowledge the facilities (confocal microscopy and flow cytometry),

scientific and technical assistance of the Australian Microscopy & Microanalysis

Research Facility at the Centre for Microscopy, Characterization & Analysis, the

University of Western Australia. In particular, my deep appreciation goes to Dr. Kathy

Heel and Mrs. Tracey Lee-Pullen for their support and help.

My thanks also go out to the support I received from Prof. Link Schmitz and all the

academics of AHAB for their support and encouragement throughout my PhD . . . I am

indebted to Prof. Alan Harvey and Margret Pollett who accepted me as one of their

team membersand Margaret provided me with her constant support and advice regarding

neuronal cell culture as well as enabling me to use neuronal cell line.

I am also very grateful to all those at the APHB office, especially Vicki, Heather, Denis,

Debbie and others who were always so helpful and provided me with their assistance

throughout my dissertation.

I should say thanks to Cell Central and in special Guy Ben- Ary, center for microscopy at

APHB for providing the microscopic facilities. Thanks Greg Cozen, Mary Lee, Leonie

Page 17: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[xvi]

Khoo, and Steve Bamfort for your always help, advice and patience in answering my

questions. I also appreciate the Graduate Research School, The University of Western

Australia for supporting this project and me all these years.

On a personal note, I like to express my heartiest appreciation to my Mum and Dad for

their support beyond measures during all stages of my life in particular during the PhD.

Their everyday encouragement, optimisms, and strength made to overcome the obstacle

and without them I would not have contemplated this road. I am very lucky to have

parents, who understand the difficulties of research and science.

Finally, my very special thank is to Houman for his endless love, understanding,

encouragement, support and optimisms during my PhD, I don’t know how should I say

thank you. You were with me, with positive opinion and hope in every parts of these PhD

during happy and sad times. I am so lucky to have you in my life, one who understands

the real meaning of research and PhD. It would have not been possible without you.

Page 18: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[xvii]

Abbreviations

AD

Alzheimer’s Disease

ALS

APCs

Amyotrophic Lateral Sclerosis

Antigen Presenting Cells

BBB Blood-Brain Barrier

BDNF

CCL

Brain-Derived Neurotrophic Factor

Chemokine (C-C Motif) Ligand

CCR C-C Motif Chemokine Receptor

CNS

CXCL

CXCLR

DG

Central Nervous System

Chemokine C-X-C motif Ligand

Chemokine C-X-C motif Receptor

Dentate Gyrus

FKT Fractalkine/CX3CL1

GM-CSF

GFAP

Granulocyte-Macrophage Colony Stimulating Factor

Glial Fibrillary Acidic Protein

HLA-DR

HMGB1

Human Leukocyte Antigen-DR

High Mobility Group Box 1 Protein

HMC3 Human Microglia Cell Line Clone 3

Iba-1 Ionized Calcium Binding Adaptor Molecule 1

IL Interleukin

iNOS

ISF

LIF

Inducible Nitric Oxide Synthase

Interstitial Fluids

Leukaemia Inhibiting Factor

LPS

MBP

Lipopolysaccharide

Myelin Basic Proteins

MCP-1

MIP-1

Monocyte Chemoattraactant Protein 1

Macrophage Inflammatory Protein 1

M-CSF Macrophage Colony Stimulating Factor

M-DC Monocyte Derived Dendritic Cells

M-MG Monocyte-Derived Microglia

MS Multiple Sclerosis

NGF Nerve Growth Factor

NGFR Nerve Growth Factor Receptor

NPCs Neuronal Precursor Cell

Page 19: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[xviii]

NSCs

NSE

Neural Stem Cells

Neuronal Specific Enolase

PBMC Peripheral Blood Mononuclear Cells

PBS Phosphate Buffer Saline

PC12 Pheochromocytoma

PD Parkinson’s Disease

PFA Paraformaldehyde

PHA Phytohemagglutinin

PMA Phorbol-12-Myristate-13-Acetate

PVR

RANTES

SDF-1

SVZ

Proliferative Viteorethinopathy

Regulated And Normal T Cell Expressed And Secreted

Stromal Derived Factor 1

Subventricular Zone

TGF-β

TECK

Transforming Growth Factor Beta

Thymus Expressed Chemokine

TLR4 Toll-Like Receptor 4

TNF-

TGF-β

Tumor Necrosis Factor Alpha

Transforming Growth Factor Beta

Trk A Tyrosine Kinase Receptor A

Page 20: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal
Page 21: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

Chapter 1

Literature Reviews

Page 22: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[1]

1. Literature Reviews

1.1. Lead

Lead is a persistent, widely spread environmental pollutant with potent toxic effects in

humans. Lead is a xenobiotic metal with many still unknown effects on cell growth,

proliferation and differentiation. Despite continuous attempts to reduce lead exposure in

humans, it is still a major global health concern. Lead can be naturally found in the

environment. However, human activities over the past three decades have increased the

lead level more than 1000 fold (NHMRC, 2009). Based on sources from the ancient

history literature, lead was known to humans as early as 4,000 BC. It has been used by

humans for thousands of years due to its wide geographical availability, easy mining and

enrichment, as well as convenient processing and usage. Lead is often found in ore with

other metals such as silver, zinc and copper which are usually extracted together. The

ancient Romans regarded lead as the father of all metals. However, Romans were aware

that lead could cause serious health problems such as madness and death (Woolley,

1984). Thus, lead and its toxicity have affected humans for many years. More recently,

lead has been widely used in building constructions, batteries, army equipment including

bullets and weapons, ceramic glazes and colouring, house paint, piping, and petrol

(Hernberg, 2000, Woolley, 1984). Epidemic incidents of lead poisoning during the 19th

century up until the mid-twentieth century, due to industrializing the world and changes

in human lifestyles, led to increased need for understanding and detection of lead

exposure sources. Thus, legislation was established in many countries in order to reduce

the level of environmental exposure from lead. For example in the 1920s, Austria,

Czechoslovakia, Finland, Norway, Poland, Spain and Sweden, abandoned the use of lead

in indoor white painting due to frequent toxic incidents in children (Hernberg, 2000).

Australia is one of the principal countries in lead mining, smelting and processing. Lead

was first mined in Australia in 1841 near Adelaide, South Australia. Hence, people

living next to lead-mining, smelting, and refining centers such as those in Broken Hill,

Port Pirie, and Mount Isa have been exposed to lead for a long time (Figure 1.1). In

1904, for the first time, lead poisoning has been reported in children from lead-based

paint in Brisbane by the Australian physician Lockhart Gibson (Rosner et al., 2005).

Later, it was shocking to discover that more than 3000 children in the town of Port Pirie,

South Australia, were reported lead poisoned in 1980-1990. Due to ignorance or

misinformation, generations of families living in lead mining and processing regions

have been and continue to be exposed to high environmental lead concentrations, despite

Page 23: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[2]

knowing about its neurotoxicity. A survey in Mt Isa , Queensland Health has shown

that 11% of children had blood lead concentrations in excess of 10 μg/dl (Health, 2008).

Similar to humans, over the past decade, several reports have shown lead poisoning in

animals such as birds and dolphins living close to the mining area in Western and South

Australia (Lavery et al., 2008).It is widely known that children, at the age of 1-5 years,

are a very vulnerable group (Finkelstein et al., 1998). In young children, lead

intoxication causes dramatic problems, including deficits in learning and memory,

behavioural disturbance and decreased IQ scores, even at low exposure levels (Nava-

Ruiz et al., 2012, White et al., 2007).

Figure 1.1: Lead mining and processing centers in Australia.

This map illustrates the many lead mining centers scattered throughout Australia. Sever lead

poisoning (BLL more than 10 µg/dl) in children living in the marked places are indicated by

black stars. Source: http://www.lead.org.au/lanv12n3/lanv12n3-2.html

Based on recent research and reports, it is not possible to determine safe levels of lead

exposure. In 1960-1990, due to the extensive world-wide efforts to decrease

environmental lead levels, average blood lead levels (BLL) were significantly reduced

from 60 to 25 µg/dl in most human populations. However, in 1991, the Centres for

Disease Control and Prevention (CDC) recommended lowering the BLL in individuals

to 15 µg/dl and started a world-wide lead poisoning prevention campaign in areas where

children had BLL > 10 µg/dl. In 2005, the CDC considered blood lead concentrations

greater than or equal 10 µg/dl (0.5 µM) excessive for infants and children (CDC),

Page 24: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[3]

2005).The CDC has set the standard elevated blood lead level for adults to be 25 µg/dl in

whole blood. However, according to recent reports about the toxic effects of lead,

reduction of recommended upper BLL in children has been now set at 5 µg/dl. The

Advisory Committee on Childhood Lead Poisoning Prevention (ACCLPP) in the United

States of America reported deficits in cognitive functions, IQ and academic skills, as

well as impairment in cardiovascular, immunological and endocrine functions in

children with BLL lower than 10 µg/dl (Figure 1.2).

Figure 1.2: Effects of lead at different concentration on children vs. adults.

This figure provides a summary of the organ-specific effects associated with different

concentrations of lead as well as physiological disorders in children versus adults. Source:

Preventing Lead Poisoning in Young Children, CDC, 1991, Gurer and Ercal, 2000.

Thus, they proposed the new upper references value of maximally 5 µg/dl as the base

line for lead poisoning in children (Prevention, 2012). Based on evidences on the effect

of low- level lead exposure, the Australian National Health and Medical Research

Council (NHMRC) suggested in 2009 that all Australians should have BLL below 10

μg/dl (NHMRC, 2009).Environmental lead is taken up through inhalation, ingestion and

dermal contact. Lead absorption in humans happens mainly through the respiratory and

gastrointestinal systems (Papanikolaou et al., 2005). Inhaling lead dust, as a result of

mining and smelting of lead, is still common in many countries, including Australia.

However, using lead in petrol was stopped in most countries by 2007. Lead

50 100 150

<

>5 10 20 40

Blood Lead

Level

learning disabilities

cognitive deficits

reduced IQ

neurobehavioral

disorders

encephalopathy

medical lead removal

chilation therapy

medical lead removal

chilation therapy

colic

anaemia

nephropathy

encephalopathy

death

Fatigue abdominal pain

Abdominal pain anaemia reproductive

problem

Reduced poor appetite

Sleep deprivation memory problem

Aggressive behaviour nausea

Headache kidney problem

nerve conducttion

velocity

hemoglubulin synthase

Vitamin D methabolisms

Page 25: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[4]

contaminated soil or water, via lead pipes, are other common sources of exposure. Lead-

based paint has been a major sources of lead exposure in children (Figure 1.3). Lead

paint tastes sweet, and thus encourages children to lick the paint. It can also easily be

peeled off walls or toys , and enters the infants’ or children’s body hand-to-mouth

(Schwartz and Levin, 1991). Approximately 40% of the inhaled lead enters the

circulation system (Philip and Gerson, 1994).

Adults absorb 10-15% of lead through the gastrointestinal tract, while infants and

children absorb as much as 50% through their gastrointestinal tract (Markowitz, 2000,

Papanikolaou et al., 2005, Goldstein, 1990a). Depending on the target tissue, lead can

persist for many years. For example the half-life of lead in the brain is about two years,

while it remains in bone for about 30-40 years (Verstraeten et al., 2008). Lead in blood

has a half-life of 35 days (Xu et al., 2012). Generally, the rate of lead transferred from

blood to soft tissue is slow and takes about 4-6 weeks. In the human body, lead has

detrimental effects on several important systems, such as the renal, reproductive,

cardiovascular, immune and central nervous system, even at low levels of exposure

(Figure 1.4). In children, the adverse effects of lead can vary from changes in cognitive

function to fatal lead poising depending on the amount of exposure (Needleman, 2004a).

It is noteworthy to mention that lead excretion from the body happens very slowly,

mainly through urinary excretion. Therefore, chelating agents are the best agents to

enhance lead excretion, these are currently being used in the therapeutic approach for

lead poisoning treatment.

Page 26: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[5]

Figure 1.3: The main sources of lead poisoning.

This image illustrates the most common sources of lead exposure: soil, drinking water, lead based

paint and leaded petrol. In Australia, due to the mining industries, lead-contaminated soil is one of

the main sources of exposure. Lead-based paint is another common source of lead exposure

especially among preschoolers. Children may eat, chew paint chips, or even lick the old-painted

window slides. Drinking water is usually contaminated with lead through lead water pipes, found

in many homes built before 1930.

Australia is one of the main leading countries in mining, smelting and processing

the lead

Page 27: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[6]

Figure 1.4: Lead toxicity targets in the human body.

The detrimental effects of lead can affect several organs during development and adulthood. The

central nervous system and the immune system are key targets of lead poisoning in human. Source:

WHO, 2010.

Page 28: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[7]

1.1.1. Lead neurotoxicity

The central nervous system (CNS) is one of the primary targets of lead toxicity,

especially during development. Lead can cross the blood–brain barrier (BBB) and

concentrates in the gray matter of the brain (Goyer and Clarkson 2001; Gwalteney-Brant

2002). Lead exposure has devastating effects on the developing nervous system,

producing morphological, cognitive, and behavioural deficits. Headaches, poor attention

span, irritability, loss of memory and dullness are early symptoms of lead exposure in the

central nervous system. High level exposure to lead, which results at blood lead levels of

70-100 µg/dl in children, may cause serious clinical encephalopathy or severely affect

neurological functions. Persistent vomiting, ataxia, stomach pain and loss of

consciousness are symptoms of acute encephalopathy (Kanwal and Kumar, 2011).

Furtunately, occurrence of this kind of lead poisoning is now extremely rare. Acute adult

lead poisoning occurs primarily as the result of inhalation or ingestion of lead particles

due to working in lead mines or smelting sites.

Despite the well-documented neurotoxic effects of lead, the mechanisms involved are not

fully understood. As lead affects numerous biological activities at different levels, several

cellular, intracellular and molecular mechanisms have been shown to be involved in lead

neurotoxicity. The prefrontal cerebral cortex, hippocampus and cerebellum have been

reported as preferred target regions in the brain for lead-induced damages (Selvin-Testa

et al., 1994). Based on observations from animal and human in vitro and in vivo models,

a number of neurotoxic effects of lead on the brain, especially during development, have

been observed. These include impaired synaptic plasticity, inhibition of glutamate

receptor activation (Guilarte et al., 1994), apoptosis (Oberto et al., 1996), learning and

cognitive deficits, memory impairment (Xu et al., 2009), excitotoxicity, altered

neurotransmitter storage and release, mitochondrial dysfunction, loss of neuronal myelin

sheaths, changes in cellular energy metabolism and disturbance in neuronal growth

(Toscano and Guilarte, 2005). Several studies have demonstrated the neurotoxic effects

of different metals, including zinc, mercury, and aluminum, on cortical neurons survival

and function, as well as their involvement in pathology of neurological and psychiatric

disorders, including autism spectrum disorders, Alzheimer’s disease, Parkinson’s disease,

epilepsy, depression and mood disorders (Xu et al., 2012, Chen and Liao, 2003).

Although there are several studies showing the developmental effects of low-levels of

lead exposure on neurons, such as reduction of the capacity of neurogenesis in the adult

rat hippocampus, deficits in synaptic plasticity, and effects on migration of the human

neural crest cells, (Gilbert et al., 2005, Zimmer et al., 2012), the mechanisms involved in

Page 29: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[8]

the effect of low-level exposure of lead on the CNS are still not known. Recent studies

have reported that lead could enhance neuron degeneration in cultured rat

cerebrocortical neurons without inducing neuronal cell death (Fujimura and Usuki,

2012).

1.1.2. Lead entry into the brain

The blood-brain barrier (BBB) is a specialized structure in the CNS, which separates the

systemic blood circulation from the interstitial fluids (ISF) and therefore protects the

brain’s microenvironment from fluctuations in ion and metabolite concentrations in the

blood (Zheng et al., 2003). The BBB is made up of the endothelial cells surrounded by

astrocytes. Toxicity of lead play a key role in the disruption of the astrocytes-endothelial

cell communication (Goldstein, 1990). Various receptor- mediated transport systems are

inherent to the BBB vascular structure in order to control the transport of metals within

the brain. The BBB has long been known as one of the targets of lead toxicity. Poisoning

by lead and other metals has been demonstrated to induce vascular destruction and

cerebral haemorrhage (Zheng et al., 2003). Lead may interact with astrocytes to

indirectly affect the BBB function (Zheng et al, 2003). Astrocytes are also known as the

metal depot cells of the brain.

Several studies suggest that lead enters the brain as a free ion or as a complex with small

molecules. In plasma, around 40% of lead is bound to albumin which stops brain lead

uptake, as albumin does not preferably cross the BBB. Lead may however, cross the

BBB by a passive process as an ion, such as Pb-OH+. In vitro studies of brain capillary

endothelial cells also revealed the involvement of the Ca-ATPase pump in the transport

of lead into the brain (Bradbury and Deane, 1993).

1.1.3. Mechanisms involved in lead-induced neurotoxicity: role of

neuroinflammation

Numerous research studies support the hypothesis that lead has an effect on altering the

cellular second messenger systems in neurons and astrocytes (Figures 1.5 and 1.6). The

ability of lead to substitute for Ca2+

the most common second messenger, is one of the

main proposed lead neurotoxicity mechanisms. It has been shown that lead enters

astrocytes and neurons via voltage sensitive calcium channels. Increased depletion of

stored Ca2+

, followed by lead exposure in HEK293 cells or glial C6 cells has been

reported, and suggests the possible of substitution of calcium by lead in these cells

(Bressler et al., 1999b). Perturbation of Ca2+

homeostasis, dysfunction of mitochondria,

Page 30: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[9]

glutamatergic excitability, disintegration of β-tubulin III protein and reactive oxygen

species (ROS) have been reported as several possible mechanisms in metal-induced

neuronal death. However, a recent study showed that the lead or Aβ-induced neurotoxic

mechanism was different to neuronal apoptosis or neuritic degeneration induced by other

metals or neurotoxicants (Fujimura and Usuki, 2012).

Figure 1.5: Lead accumulation, storage and release in astrocytes.

This diagram shows the possible mechanism involved in entry of lead into the cells, binding

with intracellular organelles, and neurotoxicity. Source: Tiffany-Castiglioni and Qian, 2001.

It has been shown that neuritic degeneration induced by exposure to 10 µM lead was not

associated with neuronal cell death (Fujimura and Usuki, 2012b). Lead can also

negatively affect the survival of neurons through changes in expression of neuronal cell

adhesion molecules. It has been well-documented that in the brain, lead interacts with a

variety of molecules involved in cell signalling pathways. Lead exposure has been shown

to stimulate the ERK1/2 and p38 MAPK

pathway in the hippocampus of the immature rats

in both in vivo and in vitro (Cordova et al., 2004). Induced apoptosis and alteration in

neurotrophic factors by lead has also been reported in the developing rat brain (Chao et

al., 2007). The ability of lead to substitute for zinc may also contribute to

neurodevelopmental toxicity. Lead can accumulate in the cell nuclei and can interfere

with the functioning of gene regulatory proteins (Hanas et al., 1999). Free zinc

concentration in microglia decreased after being challenged with lead 24 hours earlier,

which correlates with the previously mentioned mechanisms (personal communication).

In astrocytes, induced cytotoxicity and cell activation with enhanced cell detachment and

Page 31: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[10]

nuclear inclusion has been reported after exposure to lead. In addition, lead could

enhance the expression of astrocyte activation markers, glial fibrilary acidic protein

(GFAP) and S-100β protein, and induces astrocyte activation (Struzynska et al., 2007b).

Some studies suggested that lead may induce TNF-α expression, similar to

lipopolysaccharide (LPS) through activation of the PKC pathways (Cheng et al., 2004).

There is a possibility that lead suppresses the immune response through inhibiting the

signalling pathways involved in microglia activation. However, the exact molecular

mechanisms involved in lead–induced neurotoxicity/neuroinflammation are not

fully understood.

Figure 1.6: Lead interacts with different sites of actions in neurons.

This diagram demonstrates that lead can enter into the neuron through voltage channels, alter the

intracellular Ca2+

balance, affect cell signaling and interfere with the nucleus and damage DNA

and RNA. Source: Finkelstein et al. 1998.

1.1.4. Lead toxicity in animal models

Despite several studies on toxic effects of lead, understanding the underlying

mechanisms have generated much interest over past decades. The differential toxic

effects of lead exposure in young children and adults, as well as, inconsistency between

in vitro and in vivo models, imply the involvement of multiple mechanisms in lead-

induced toxicity. Due to the versatile effects of lead poisoning on human health,

Page 32: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[11]

particularly on nervous system functions, to study lead neurotoxicity using experimental

animal models is a convenient approach. Animal models are the beneficial systems in the

interpretation of behavioural and neurobiological mechanisms involved in observed

deficits in lead-exposed humans (Bellinger, 2004). The role of lead toxicity in the

pathogenesis of several neurodegenerative diseases, including Alzheimer’s disease and

Parkinson’s disease, has also been shown using animal models (Charlet et al., 2012). In

addition, involvement of microglia activation has been reported in inflammation-

mediated neurodegenerative diseases (Liu et al., 2012). Based on recent studies, lead

could induce the glial cell reactivity as a hallmark of brain inflammation. Enhanced

expression of beta amyloid precursor protein by lead has also been suggested as one

possible mechanism underlying the pathogenesis of Alzheimer’s disease (AD) (Monnet-

Tschudi et al., 2006). In vitro and in vivo animal experiments have revealed the dramatic

effects of lead on growth and development of the CNS. Studies in rat and mice show that

absorption of lead in pups is greater than in adults. Reduction in the number of neurons,

glia cells, as well as synapses per neurons was reported in lead-exposed pups. Pro-

inflammatory cytokine gene expression, IL-6 and TGF-β1, increased in the whole brain

of mice exposed to lead (Kasten-Jolly et al., 2011). Increased gene expression of IL-6

and TGF-β1 during development may adversely affect the growth and development of

neurons. In the hippocampus of developing rats, lead caused an increase in apoptosis

through suppressing the expression of apoptotic inhibitor proteins (Liu et al., 2010). Lead

exposure was also found to reduce hippocampal long-term potentiating. Recent studies

showed the developmental effects of lead on behaviour and gene expression patterns in

BALB/c mice. Lead caused significant impairment of spatial memory tested using the

Morris water maze (Kuhlmann et al., 1997). Lead-exposed mice manifested abnormal

exploratory behavior compared to controls in the activity monitor. However, lead showed

no changes in motor function (Gilbert et al., 2005). Lead differentially affected the

behavior of male and female mice. Lead-exposed males showed more violent behavior

towards the same cage mates (Gilbert et al., 2005). At the gene expression level,

developmental lead-exposure showed perturbation in pathways associated with region

formation in the developing brain. Lead has a high impact on genes related to the

immune system; for instance, increased transcription of genes for MHC protein and

decreases expression level of IL-10 in the cortex of lead-exposed mice were reported

(Kasten-Jolly et al., 2012). Altered cytokine gene expression patterns, ROS generation

and lipid peroxidation have been reported in developmental lead toxicity at relatively

low, physiological doses (Kasten-Jolly et al., 2011).

Page 33: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[12]

In vitro studies, using the rat PC12 neuronal cells, showed that lead could induce

apoptosis and DNA damage through changes in the expression of apoptotic genes. Lead-

induced neurotoxicity has been reported in a dose- dependent manner in PC12 (Xu et al.,

2006). Furthermore, mitochondrial activity and synaptosomal neurotransmitter decreased

in the lead-exposed rat brain (Devi et al., 2005).

1.1.5. Lead and the immune system

Immune modulatory effects of heavy metals, such as lead, zinc and copper, have been

reported previously, although not much is still known about the underlying mechanisms.

Changes in immune responses following lead exposure have been shown using both

animal and human models. For example, in vitro exposure of murine spleenocytes to lead

(1-100 µM) resulted in the enhancement of both B and T-cell activities (Lawrence and

McCabe Jr, 2002, McCabe and Lawrence, 1990). Interestingly, in this model, higher

levels of lead suppressed immune activity and increased bacterial susceptibility in vivo.

Thus, effects of lead on the immune system may differ depending on lead dosage and

exposure time, as well as the sub-population of responsive immune cells (Lawrence,

1981). Based on in vivo and in vitro studies, lead preferentially enhances Th2 cell

development and affects the development of B cells (Lawrence and McCabe Jr, 2002,

McCabe and Lawrence, 1990). Moreover, in vivo lead acetate treatment inhibits cell

adhesion properties and alters cell morphology in the splenic murine macrophages

(Sengupta and Bishayi, 2002). Gao and colleagues (2007) showed that lead can modulate

the development and function of the murine bone marrow derived dendritic cells. In

addition, lead-exposed dendritic cells skewed the immune response from Th1 to Th2

responses (Gao et al., 2007). It has been suggested that systemic immunosuppression,

which is characterized by a shift towards a T-helper cell type 2 (Th2) cytokine pattern, is

beneficial in the context of CNS lesions, as it results in reduced cell mediated and

humoral immune responses (Hendrix and Nitsch, 2007). Animal studies also showed that

lead may affect the immune-nervous system interaction through altering the

immunogenicity of two neuronal proteins, glial fibrilary acidic protein (GFAP) and

myelin basic proteins (MBP). Secretion of auto-antibodies against these proteins which

could enhance the progression of lead-induced neurotoxicity through auto-immunity

responses. Recent studies on human peripheral blood monocytes (PBMC) showed that

lead can activate IL-8 secretion, as well as induce secretion of pro-inflammatory

cytokines, such as IL-6 and TNF-α, though the mitogen-dependent activated pathway

(MAPK) (Gillis et al., 2012).

Page 34: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[13]

1.2. Microglia

Microglia are the resident macrophages of the brain and are highly responsive with key

functions in the host’s defense, as well as in neuroprotection and regeneration. Microglia

were first identified in the late 19th century. In 1899, the psychiatrist Franz Nissl came

across rod shaped cells in the CNS and described them as reactive glial elements

(Buchholz, 1899). Then, Santiago Ramón y Cajal in 1913 determined that glial cells were

the third major element of the CNS, in addition to neurons and astrocytes (Cajal, 1913).

However, Pio del Rio-Hortega, a Spanish neuroanatomist, was the first person to

determine that there were two types of glial cells, oligodendroglia and microglia, based

on morphological differences using silver- staining methods in 1932. He described

several fundamental characteristics of microglia that are still accepted and cited in

current microglia research (Del Rio-Hortega, 1932).

1.2.1. Origin

The origin of microglia has been debated for more than a century. Most researchers agree

that microglia have a myloid origin. Recently, Ginhoux et al (2010) showed that during

the embryonic and perinatal development of mice, two hematopoietic waves of microglia

are recruited to the CNS, where they differentiate. Ginhoux et al (2010) also reported that

myeloid cells expressing the hematopoietic marker CD45 and the adult

macrophage/microglia markers CD11b, F4/80, and CX3CR1 were detectable in the

developing brain starting from E9.5.It has also been reported that the primitive myeloid

progenitors have limited potential to give rise to adult blood leukocytes. Current evidence

suggests that in mammals, microglia are derived from primitive myeloid progenitors that

arise before embryonic day 8 in mice (age E8.0) (Ginhoux et al., 2010).

Throughout life, microglia are replenished by limitedproliferation of resident microglial

cells. Replenishment by bone marrow-derived progenitor cells is still underdebate.

Numerous reports have shown that bone-marrow-derived cells have the ability to

populate the CNS and differentiate into functional parenchymal microglia as well as

perivascular microglia (Soulet and Rivest, 2008, Simard and Rivest, 2004). Although

bone-marrow-derived cells can enter the brain parenchyma and throughout the CNS in

normal mice, studies using a panel of bone-marrow chimeric and transfer experiments

have shown that Ly-6Chigh

CCR2+ monocyte were preferentially recruited to the lesioned

adult brain and differentiated into microglia (Mildner et al., 2009). It has also been

reported that microglia and astrocytes chemokines could regulate monocyte recruitment

Page 35: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[14]

through the BBB in human immunodeficiency virus-1 encephalitis (Persidsky et al.,

1999).

1.2.2. Turnover

The turnover rate of microglia has been the subject of multiple studies since their

identification. Several studies have reported an amplified number of microglia during

pathological conditions. This may be due to the accelerated turnover in order to

compensate for the need of new cells. Investigators believe that microglia are a long-

lived population of macrophages, but how these cells are maintained during different

conditions is still unknown (Ransohoff et al., 2007). Two subsets of phagocytes have

been found in the adult brain: resident microglia in parenchymal microglia and

perivascular macrophages. Studies have shown that under healthy conditions, the

microglial replacement rate is low. However, during injury or inflammation, the turnover

rate of microglia is increased. Supplementation of resident populations of microglia with

bone-marrow derived cells has been seen in several CNS injury models. It has been

suggested that this phenomenon may also occur in the normal CNS, but at a very low rate

(Simard et al., 2006, Ajami et al., 2007).

1.2.3. Distribution

Microglia make up approximately 5-20% of the cells in the CNS (Lawson et al., 1990,

Benveniste, 1997, Tambuyzer et al., 2009). Based on animal and human studies, there

seems to be a differential distribution of microglia in separate regions of the brain. The

percentage of microglia in the healthy human brain show significant regional differences

ranging from 0.5% to 16.6% in the brain parenchyma with significantly

more microglia in white than in gray matter (Mittelbronn et al., 2001). In adult mice,

microglia are predominant in grey matter, with the highest concentration found in the

hippocampus, olfactory telencephalon, basal ganglia and substantia nigra (Lawson et al.,

1990).

1.2.4. Morphology

In the healthy brain, resting microglia manifest a typical ramified morphology, consisting

of a small cell body and long slender processes with secondary branching. In response to

stimuli, such as injury, toxins or inflammation, the processes are retracted and the cells

become more amoeboid-like, and transform into an activated phenotype (Figure 1.7)

(Xiang et al., 2006) . Del Rio-Hortega has also described the amoeboid cells showing

phagocytosis and migration similar to macrophages (Del Rio-Hortega, 1932). Resting

Page 36: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[15]

microglia with ramified morphology act as the gatekeepers of the brain and they restrict

changes or insults through constant surveying of the brain’s microenvironment. Recent

studies, using in vivo two-photon imaging in the neocortex, have reported that resting

microglia exhibit very dynamic processes that project and retract extensively to survey

and control the local environment (Kofler and Wiley, 2011, Nimmerjahn et al., 2005).

There is evidence showing that microglia activation occurs in response to recognition of

foreign pathogens, serum components, intercellular structures, stress, protein aggregates,

or receptor signaling pathways (Prinz et al., 1999). In the context of receptor signaling,

neuron-microglia interaction are involved in modulating the microglia phenotype and in

maintaining the ramified morphology. Disturbance in expression of some neuronal

ligands, such as fractalkine, CD200 and CD47, and reduced interaction with their

microglia receptors are associated with activated morphology (Lyons et al., 2009, Walker

et al., 2009).

Figure 1.7: Microglia is the immune effectors cells in the brain.

In the CNS, quick responses to stimuli results in activation of resting microglia, which is followed

by changes in function and morphology, from, ramified to amoeboid. Activated microglia increases

the expression of MHC molecules, enhances secretion of pro- and anti-inflammatory cytokines and

chemokines, and become potent APC as well as phagocytes. In the brain, activated microglia play

either a neuroprotective or neurotoxic role depending on the immune stimulation and secretions.

Source: Block et al., 2007, Trudler et al., 2010.

Page 37: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[16]

1.2.5. Microglia function: Immune effectors of the CNS

A quick response to any insult that disturbs the CNS homeostasis leads to microglia

activation, which results in changes in microglia morphology (from ramified to

amoeboid) and function. Based on recent published evidence, several surface molecules

expressed by microglia are involved in recognition of antigens, non-self or stressed-self

structures, phagocytosis, and migration, as well as pro- and anti-inflammatory responses,

which then stimulate the immune system (Figure 1.8). Like macrophages, microglia have

a wide range of receptors that are present on their surface which are responsible for the

detection of damage, associated with molecular pattern molecules (DAMP), which are

released by injured neurons or degenerating cells. Heat shock proteins, stress, oxidized

lipids, DNA, ATP and high mobility group box-1 (HMGB1) proteins are the main known

DAMPs. The receptors that detect DAMPs are scavenger receptors, Toll-like receptors

(TLRs), and receptors for advanced glycation end products (RAGE). TLRs play an

important role in recognition of the pathogen-associated pattern molecules (PAMP). TLR

signaling plays an important role not only in the innate immune system response, but also

in inflammation and demyelinating disorders. Ten TLRs have been identified in humans,

which can be expressed by microglia in the brain (Olson and Miller, 2004). Each member

of the TLR family recognizes different pathogens. TLR4 is the most studied member of

the Toll-like family, which is responsible for the recognition of LPS and the outer

membrane component of gram negative bacteria. The LPS-TLR4 interaction induces

activation of several intracellular signaling pathways, which initiate immune responses

through the secretion of pro-inflammatory cytokines (Luna et al., 2012, Kawai and Akira,

2011). Microglia express many other surface receptors, including phosphatidyl serine

(PS) receptor, LPS receptor CD14, the scavenger receptor CD36 and the purine receptor

P2Y6, which interact directly with the target to initiate phagocytosis (Hirt and Leist,

2003, Jones et al., 2012, Liu et al., 2005, Koizumi et al., 2007).

Microglia are in close contact with their cellular neighbours: neurons, astrocytes and

oligodendrocytes (HANSSON and RÖNNBÄCK, 2003). These connections have

significant effects on the microglia phenotype and function through receptor-ligand

interaction. There are a growing number of known molecules expressed or secreted by

neurons that bind to receptors on microglia. Fractalkine is one of these molecules which

is secreted by neurons and can modulate microglial activation through its receptor,

CX3CR1.

Page 38: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[17]

Figure 1.8: Chemo taxis or phagocytosis-involved receptors in microglia and correlation of the inflammatory or anti-inflammatory response.

Various receptors expressed by microglia are correlated with migration, phagocytosis, pro- and

anti-inflammatory responses. Source: Noda and Suzumura, 2012.

Microglia as the resident immune cells of the CNS have ability to act as an antigen-

presenting cell (APC) to stimulate the immune system through processing and presenting

antigens to T-cells. Previous studies provided evidence showing that constitutive MHC

class II molecules (HLA-DR) are expressed by resting microglia (Gehrmann et al., 1993).

Several studies have shown that microglia activation in response to neuronal damage,

infection, or inflammation is usually followed by increased expression of MHC class II,

as well as adhesion/co stimulatory molecules ICAM-1, CD40, CD80, and CD86. In

response to interferon-gamma (IFN-γ) and/or LPS stimulation in vitro, microglia are able

to prime allo-reactive T-cell responses and stimulate T-cell lines to proliferate and

secrete cytokines (Becher and Antel, 1996, Aloisi et al., 1998, Williams et al., 1993).

Microglia can be regarded as more efficient APC of the CNS than astrocytes for the

stimulation of both Th1 and Th2 cells (Aloisi et al., 1998).

The phagocytic ability of microglia plays a key role during development, as well as

during injuries and inflammation. Depending on the engulfed particles and the receptors

involved, phagocytosis can occur together with secretion of pro- or anti-inflammatory

mediators. During bacterial invasion, the activation of receptors, such as TLRs, induces

the secretion of pro-inflammatory cytokines, as well as active phagocytosis. However,

during debris removal, microglia secretion moves towards the anti-inflammatory

molecules (Hanisch and Kettenmann, 2007, Hanisch et al., 2001, Chan et al., 2001,

Magnus et al., 2001).

Page 39: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[18]

1.2.6. Role of microglia: neuroprotection versus neurodegeneration

The role of microglia in neuropathology is dichotomised into neurodegeneration and

neuroprotection (Ransohoff et al., 2007). There is increasing evidence that microglia

activation can either enhance or decrease neurogenesis depending on the stimuli and

situation (Gemma et al., 2010, Morrens et al., 2012). Microglia are important in synapse

remodelling and plasticity during development. The central nervous system is remarkable

for the large amount of developmental apoptosis that occurs during embryogenesis.

Microglia have been shown to play an important role in inducing developmental neuronal

apoptosis. Recent studies have demonstrated the important contribution of microglia to

synaptic remodelling. Microglia also play a role in synaptic striping in the pathogenesis

of neurodegenerative disorders (Perry and O'Connor, 2010).

A considerable amount of evidence supports the occurrence of adult neurogenesis in the

mature brain. One piece of supporting evidence for this view is the existence of neural

stem cells (NSCs) in the mammalian central nervous system. Microglia can promote

adult neurogenesis through the induction of NSCs proliferation, differentiation and

migration (Ekdahl et al., 2009).

It has been shown that disturbances in neurogenesis are also involved with induced

neuronal apoptosis as a result of injuries or inflammation during neurodegenerative

disorders. Notably, recent collective data indicate that neurogenesis is controlled by

endogenous factors such as cytokines, chemokines, neurotransmitters and reactive

oxygen species (ROS) that are released by activated microglia, astrocytes and injured

neurons (Whitney et al., 2009). It has been demonstrated in animal and human models

that microglia activation plays a critical role in the progression of various

neurodegenerative disorders, such as Parkinson's disease (PD), Alzheimer's disease (AD)

and Huntington's disease (HD), as well as neuro-inflammatory disorders including AIDS,

encephalitis and multiple sclerosis (Sugama et al., 2009, Nelson et al., 2002, McGeer and

McGeer, 2002).

1.2.7. Cytokines and neuroinflammation

Increased secretion of pro-inflammatory cytokines is one of the typical characteristics of

activated microglia, which induces an inflammatory reaction in the brain. Indeed, it is

well established that inflammation and activated microglia could play a positive role in

neurogenesis during brain injuries, such as brain ischemia. Interestingly, several

cytokines can induce both pro- and anti-inflammatory responses. For example, IL-6 has

Page 40: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[19]

the ability to induce NSC differentiation. On the other hand, an elevated level of IL-6 has

a dramatic effect on attenuating neurogenesis (Tang et al., 2009, Vallieres et al., 2002).

Opposing dual effects on neurogenesis has also been reported for IFN- (Song et al.,

2005). TNF- is a potent pro-inflammatory cytokine, mainly secreted by activated

microglia, and to a lesser extent, by astrocytes or neurons. Increased levels of TNF-

have been reported for many neurodegenerative disorders. Both, stimulatory and

inhibitory effects of TNF- have been shown in proliferation, differentiation and

migration of neural precursor cells (Ben-Hur et al., 2003).

1.3. Lead and microglia

Despite the importance of microglia in the physiology and pathology of the brain, there is

not much known about the exact role of microglia in lead-induced neurotoxicity. Recent

studies have raised the idea that microglia and astrocyte activation may be involved in

lead- induced neurotoxicity (Liu et al., 2012, Struzynska et al., 2007). However, most

research data have been obtained from in vitro and in vivo animal models, which may not

be the ideal models, especially in the context of the cellular immunity. To my knowledge,

there are no published data about the contribution of human microglia in lead-induced

toxicity. It is noteworthy that most of the studies on human microglia have been done

using fetal brain samples, post-mortem material and two non-commercialized cell lines.

In addition, the dosage of lead used in several studies was much higher than the level, the

human body may be exposed to in vivo. Recent studies found that lead-exposure induced

microglia activation was followed by the release of TNF-, IL-1β and enhanced

expression of inducible nitiric oxide syntase (iNOS) in vitro (Liu et al., 2012). Lead also

enhanced neuronal injuries in co-cultures of microglia with hippocampal neurons (Liu et

al., 2012). Low dose lead exposure caused considerable reduction in long-term

potentiation (LTP). It has been suggested that lead could impair spatial learning and

memory through the impairment of LTP. Indeed, microglia activation also could affect

LTP. Thus, in this study microglia activation has been suggested as a possible

mechanism involved in lead -induced LTP impairment (Liu et al., 2012).

In vitro studies have revealed that cytotoxicity and the activation of astrocytes occur

following lead exposure. An increase in densities of lysosomes, glial fibrillary acidic

protein (GFAP) and S 100 protein, along with the enlargement of rough endoplasmic

reticulum (RER), has also been reported. Enhanced detachments of astrocytes in presence

of lead in culture has been reported as well (Tiffany-Castiglioni and Qian, 2001).

Page 41: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[20]

1.4. Role of chemokines and chemokine receptors in CNS development

Chemokines, which are chemoattractant cytokines, and their G protein-coupled receptors

are essential for the selective recruitment of leukocytes to inflammatory sites. Besides

chemotaxis, chemokines are also involved in neuronal development, synaptic

transmission, modulation of cell adhesion, phagocytosis and cytokine secretion (Cartier

et al., 2005, Biber et al., 2002). Due to their wide range of biological functions,

chemokine receptors can be grouped as either constitutive or inflammatory (Figure 1.9),

depending on their involvement in development or inflammatory responses (Bennett et

al., 2011). The role of chemokines as neuromodulators is important in the neuron-

microglia interactions, as well as in the coordination of communication between the

nervous system and the immune system.

Figure 1.9: Constitutive vs. inducible chemokine

Chemokine receptors and their ligands can be broadly categorized into two classes depending on

whether they are constitutively produced or are inducible. Constitutive chemokine-receptors play a

basal role in leukocyte trafficking during development. However, inducible chemokine-receptors

are mainly involved in inflammatory responses. Source: Proudfoot, 2002.

There is increasing evidence that cytokines and chemokines in the CNS are involved in

neuronal development by regulating proliferation, survival, migration and differentiation

of the neuronal stem cells under physiological and inflammatory condition (Table 1.1).

Studies have shown that chemokines released from astrocytes, CCL3 (MIP-1) and CCL5

(RANTES) enhance the rat embryonic neuronal CCR5–mediated differentiation (Park et

al., 2009). Embryonic neuronal differentiation is promoted by the treatment with

Page 42: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[21]

astrocyte-microglia conditioned medium (Park et al., 2009). Chemokines have been

shown to be important in modulating the adult neurogenesis through expression of

chemokine receptors (CCR2, CXCR4) by neural stem cells. During inflammation,

expression of reciprocal ligands, CCL2 and CXCL12 is up-regulated by activated

microglia, as well as injured neurons, and can attract progenitor cells to sites of injuries

for repair (Belmadani et al., 2006, Liu et al., 2007, Miller et al., 2008).

Similar to the haematopoietic factor Granulocyte-colony stimulating factor (G-CSF),

granulocyte -macrophage colony stimulating factor (GM-CSF) have been reported to

have a stimulatory effect on rodent neuronal stem cells during differentiation (Kruger et

al., 2007). The expression of CCR9 and its ligand CCL25 (TECK) has been reported in

PC12 rat cells, a useful model for neuronal differentiation. It has also been shown that

CCL25/CCR9 interaction can promote PC12 survival (Cao et al., 2012).

Recent in vivo and in vitro studies have reported that some chemokines and chemokine

receptors, such as CCL2/CCR2, FRACTALKINE/CX3CR1, CXCL8/CXCR1/CXCR2

and CXCL12/CXCR4, are constitutively expressed in the CNS with roles in the

modulation of neuro-protection and neurogenesis, as well as neuro-inflammation

(Semple et al., 2010). Several studies have shown that along with their involvement in

inflammatory responses, cytokines play an active role in the CNS developmental process

especially in the proliferation, migration and differentiation of neuronal stem cells

(NSCs). Interleukin-4 (IL-4), interleukin-6 (IL-6), leukaemia inhibitory factor (LIF),

transforming growth factor beta (TGF-β), MCP-1/CCL2, GM-CSF and G-CSF have been

shown to be involved mainly in NSCs differentiation. Tumor necrosis factor alpha (TNF-

), LIF and stromal cell -derived factor (SDF-1)/ CXCL12 are considered to be the key

regulators of proliferation. Meanwhile, MCP-1 and SDF-1 play an active role in cell

migration (Taga and Fukuda, 2005, Gonzalez-Perez et al., 2010, Cameron et al., 1998,

Buisson et al., 2003). Like microglia, astrocytes also promote the NSCs differentiation.

Studies showed that microglial IL-6 can enhance the differentiation of astrocytes. On the

other hand, microglia can modulate the NSC differentiation either directly or indirectly,

through the secretion of IL-6 (Nakanishi et al., 2007).

Glia cells, microglia and astrocytes are the main sources of IL-6 in the CNS. However,

recent studies have shown the neuronal expression of IL-6 has a role in physiology and

inflammation. It has been reported that secretion of chemokines, such as IL-8, in

microglia can be regulated by pro- or anti-inflammatory cytokines. Human fetal

microglia showed enhanced secretion of IL-8 in the presence of LPS or pro-inflammatory

cytokines such as IL-1 β. However; IL-8 expression was attenuated when microglia were

Page 43: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[22]

treated with anti-inflammatory cytokines such as Il-4, Il-10 and TGF- β (Ehrlich et al.,

1998). IL-8/CXCL8 is a chemokine which is in humans can bind to both CXCR1 and

CXCR2 receptors. Mouse and rat can not express IL-8 and its receptos due to the

deletion of the genes encoding IL-8 and its receptor CXCR1 in a common ancestor of

mouse and rat, and therefore IL-8 is regarded as unique chemokine in human studies.

Human. IL-8 enhances progenitor cell division, neurogenesis, ventral midbrain

development and increases the number of dopaminergic neurons in the rat brain (Edman

et al., 2008). CXCR1 exclusively binds IL-8 and CXCL6 with high affinity, whereas

CXCR2 can be activated by additional ligands such as CXCL1,CXCL3 and CXLCL5-7.

Activation of the two IL-8 receptors triggers the formation of distinct second messengers,

which activate several signaling pathways through protein kinases and phospholipases.

The main signal transduction pathways induce the transient mobilization of free Ca+2

from internal stores.

For instance, distinct expression of CCL2/CCR2 has been identified during brain

development (Rezaie et al., 2002). CCL2 is mainly secreted by microglia and astrocytes.

Neuronal CCL2 secretion has also been shown to occur following trauma. Enhanced

secretion of CCL2 occurs in response to several stimuli during neuro-inflammation. It

has been shown that CCL2 could induce neuronal precursor migration to the site of

inflammation, and may be involved in neurogenesis (Semple et al., 2010).

Increased neuronal death has been reported in CCL2/CCR2 knock-out animal models of

methyl mercury neurotoxicity which implies a possible protective role of CCL2 in brain

deficits caused by toxicity (Godefroy et al., 2012). In addition to proliferation,

CCL2/CCR2 is involved in differentiation, as well as cell migration. Recent studies have

shown that treatment of embryonic rat cells with CCL2 and CCL7 enhances cell

differentiation towards dopaminergic neurons (Edman et al., 2008). CCL2 may also act

as the modulator of the blood-brain barrier permeability through interaction with

endothelial cells expressing CCR2 that may facilitate leukocyte trafficking to the brain.

1.5. Role of cytokines, chemokines and chemokine receptors in

neuroinflammation

Neuro-inflammation, neuronal damage and cell death in the CNS is usually accompanied

by the activation of microglia and astrocytes following the secretion of pro-inflammatory

cytokines and chemokines. There is published evidence showing that neuronal damage in

the CNS, after long term exposure to low-level lead can be attributed to the over

expression of TNF-, which is primarily released by microglia and astrocytes (Viviani et

Page 44: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[23]

al., 1998). Recent studies on human glioma cell lines have shown that lead and

liposaccharid, both share common signaling pathways that result in secretion of TNF-

(Cheng et al., 2004). Based on recent published evidence, chemokines and chemokine

receptors are expressed in CNS at low levels under healthy conditions and mediate the

innate immune system. In response to neuro-inflammatory stimuli, their expression is

rapidly induced and this has been reported in various neurological disorders (e.g. trauma,

stroke and Alzheimer's disease), in tumour induction and in neuroimmune-related

diseases, such as multiple sclerosis or acquired immunodeficiency syndrome (AIDS)

(Mennicken et al., 1999). Therefore, they play a key role in the pathogenesis of several

neurodegenerative or neuro-inflammatory diseases. Increased secretion of pro-

inflammatory cytokines and chemokines, such as IL-6, is one of the marked

characteristics of the activated microglia which can result in the inflammatory response,

activating more microglia, as well as astrocytes. Reactive gliosis, demonstrated by an

increase in the number and size of glia cells, has been reported as one of the symptoms of

neurodegenerative diseases ((Rubio-Perez and Morillas-Ruiz, 2012, Sheng et al., 2001,

Mrak and Griffin, 2005, Lee et al., 2013). Over-expression of cytokines may also cause

neuronal cell death through changes to neurotransmitter receptors and neuronal activation

(Kaindl et al., 2012).

Page 45: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[24]

Table 1.1: Chemokine and chemokine receptors in CNS

Chemokine receptor Chemokine

Common name Specific name

CCR1

MIP-1

RANTES

MCP-3

CCL3

CCL5

CCL7

CCR2

MCP-1

MCP-2

MCP-4

MCP-3

CCL2

CCL8

CCL13

CCL7

CCR3

Eotaxin

MEC

RANTES

MCP-4

CCL CCL11

CCL28

CCL5

CCL13

CCR4 TARC

MDC CCL17

CCL22

CCR5

MIP-1β

MIP-1

RANTES

CCL4

CCL3

CCL5

CCR6 MIP-3 CCL20

CCR7 SLC CCL21

CCR9 MEC CCL28

CXCR1 IL-8

GCP-2

CXCL8

CXCL6

CXCR2

NAP-2

ENA78

GRO-

GRO-β

GRO-

IL-8

CXCL7

CXCL5

CXCL1

CXCL2

CXCL3

CXCL8

CXCR3

IP-10

MIG

I-TAC

CXCL10

CXCL9

CXCL11

CXCR4 SDF-1 CXCL12

CXCR5 BCA-1 CXCL13

CX3CR1 Fractalkine CX3CL1

Page 46: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[25]

Chapter 2

Research aims and outline

Page 47: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[26]

2. Research aims and outlines

2.1. Aims

The literature review highlighted several potential mechanisms of lead poisoning in

humans. It focused on the current understanding and knowledge about the toxic effects of

lead, mainly on the developing central nervous system via changes in proliferation,

migration and differentiation of precursor cells, as well as possible involvement of

immune cells and inflammatory responses in lead-induced toxicity.

The literature review also underlined the key roles of microglia as the resident immune

cells of the brain during health and disease. In the context of microglia, more attention

was on the contribution of cytokines and chemokines as the immune modulator of the

CNS in both, neurogenesis and neuroinflammatory responses. The chemokine-

chemokine receptor axis has been shown to play an active role in the microglia- neuron

interaction.

Most of the data about the biology and function of microglia are gathered from in vitro

studies using the primary culture of embryonic or neonatal murine brain or established

cell lines. In humans, primary culture of microglia is not easily feasible due to ethical

reason and lack of sources. On the other hand, fetal microglia may behave differently in

some aspects from adult microglia. In addition, few established human cell lines have

been generated. Therefore, lack of a convenient and appropriate model for in vitro human

microglia studies is one of the current challenges for research into the human microglia.

It is well documented that lead exposure, even at low concentrations, can be associated

with deficits in neurogenesis and spatial learning abilities in adulthood through

inflammatory reactions.

Activation of microglia as the immune effectors of the brain has been reported in several

neurodegenerative diseases, including heavy metal toxicity. There is evidence showing

that a number of heavy metals can induce microglia activation followed by secretion of a

variety of immune modulators. In most studies, metal-induced microglia activation

enhanced the neuorinflammatory/neurodegenerative responses. Despite the reported

neurotoxic effects of lead, especially in children, little is known about the role of

microglia as the immune resident of the CNS in lead-induced

neurotoxicity/neuroinflammation. Therefore, in this thesis, I investigate the role of

Page 48: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[27]

microglia in lead-induced neuroinflammation using in vitro human models.The overall

aims of this thesis were to test the following hypotheses (Table 2.1).

Table 2.1: Testing hypotheses of microglia involvement in lead-induced neurotoxicity

In this thesis, several experimental studies were developed and designed to obtain

information to answer the hypotheses above. Main aims addressed in this thesis are

shown below (Table 2.2).

Table 2.2: The main aims of this thesis

Aim 1

Determine the effect of lead exposure on the in vitro human

neural precursor cells, with specific focus on immune-related

factors.

Aim 2

Establish and characterize the novel in vitro human microglia

model derived from human blood monocytes.

Aim 3 Study the effect of lead on the in vitro human microglia model

in the context of morphology, phenotype and function.

Aim 4

Investigate the effect of lead exposure on patterns of chemokine

and chemokine receptor expression in microglia in vitro

models.

Hypothesis 1

Lead has cytotoxic effects on survival and morphology of in

vitro microglia models.

Hypothesis 2

Lead activates microglia towards enhanced inflammatory

responses.

Hypothesis 3

Lead does not have a substantial effect on human neuronal

precursor cell in vitro.

Page 49: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[28]

2.2. Research outline and methodology

Several qualitative and quantitative tools and techniques, as well as support and

experience of experts were employed. Table 2.3 summarizes the main techniques applied

in this thesis. This study will provide new insights about the role of microglia in lead-

induced inflammation using the feasible and convenient in vitro human microglia model.

Knowledge on the effects of lead on the cytokine/chemokine receptor interactions in

microglia will provide new approaches to understand the role of lead as one of the known

environmental pollutants in modulating the immune responses in brain.

Table 2.3: Main methods applied in this study and the corresponding chapters.

Chapters Methods

Chapters 4, 5

and 6

Separation and isolation of the blood mononuclear cells

(PBMC) using human blood buffy coats

Chapters 3, 4,

5, 6 and 7

In vitro culture of the human blood monocytes, lymphocytes,

dendritic cells, microglia, human microglia and neuronal cell

lines

Chapters 3, 5

and 7

Viability test (MTS assay, SYTO-green and Propidium iodide

staining)

Chapters 3, 5

and 7 Proliferation test (BrdU Assay)

Chapters 3, 4,

5 and 7

Morphological studies ( phase-contrast and confocal

microscopy)

Chapters 3, 4,

5,6 and 7

Phenotype characterization using the flow cytometry and

immune-histochemistry staining

Chapter 4 Functional assay (phagocytosis, mixed leukocyte reaction -

MLR)

Chapters 5

and 7

Intracellular lead uptake using Leadmium green staining

Chapters 3, 5

and 7

Cytokine measurements

In order to investigate the proposed aims of this thesis, five separate studies were

undertaken. Each of these make-up their own chapter and each consists of their own

background information, methods, results, discussion, conclusion and references (Figure

2.1).

Page 50: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[29]

Figure 2.1: Research outline

To investigate the proposed aims, five separate studies were undertaken. The results of these

studies are presented in five papers found in this thesis.

Page 51: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[30]

Chapter 3

Effect of lead on human cortical neuronal

precursor cells

Page 52: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[31]

3. Effect of lead on human cortical neuronal precursor cells

3.1. About the paper

Lead poisoning remains a significant global health problem due to causing cognitive

deficits and neurological problem in children even at lower concentrations. In human,

brain formation begins in the third gestational week with the differentiation of neuronal

precursor cells (NPCs) and continues to develop in the first years of life. Therefore, brain

at the early time of development is sensitive to exposure of toxicants such as lead, which

can affect further developmental, and maturation processes. Despite the fact that lead

exposure can occur both during prenatal or postnatal exposure, it is still little known of

effect of lead exposure on neural precursor cells morphology and function, or to what

extent development and maturation of neuronal precursor cells are affected by lead.

There is evidence showing that lead exposure can differentially affect the proliferation

and differentiation of neuronal precursor cells originating from different region in brain.

Recent animal studies showed that, despite the region where neural precursor cells

originate from, the age of the animal play a key role in the degree of sensitivity to lead.

For example, hippocampal neuronal stem cells from newborn rats were more vulnerable

to the toxicity of lead than those from adult rats. Lead also could alter the differentiation

of neural stem cells towards oligodendrocytes or astrocytes. In summary, these data

implicate the importance of lead exposure prevention during brain development. One

suitable model to study the neurotoxic effects of metals such as lead, on neuronal cell

morphology and function is using a human neuronal precursor cell line. In this study, we

used the human cortical neuronal cell line. Undifferentiated HCN2 cells have been

reported to express several neuronal markers including neurofilament protein, neuron

specific enolase (NSE), β-tubulin III, vimentin, somatostatin (SST),etc. However, cells

have been reported to be negative for glial markers, such as glial fibrillary acidic protein

(GFAP) and myelin basis protein (MBP), which make them a suitable model for

investigating human neuronal precursor cell biology.

Neural precursor cells, due to their unique capacity for self renewal and multipotency,

can give rise to neurons, astrocytes and oligodendrocytes during development. Over the

past decades, it became evident that neurogenesis occurs in distinct areas of the adult

brain, where neural stem cells, progenitors and precursors reside. Alteration in

proliferation and differentiation of neuronal precursor cells (NPCs) has been reported in

impaired adult brain cognitive function, as well as in the pathology of neurodegenerative

Page 53: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[32]

diseases. In addition, it became apparent that the immune system plays a key role in

modulating the CNS function. Recent evidence shows that microglia have the capacity to

influence the differentiation of both, adult and embryonic neural precursor cells toward a

neuronal phenotype emphasizing the importance of the microglia- neuron interaction.

In order to answer the main question of this thesis, which is the role of microglia in lead

induced neurotoxicity, we investigated toxic effects of lead on the human neuronal cell

line HCN2 over 24 hours in vitro. The results of this study showed that lead at the

concentration of 10 µM had no detrimental effects on morphology or viability of these

cells. Reduction in the number of cells was observed in the presence of 100 µM lead

acetate. However, lead significantly altered the expression of a number of chemokine

receptors and cytokines. Given that the key role of the immune system in the

differentiation and migration of NPCs, these results indicate that immunomodulatory

effects of lead may have an impact on HCN2 maturation and function as well as their

interaction with other cells in the CNS.

Declaration of the work:

All the experiments were designed and conducted by SE involving cell culture,

proliferation assay, viability assay, morphology study, surface marker expression

(FACS), cytokine measurement and statistical analysis under supervision of Prof. Luis

Filgueira.

3.2. Abstracts

Lead is a developemental toxicant with no known physiological function. Lead toxicity

has been recognized for at least thousands of years with diverse harmful effects on

human body. It has been found that early childhood exposure to excessive amounts of

lead may produce lasting adverse effects upon brain function. Although, several studies

reported the negative effects of lead on adult neuronal cells, not many studies have been

investigated the effects of lead on human neuronal precursor cells.

In this study, the effects of lead on human neural precursor cells in vitro at

concentrations, which are expected to be found during lead intoxication in vivo were

investigated. For that purpose, the human cortical neuronal cell line HCN2 was used as a

model. First, we studied the expression of neuronal-restricted precursor marker such as

nestin, vimentin as well as β-tubulin III and NGF-R as developmental-dependent

maturation marker. After 10 days culture in standard medium, HCN2 were exposed to 1,

10 and 100 µM of lead acetate for 24 hours. Then, changes in morphology, viability,

Page 54: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[33]

chemokine receptor expression and cytokine secretion were investigated. Our result

showed that lead at lower concentration (1 and 10 µM) had no effect on morphology and

viability of HCN2 compared to negative and positive controls. HCN2 under non-

stimulatory control conditions expressed most of the chemokine receptors. Alteration in

expression pattern of chemokine receptors has been seen in response to lead exposure.

These changes included significant down regulation in pro-inflammatory chemokine

receptors CCR1, CCR2, CCR3 and CCR5. However, expression of CCR4, CXCR1 and

CXCR3 significantly increased in lead-treated HCN2. Interestingly, we have found

contrary effects of lead exposure on the expression pattern of neuronal fractalkine

(CX3CL1) and its receptor (CX3CR1). HCN2 at basal condition expressed fractalkine

and CX3CR1 at low and medium levels. After lead exposure, expression of CX3CL1

was enhanced and of CX3CR1 decreased. Beside the chemokine receptors, β-tubulin III,

neuron-specific marker showed significant reduction in lead-exposed HCN2, including a

reduction in differentiation potential of the cells. We have also investigated the effect of

lead on secretion of cytokines by HCN2. Of all cytokines tested, only IL-8 and IL-6 were

secreted by HCN2 at basal culture conditions. However, lead significantly reduced

secretion of IL-6, but had no effects on IL-8. In summary, lead (10 µM, 24 hrs) had no

cytotoxic effects on human neural precursor cells. However, lead changed functional

properties of the cells, indicating that lead may interfere with neuronal turnover,

recruitment and differentiation.

Keywords: human neuronal cell line, HCN2, lead, neuronal precursor, chemokine

receptor

3.3. Introduction

Lead is a xenobiotic metal with no apparent reported essential function in cell growth,

proliferation or differentiation. Lead exposure has devastating effects on the developing

nervous system, producing morphological, cognitive, and behavioural deficits.The

neurotoxicity of lead, even at blood concentrations as low as 10 µg/ dl (~ 0. 5 µM), has

been repported in children (Finkelstein et al., 1998). Despite the well-documented

neurotoxic effects of lead, related mechanism are not fully understood. Lead affects

numerous biological activities, including mechanisms at cellular, intracellular and

molecular level (Finkelstein et al., 1998, Zurich et al., 2002, Lidsky and Schneider,

2003). Lead crosses the blood-brain barrier and causes behavioral alterations, and may

even disrupt the main structural components of the BBB (Bressler et al., 1999b). The

prefrontal cerebral cortex, hippocampus and cerebellum have been shown to be the

preferred target region in the brain for lead-induced damages (Finkelstein et al., 1998). In

Page 55: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[34]

vitro and in vivo animal and human studies have shown neurotoxic effects of lead,

especially during development, including impaired synaptic plasticity, inhibition of

glutamate receptor activation (Guilarte et al., 1994), apoptosis (Oberto et al., 1996),

learning and cognitive deficits, memory impairment (Xu et al., 2009), exicitotoxicity,

altered neurotransmitter storage and release, mitochondrial dysfunction, loss of neuronal

myelin sheaths, changes in cellular energy metabolism and disturbance of neuronal

growth (Toscano and Guilarte, 2005). There is evidence showing reduced expression of

neuronal cell adhesion molecules (e.g. NCAM), delayed neurite outgrowth and reduced

survival rate of cultured hippocampal neurons after exposure to low level of lead (Hu et

al., 2011). Prospective studies in animal studies have also correlated the elevated blood

lead level with memory deficits associated with changes in neuronal adhesion molecules

in the hippocampus (Murphy and Regan, 1999). Neural cell adhesion molecule (NCAM)

plays an important role during neural development and in the adult brain (Boutin et al.,

2009). NCAM are also shown to be involved in synaptic plasticity and long term

memory formation in adults. Attenuation in expression of NCAM along with memory

deficits and changes in synaptic plasticity has been shown in animal models (Welzl and

Stork, 2003, Baydas et al., 2003). Previous studies also reported the morphological

changes in hippocampus, as well as long lasting decrease in density of cholinergic

neurons in rats exposed to lead at concentration less than 200 µM prenatally (Slomianka

et al., 1989).

In the context of sensitivity of neurons and glia cells to lead, different experimental

models have been used resulting in various ideas about lead toxicity. Holtzman et al.

(1987) showed that that lead induced neuronal necrosis in rat neuronal culture, while no

effects on astrocyte organelle morphology was seen. They reported that neurons seemed

to be more sensitive to lead toxicity than astrocytes. They also suggested that astrocytes

are able to sequester lead as protective mechanisms (Holtzman et al., 1987). On the other

hand, Zurich and colleagues showed that enriched culture of neurons store more lead

than astrocytes after 25 days exposure to 1µM lead. Moreover, they showed that glia are

more affected than neurons in glia-neuron culture (Zurich et al., 1998).

Although in mammals the process of neurogenesis is almost terminated after

development, neurogenesis occurs throughout the life in two regions in the hippocampus

(Zhao et al., 2008). Several studies showed a positive correlation between adult

hippocampal neurogenesis and memory function. Changes in hippocampal neurogenesis

have been reported in several neurodegenerative disease pathology and age-dependent

memory-learning deficits (Voloboueva and Giffard, 2011).The hippocampal region is

Page 56: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[35]

known to accumulate lead, which has been experimentally proven in animals or in

environmentally exposed humans (Sandhir et al., 1994). Recent studies reported that

perinatal exposure to even low level of lead with corresponding low blood lead

concentrations (below 10 µg/dl), decreased the number of hippocampal neurons with no

changes in apoptotic or necrotic numbers indicating that there may be reduced

neurogenesis (Baranowska-Bosiacka et al., 2012).

Several studies have reported the link between pro-inflammatory cytokines as immune

modulators and neurogenesis. There is evidence showing that inflammation could play a

positive role in neurogenesis during brain ischemia (Tang et al., 2009). Elevated levels of

pro-inflammatory cytokines with enhanced neurogenesis has also been reported in

Alzheimer disease as a chronic inflammatory condition (Yoneyama et al., 2011). Among

pro-inflammatory cytokines, IL-1 and IL-6 showed inhibitory effects on neural stem cells

(NSCs) proliferation or neurogenesis. Although, the ability of the cytokine to induce

NSC differentiation to astrocytes has also been shown (Vallieres et al., 2002, Wang et al.,

2007). Based on in vivo and also in in vitro studies, higher levels of pro-inflammatory

cytokine, such as TNF-α, IL-1β, IL-6, IL-8, monocyte chemoattractant protein-1,

macrophage inflammatory protein-1 and CXCL10, are involved in HIV-1-induced

neuropathogenesis (Guha et al., 2012, Kelder et al., 1998).

Growing evidences suggest that IL-8/CXCL8, pro-inflammatory cytokine/chemokine,

and its receptors, CXCR1/2 play a key role in modulating the neuronal function. The

neuroprotective role of IL-8 in the AD pathogenesis has recently been reported. IL-8

inhibits Aβ-induced neuronal apoptosis and up-regulates neuronal BDNF production

(Ashutosh et al., 2011).Up-regulation of IL-8 has been shown in AD brain tissue.

Increased neuronal secretion of IL-8 has also been reported in the presence of both Aβ

and pro-inflammatory cytokines such as IL-1β or TNF-α (Ashutosh et al., 2011). In

contrast, overexpression of IL-8 could induce the pro-apoptotic proteins and cell death in

AD models (Thirumangalakudi et al., 2007). IL-6 is another pro-inflammatory cytokine

shown to be involved in modulating neurogenesis. Both IL-6 and the IL-6 receptor are

expressed in the postnatal hippocampus (Vallieres and Rivest, 1997), overexpression of

IL-6 in the hippocampus suppresses hippocampal neurogenesis (Vallieres et al., 2002)

and complete lack of IL-6 attenuates the proliferation and survival of neuronal precursor

cells (NPCs) (Bowen et al., 2011). TNF- and IFN- are the pro-inflammatory cytokine

involved in neuronal precursor cells survival and differentiation during neurogenesis or

neuronal remodelling. TNF- is expressed predominantly by activated microglia and at

lesser extent by astrocytes and neurons. Several studies showed the role of TNF- in

Page 57: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[36]

proliferation and differentiation of newly generated cells in the CNS. TNF- showed

inhibitory effects on NSCs proliferation but stimulating its migration in the new born rat

(Keohane et al., 2010, Turbic et al., 2011). IFN-γ-induced neural differentiation and

neurite outgrowth has also been shown in murine neural precursor cells (Song et al.,

2005).

Chemokine and chemokine receptors play an important role in CNS health and pathology

(Bacon and Harrison, 2000, Tran and Miller, 2003). Studies showed that various

chemokine and chemokine receptors are expressed at different levels by neurons during

development, which implicate their role in development, differentiation and even

inflammation through recruitment of leukocyte to the site of injuries (Tissir et al., 2004).

Furthermore, the chemokine/receptor axis play a key role in the neuron-microglia

interaction in the brain in both, neurogenesis and neurotoxic pathway. Chemokines have

been shown to be involved in regulating the migration of NSCs and NPCs (Belmadani et

al., 2006). Expression of a number of chemokine and chemokine receptors have been

reported in rodent and human neurons during physiology and pathology of the brain

(Robin et al., 2006, Losy and Zaremba, 2001, Abrous et al., 2005). A recent study

demonstrated that the CXCR1 and CXCR2 transcripts are widely expressed in

glutamatergic, GABAergic, and cholinergic neurons in the rat brain where they may

modulate transmitter release or ion channel activities (Puma et al., 2001). CXCR4, alike

CX3CR1, is one of the chemokine receptors expressed constitutively in the brain

(Hesselgesser and Horuk, 1999). It has been shown that neurons and astrocytes

expressing CXCR4 in the cerebral cortex, hippocampus, and substantia nigra frequently

coexpress CCR2 (Coughlan et al., 2000, Van Der Meer et al., 2001) which has been

shown to play critical roles in the migration of the NPC during neurogenesis or after

injury.

Among the chemokine/receptors, the fractalkine/CX3CR1 axis was the focus of several

in vitro and in vivo studies in recent decades. Fractalkine is widely expressed in the CNS

with constitutive expression by neurons (Meucci et al., 2000). First, it has been thought

that CX3CR1, the only fractalkine receptor, is expressed by microglia and astrocytes,

considered as one of the main neuron-glia communication in the CNS. Later, studies

showed that CX3CR1 is expressed by neurons, as well. Several studies have shown the

neuroprotective and/or neurotrophic role of fractalkine/CX3CR1 activation in the CNS.

For instance, there are evidences showing fractalkine protects hippocampal neurons from

HIV induced neurotoxicity (Meucci et al., 2000). It has been noted that on one hand,

neuronal fractalkine could supress the migration of neurons by inducing the molecules

Page 58: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[37]

active in cell attachments, such as integrins. On the other hand, it could enhance

microglia migration by means of expressing the neuronal CX3CR1 and facilitate

microglia-neuron interactions (Lauro et al., 2006). Fractalkine is considered for its

therapeutic potential in limiting inflammation, microglial over-activation, and neuronal

injury in neurodegeneration models (Suzuki et al., 2011). CX3CR1 is mostly activated by

soluble fractalkine, which modulates the cell activation and survival through pro-survival

signalling pathway, such as the Akt pathways (Meucci et al., 2000). The neuronal

CX3CR1 receptor could mediate the neurotrophic effects of fractalkine, suggesting that

fractalkine and its receptor are involved in a complex network of both, paracrine and

autocrine interactions between neurons and glia. During the CNS development,

regulation of the neuronal survival and programmed cell death (apoptosis) has a key role

in formation of the neuronal network and different parts of the CNS. It has been shown

that cortex is one of the region which is greatly vulnerable to inflammation or infection

occurred during prenatal or early post natal stages. Published studies showed that LPS-

induced inflammation alters synchronized neuronal patterns and increases apoptosis

(Nimmervoll et al., 2012). Furthermore, decrease in neuronal survival and induced

apoptosis followed by LPS-induced inflammation in CNS has also been a reported effect

of fractalkine and its receptor (Yeh et al., 2008).

Quick and constant up-regulation of neuronal fractalkine along with TNF- as a

biomarker of inflammation, followed by induced neuronal death, has been reported in

patients with epilepsy (Xu et al., 2012). Neuronal expression of TLR2 and TLR4 has

also been reported. Up-regulation, activation of TLRs and down-stream signalling via

JNK has been reported in response to ischemic condition in neurons.

In this study, we used the human cortical neural cell line HCN2 as an appropriate model

for in vitro study of human neuronal precursor cells. HCN2 was derived from cortical

tissue removed from a 5 years old girl undergoing hemispherectomy.These cells are a

really slow growing cell line (doubling time greater than 120 hours).The growth rate of

HCN2 can be stimulated in the presence of phorbol esters (Ronnett et al., 1994). Induced

differentiation in the presence of nerve growth factor (NGF) has also been reported

(Ronnett et al., 1994). In addition, neurotrorphic effects of NGF on neuronal precursor

cell survival and differentiation has been reported (Ronnett et al., 1994). According to

reported studies, HCN2 is positive for several neuronal markers, such as β-tubulin III,

vimentin, somastatin (SST), glutamate, gamma aminobutyric acid (GABA),

cholecytokinin -8 (CCK-8). The cells are negative for glial fibillary acidic proteins

(GFAP) and myelin basis protein (MBP) (Ronnett et al., 1994).To our knowledge, there

Page 59: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[38]

is no study showing the effcts of lead exposure on human cortical neuronal cell line or

HCN2. As the CNS is one of the main targets of lead toxicity and several studies have

reported behavioural and cognitive deficits, as well as neural morphological

abnormalities on developing brain, we were interested in studying the effects of brief

exposure of lead (10 µM, 24 hrs) on HCN2, as an appropriate representitve of human

cortical neuronal precursor in vitro.

3.4. Material and methods

3.4.1. Characterization of HCN2

The human cortical neuronal cell line HCN2 (CRL-10742) was purchased from

American Type Culture Collection (ATCC, Manessa, VA) . Cells were thawed and

cultured in DMEM Glutamax (invitrogen, Carlsbad, CA) supplemented with 10% fetal

bovine serum (FBS, Serana, Bunbury, WA, Australia) and 1% antibiotic/antimycotic

mixed solution (10,000 units/mL of penicillin, 10,000 µg/mL of streptomycin, and 25

µg/mL of Fungizone, Gibco, Grand Island, NY) at 37oC, 5% CO2 . HCN2 is a slow

growing cell line (doubling time greater than 120 hours), therefore, they were maintained

for about 3 weeks after thawing. They did not reach the 100 % confluence (maximum

60%). NGF (10 ng/ml, PeproTech, Rocky Hill, NJ) was also added to culture medium to

induce differentiation and mature morphology in HCN2 (Ronnett et al., 1994). Under

other experimental conditions, we cultured the HCN2 in the presence of fractalkine (10

ng/ml, PeproTech) and LPS (10 ng/ml, E. coli serotype 0111:B4, Sigma -Aldrich, St.

Louis, MO).

Lead acetate [Pb(CH3CO2)2·3H2O] (Sigma-Aldrich) was prepared at the concentration of

1, 10 and 100 µM in medium. After 10 days in culture, HCN2 cells were exposed to

different concentration of lead for 24 hours at 37oC. Then, cells were used for further

characterization and functional studies.

To study the expression of neuronal markers, immunostaining was performed with

HCN2 cultured on cover slips, fixed with 3% paraformaldehyde in PBS, containing 2 %

sucrose for 10-15 min. After permeabilization with 0.1% TritonX100 in PBS for 10 mins,

cells were incubated overnight at 4°C with one of these primary antibodies: a mouse

anti- β-tubulin III (1:100, Stemgent, Cambridge, MA), mouse-anti nestin (1:50, Sigma-

Aldrich) and rabbit-anti ionized calcium-binding adapter molecule 1 (Iba-1) (1:100,Wako

Pure Chemical Industries, Osaka, Japan). Next day, cells were incubated for 1 hour with

Alexa Fluor 488-conjugated donkey anti-mouse and anti-rabbit antibodies (1:200,

Page 60: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[39]

Invitrogen) and DAPI (1:400, Roche, Manheim, Germany). Cells were also stained with

a mouse-anti NGFR (1:50, BD bioscience, San Jose, CA) and rabbit-anti fractalkine

antibody (1:50, AbD Serotec, Oxford, UK) to detect surface expression of tested

markers. Thus, fixed cells were directly incubated with primary antibody without being

perrmeabelized. In the next step, cells were incubated with Alexa Fluor 488-conjugated

donkey anti-mouse as secondary antibody, anti-rabbit Ig G antibodies as isotype control

(1:200, Invitrogen) and DAPI (1:400, Roche) for 1 hour. Stained cover slips were

mounted on slide using the fluorescent mounting medium (Dako, Glostrup, Denmark)

and slides were ready for analysis under fluorescent microscopy. It needs to be pointed

out that . excitation and emission wavelengths of Alexa Fluor-488 and DAPI were

respectively 495, 519 and 350,470.Morphology of HCN2 was studied after 10 days in

culture in the presence/absence of lead using an inverted Leica DM IL phase contrast

microscope. Images were taken with Photometrix Cool SNAP Fx digital camera and

processed with NIS-elements F3.0 software.

3.4.2. Viability assay

Cell viability was analyzed by the MTS assay. When cell reached the optimal confluency

(60%), they were challenged with 1, 10 and 100 µM lead for 24 hours at 37 °C. Then, 20

µl of MTS Aques solution working solution (1:100 from stock solution) was added

directly to each well and cells were incubated for 4 hours at 37°C. In addition, in three

experimental groups, HCN2 were treated with NGF (10 ng/ml, PeproTech), CX3CL1 (10

ng/ml, PeproTech) and LPS (10 ng/ml, Sigma-Aldrich) for 24 hours at 37°C to compare

the effect of these agents with lead treatment at different concentration on cell viability.

Samples were measured with a microplate reader (Multiskan) at 490 nm. Data were

analyzed and results are shown as percentages of cell survival (viability). Non-lead

treated cells viability was considered as 100 in this experiment. Same experimental

conditions (plated cell number, volumes of reagents) were applied in all tests.

Measurements were done in tetraplicate and repeated with three cellular experimental

repeats (n=3).

3.4.3. Surface marker expression

We studied the effects of lead exposure (10 µM) on the expression patterns of specific

surface molecules in HCN2 using flow cytometry. Cells were divided into two groups,

non-treated and lead-treated, which were exposed to lead for 24 hours. After detaching

and washing twice with PBS, HCN2 were stained with direct labelled antibodies, or

using primary unlabelled and corresponding labelled secondary antibodies. A panel of

Page 61: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[40]

directly labelled mouse anti-human antibodies, i.e. CCR1-AF647, CCR2-AF647, CCR3-

PE, CCR4-PE-Cy7,CCR5-FITC, CCR9-fitc, CXCR1-PE, CXCR3-AF488, CXCR4-PE

and CD 271-PE (NGFR p75) (all from BD Bioscience), were added at the supplier’s

recommended concentration. Corresponding labelled isotype control antibodies were

included in all experiments. A rabbit anti-human fractalkine (CX3CL1) and rabbit anti-

CX3CR1 polyclonal antibody was used at the final concentration of 1ug/ml (AbD

Serotec), followed by a secondary donkey anti-rabbit-AF647 used at the final

concentration of 1:200 (Invitrogen). A mouse anti-human β-tubulin III (Stemgent) was

also used at final a concentration of 1:200. In order to detect β-tubulin III intracellularly,

the cells were fixed with paraformaldehyde (PFA) 1% for 10-15 min. Then, they were

incubated with PBS Tween 0.5% as detergent for 10 min. After washing, they were

incubated with anti-β-tubulinIII, diluted in PBS Tween 0.5% for 1 hours, followed by

secondary donkey anti-mouse AF488 at final concentration of 1:200 for 30 min. After

washing twice with PBS-Tween 0.5%, cells were ready for measurement. Cells that were

not labelled with the primary antibody were used as negative control. Excitation and

emission wavelengths of the used fluorochrome were resspectively: FITC: 494, 519; PE:

496/546,578; PE-CY7:496/565,774; APC: 650, 660 and Alexa Fluor-647: 650, 668. The

stained samples were measured with a FACSCallibure (BD Biosciences). Data analysis

was performed using FlowJo software (Tree Star, San Carlos, CA). β

3.4.4. Cytokine production measurement

The cytometric bead array (CBA) human inflammatory kit (BD Biosciences) was used

for the quantitative measurement of the cytokine production and secretion by HCN2 cells

cultured under control and lead-exposed conditions. For that purpose, HCN2 cells were

incubated with lead 10 µM for 24 hours 37oC. The following day, after detaching cells

using cell scraper and washing twice with PBS, cells suspension were centrifuged to

obtain cell-free supernatant. Afterwards, 50-100 µl of the supernatants were collected

and stored at -20oC. The assay was performed according to the manufacturer’s instruction

using 50 µl of the human inflammatory cytokine standards and sample dilution. Cytokine

concentrations were measured using the FACSCanto flow cytometer (BD Bioscience).

FCAP Array version 3 software was used to analyse the data. Measurements were done

in duplicates and repeated with three cellular experimental repeats (n=3).

3.4.5. Statistics

Data were statistically analysed through the SPSS statistics 17.0 software. A one-way

ANOVA and uni-variant linear model were used to compare means. The LSD post-hoc

Page 62: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[41]

test was also used to determine the significant difference between groups. In all the

analysis P<0.05 were considered as statistically significant. Graphs with error bars

represent the standard deviation (SD).

3.5. Results

3.5.1. Characterization of human neuronal cells HCN2

The cortical neuronal cell line HCN2 has been established and reported as convenient

human neuronal precursor in vitro model (Zhang et al., 1994, Ronnett et al., 1994).

However, they have not been often used for research and only few reports have been

published. Therefore, these cells have not yet been characterized thoroughly.

Consequently, we examined the neuronal precursor properties of these cells. For this

purpose, we seeded HCN2 on glass cover slips under standard culture conditions for 7

days to cover 50-60% of the surface. It needs to be pointed out that HCN2 is very slow-

growing cell line and this range was the maximum could be reached over the 7 days.

Following fixation, the cells were stained with different markers specific for neuronal

precursor. Images were taken using the fluorescent microscopy. Nestin and β-tubulin III,

which are the significant markers of neural precursor cell, were highly expressed by the

immature HCN2 cells and decreased after induction of neuronal maturation upon nerve

growth factor (NGF) stimulation (Figure 3.1 A, B and C). We also tested expression of

nerve growth factor receptor p75 (P75 NGFR) and fractalkine. HCN2 expressed both

NGFR and CX3CL1, (Figure 3.1 D and E). No expression of Iba-1, a

microglia/macrophage marker was detectable in HCN2 (Figure 3.1 F). All these results

implicate that cultured HCN2 under basal (non-differentiated) condition show the

neuronal-restricted precursor properties, including expression of neural precursor

markers (nestin, vimentin, β-tubulin III), as well as specific morphology with elaborated

flat polygonal cell body with short neuritic processes. For further characterization, we

also studied the chemokine receptor expression patterns in HCN2 under non-lead treated

condition (Section 3.5.3).

Page 63: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[42]

Figure 3.1: Expression of neuronal markers by HCN2.

Fluorescent microscopy images represent the expression of β-tubulin III (A), nestin (B), vimentin

(C), NGFR (D), fractalkine (E), and Iba-1 (F) in HCN2. Green and blue colours represent the

expression of fluorescent markers and nucleus, respectively. Neural precursor markers (β-tubulin

III, vimentin and nestin) were expressed at higher levels, showing the precursor characteristics of

HCN2. Bar indicates 100 µM (n=3).

Page 64: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[43]

3.5.2. Lead effects on HCN2 morphology and viability

Several animal models have shown neuronal apoptosis and decreased number of neurons

after lead exposure in the developing brain (Tang et al., 2010).To verify whether lead

affects human neuronal cells in vitro, we studied the effect of lead on the viability of

HCN2 using the MTS assay (Figure 3.2). After 5 days in culture, HCN2 were treated

with various concentration of lead (1, 10 and 100 µM) for 24 hours. Viability of the non-

treated cells (control group) was considered as 100% (baseline) and the experimental

groups were correspondingly compared to the control. NGF and LPS were included as

negative and positive controls, respectively.

Interestingly, only high concentration of lead exposure at 100 µM reduced significantly

the viability of the HCN2 cells (P<0.05), whereas low lead concentrations (1 and 10 µM)

increased slightly, but not significantly the viability of the HCN2, similar to our positive

neurotrophic control agents, NGF and fractalkine (P>0.05). However, low lead

concentrations (≤10 µM) did not have a neurotoxic effect on HCN2 cells.

In the context of morphology, un-differentiated HCN2 showed the flat polygonal

morphology with few short-branched processes in the absence of lead. No significant

changes in morphology and number of cells were seen in presence of ≤10 µM

lead(Figure 3.3 A-B), although increases in length and number of branches as well as

typical differentiated morphology of neurons were observed in a number of cells in

culture in higher concentartion of lead (100 µM) (Figure 3.3 C-D).

Figure 3.2: Effect of various concentration of lead on viability of HCN2 using the MTS assay.

Bar charts represent the viability of HCN2

when challenged with 1, 10 and 100 µM

lead for 24 hours (black bars) when

compared to the non lead-treated cells as

negative control (white bar). HCN2 were

cultured in 96-well plate in standard

culture medium for 5 days in 37oC. Then,

cells were incubated with lead for 24

hours. Cells were also challenged with

LPS (10 ng/ml), fractalkine (10 ng/ml)

and NGF (10 ng/ml) for 24 hours as

positive controls. Viability of non-treated

cells considered as 100%. There were no

significant changes were seen in presence

of lower concentration of lead (1 and 10

µM).Only 100 µM lead reduced HCN2

viability at the significant level (n=3)

(P<0.05). Colorimetric measurements

were analyzed statically using the ANOVA and Dennett’s tests.

Page 65: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[44]

Figure 3.3: Effect of lead exposure on HCN2 morphology

HCN2 were cultured in DMEM supplemented with 10 % FCS for 10 days (A). Then, cells were

exposed to 10 µM lead (B) and 100 µM (C and D).for 24 hours, 37oC. Images were taken using the

phase contrast microscope (n=5). Bar indicates 100 µM (A-C) and 200 µM (D).

3.5.3. Effect of lead exposure on expression of chemokine receptors in

HCN2

As mentioned above, a variety of chemokine receptors are expressed by HCN2 cells.

Results showed that HCN2 expressed most of the chemokine receptors at moderate

levels. However, among the expressed receptors, high expression levels were found for

CCR1, CCR2, CCR3, CCR4 and CCR5 (MFI ± SD: 40.06 ± 2.09, 66.68 ±1.64, 114

±4.35, 25.4 ± 1.54 and 409.4 ± 37.9) (Figure 3.5 A-E). Both fractalkine and CX3CR1

(Figure 3.5 J and K) were expressed at high levels (101.2±10.23 and 175.8±17.07) In

accordance with immunohistochemistry data, high expression of β-tubulin III was also

shown by flow cytometry (201.62 ± 14.42) (Figure 3.4 A). As expected, low affinity

nerve growth factor (NGF) receptor (NGF-R p 75) was shown to be expressed by HCN2

at low levels (10.7 ± 1.27) (Figure 3.4 B). According to the important role of chemokine-

Page 66: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[45]

receptor axis in neuronal development, migration and even inflammation, we studied

possible changes in chemokine receptorexpression patterns after exposure to lead for 24

hours. Figure 3.5 summarizes the expression pattern of chemokine receptors in the

presence or absence of lead. Following 10 µM lead exposure, expression levels of CCR1,

CCR2, CCR3 and CCR5 drastically declined (MFI from 40.06 ± 2.09 to 23.86 ±1.96,

66.68 ± 1.64 to20.7 ± 2.54, 114 ± 4.35 to 48.08 ± 5.4, and 409.4 ± 37.9 to 67.64 ± 4.32)

(P=0.034, 0.005, 0.024, 0.020 respectively). Lead did not induce significant changes in

the expression of CCR9 nor CXCR4 (p>0.05). However, CCR4, CXCR1 and CXCR3

expression increased significantly in lead treated HCN2 (MFI from 25.4±1.54 to 60.54±

5.54, 51.22 ± 3.87 to84.52 ± 8.76 and 30.92 ± 1.17 to 42.16 ± 1.18) (P= 0.038, 0.023 and

0.037). Lead-exposed HCN2 showed significant down-regulation in β-tubulinIII

expression (from 201.62 ± 14.42 to 21.42 ± 2.3) (P=0.006). In terms of

fractalkine/CX3CR1 expression, lead exposure induced expression of membrane-bound

fractalkine while the expression of CX3CR1 decreased (MFI from 101.2 ± 10.23 to 239

± 14.2 and 175.8 ± 17.07 to71.32 ± 5.4) (P=0.017 and 0.024).

The effect of low level of lead exposure on the expression of β-tubulin III as young

neuronal marker and the p75 NGF receptor in HCN2 was also investigated. Result

showed that non-lead treated HCN2 expressed β-tubulin III at high level of intensity,

however, 10 µM lead exposure down-regulated significantly the expression of β-tubulin

III (P=0.006) (Figure 3.4. A). Lead had no significant effects on expression level of NGF

receptor p75 in HCN2, although a slight decrease was observed in lead treated cells

comparing with the control group (Figure 3.4. B).

Figure 3.4: Effect of lead exposure on expression pattern of neural marker.

Representative histograms of one

experiments showing the comparison

between the expression level of β-

tubulin III (A) and NGF-R (B) in

HCN2 in presence and absent of lead

(black and white histograms

respectively).Cells were incubated

with 10 µM lead for 24 hours at

37oC. Then, mean fluorescent

intensity of tested markers were

measured using the flow cytometry.

Data were analyzed using the FlowJo

software Analysis of the significant

differences between lead-treated cells

and non-treated cell was done by

Student’s t-test. (n=5).

Page 67: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[46]

Figure 3.5: Effect of lead on expression patterns of chemokine receptor in HCN2.

Flow cytometry data of one representative experiments is shown (n=5). Histograms represent the

expression level of CCR1-CCR5 (A-E), CCR9 (F), CXCR1 (G), CXCR3 (H), CXCR4 (I),

CX3CL1 (J) and CX3CR1 (K) in lead-treated HCN2 (black) comparing to control cells (white).

Cells were incubated with 10 µM lead for 24 hours at 37oC. Data were analyzed using the FlowJo

software Analysis of the significant differences between lead-treated cells and control ones was

done by Student’s t-test.

Page 68: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[47]

3.5.4. Effect of lead on cytokine secretion in HCN2 cells

Up-regulation of pro-inflammatory cytokines in the brain in response to lead exposure, as

well as in induced neuronal injuries and apoptosis has been reported in in vivo and in

vitro animal models (Struzynska et al., 2007). However, there is not much known about

possible changes of cytokine secretion patterns after lead exposure on human neuronal

precursors. Therefore, we studied whether lead exposure altered the expression of pro-

inflammatory cytokines. We measured the secretion of IL-1β, IL-6, IL-8, IL-10, IL-12,

TNF- and IFN-γ in lead-treated HCN2 compared to the control group. Results showed

that non-treated HCN2 secreted IL-8 and IL-6 (Figure 3.6). After exposure to 10 µM lead

for 24 hours, IL-8 expression remained unchanged, while IL-6 secretion decreased

significantly. HCN2 in both experimental condition, in the presence or absence of lead,

showed non-detectable secretion level of IL-10, IL-1, TNF- and IL-12 (n=4).

Figure 3.6. Effect of lead on cytokine secretion in HCN2.

HCN2 were incubated with 10 µM lead for 24 hours. Various secreted cytokines were measured in

collected culture media using the cytokine bead cytometric assay. Non-lead treated cells were

considered as control. Data was analyzed by FCAP software. Final concentration are shown in

pg/ml (n=4), * indicates P<0.05.Data analyzed using the Student’s t-test.

0

1000

2000

3000

4000

5000

6000

7000

IL-8 Il-6

Fin

al c

on

cen

trat

ion

(p

g/m

l)

control

pb

* p=0.000

Page 69: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[48]

3.6. Discussion

In this study, we investigated the effect of lead exposure on morphology, expression

pattern of chemokine receptors and cytokine secretion in the human cortical neural cell

line HCN2. To our knowledge, there are no published studies on HCN2 and lead. We

showed that at low concentrations (≤10 µM), lead had no effect on the morphology and

viability of HCN2. Lead altered the expression patterns of chemokine receptors and

cytokines in HCN2 with significant changes on expression of fractalkine/CX3CR1 axis,

as well as IL-6 secretion.

Lead is a well-known neurotoxicant with potent detrimental effects on the development

and function of the CNS. Lead exposure is still treated as a major global health problem

especially in children with long-lasting cognitive and behavioral impairment(Lidsky and

Schneider, 2003).

In humans, the CNS development begins in the third gestational week with proliferation,

differentiation and migration of neuronal precursor cells (NPCs) and continues in the

postnatal periods. The blood-brain barrier is not fully developed until the middle of the

first year of life (Stiles and Jernigan, 2010). The number of synaptic connections between

neurons reaches a peak around the age two and is then trimmed back to about half of it.

Similarly, there is great postnatal activity in the development of receptors and transmitter

systems, as well as in the production of myelin. All these facts support the sensitivity of

the developing brain to lead exposure (Rodier, 1995, Rodríguez-Martínez et al., 2012).

Recent studies demonstrated that low level of lead exposure produces blood lead levels,

near the accepted international safe limit, and triggers a wave of apoptotic

neurodegeneration in cortical and subcortical neurons in the neonatal mouse brain

(Dribben et al., 2011). There is also evidence from animal studies showing that short-

term exposure to moderate level of lead during neurogenesis can influence cell survival

and the process of development, by affecting neuronal precursor cells and by altering the

cortical arrangement (Davidovics and DiCicco-Bloom, 2005). Over the past decades,

several studies have reported lead-induced neurotoxicity in human and animals, using in

vivo and in vitro models. However, not many of them investigated the effect of lead on

human NPC.

Human cortical neurons are one of the principal cell types affected in neurotoxicity and

several neurodegenerative diseases. Due to their importance in healthy brain function, in

vitro and in vivo animal and human cortical neurons have been used commonly as

appropriate model, in neurotoxicity research (Schmuck et al., 2000). HCN2 are human

Page 70: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[49]

cortical neuronal non-transfected cells obtained from a 7 year old girl with Rasmussen's

encephalitis. Undifferentiated HCN2 showed characteristic of neural restricted precursor

cell including expression of neural precursor markers such as nestin, vimentin and β-

tubulin III, as well as polygonal flat soma with number of short neuronic branches

process resembling the immature/undifferentiated neuron morphology. These results are

in accordance with previous published data (Ronnett et al., 1994). HCN2 as immature

cortical neurons expressed the nerve growth factor receptor-gp75 and membrane-bound

fractalkine. HCN2 did not express Iba-1, a common microglia marker, which made it a

suitable in vitro neuronal precursor model for our study. Nerve growth factor (NGF) is a

well-characterized neurotrophic factor that plays a crucial role during development in

growth, differentiation and maintenance of brain neurons, as well as in the reparative

responses of the adult brain to damage. Expression of neurotrophin receptors by

neurosphere cultured from embryonic precursor cells has been reported (Lachyankar et

al., 1997, Maisonpierre et al., 1990). It has also been reported that NGF can induce the

differentiation of HCN2, which is in agreement with our data. Thus, based on our results,

undifferentiated HCN2 showed the properties of neuronal restricted precursor cells.

In the next step, we investigated the effect of lead exposure at different concentration (1,

10 and 100 µM) on the morphology and viability of the HCN2. HCN2 at the non-

stimulated condition showed the flat polygonal morphology with few short spin–like

processes. Piper et al. (2000) has reported that nestin positive human neuronal precursor

cells in an undifferentiated condition appeared morphologically flat with few processes,

and they formed tight clusters. When cells were replated into differentiation conditions,

cells altered their morphology and generated long processes (Piper et al., 2000,

Kerkovich et al., 1999). HCN2 showed no changes in morphology after exposure 0, 1 or

10 µM. However, in the presence of 100 µM lead, few cells showed the bipolar cell body

with long processes resembling the more differentiated cells. There is no evidence

showing the effect of lead in morphology of human neuronal precursor cells. However,

morphologic changes were observed in the rat hippocampus following low level (< 0.5

µM) exposure to lead such as a significant increase in the size and numerical density of

the dendrites and granulr cells. The opposite effects (i.e. decreased density of cell layers)

were observed at much higher concentration of lead, suggesting a bimodal effect of lead

on the developing hippocampus (Slomianka et al., 1989, Finkelstein et al., 1998). Lead

involvement has also been suggested to potentiate the PC12 morphological

differentiation that may be related to lead-induced alteration in binding transcription

factor to DNA (Crumpton et al., 2001).

Page 71: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[50]

As we did not observe any significant changes in morphology, we studied whether lead at

the low concentration could alter the HCN2 viability. Several in vitro and in vivo studies

have shown detrimental effects of lead on viability of CNS cells through different

mechanisms such as alteration of the cytoskeletal reorganization, increase in oxidative

stress, up-regulation in expression of apoptotic proteins and disturbance intracellular

Ca2+

(Choi et al., 2011, Penugonda et al., 2006). However, on the contrary, some studies

showed an inhibitory effect of lead on neurite outgrowth both, at high and low

concentration, with no effect at the intermediate levels (Kern et al., 1993, Schneider et

al., 2003). Thus, there is still debate on toxic effects of lead on neuronal cells depending

on the different concentration, exposure time, the roigin of the brain region cells and

developmental stage. Similar to previous studies in rat cortical neurons, no significant

toxic effect of lead (1 and 10 µM) on cell viability of HCN2 was observed in our study

(Fujimura and Usuki, 2012). In this study, we have also reported that lead could induce

cytotoxicity in HCN2 in vitro at the higher concentration (100 µM), over short period of

exposure (24 hours) a concentration which is not expected to be reached in vivo. A

similar cytotoxic effects of higher concentration of lead has been reported for rat cortical

neurons (Tang et al., 2010). It needs to be pointed out that MTS assay was used in this

study as a standardized cellular viability and cytotoxicity test. However, as MTS assay is

reflecting the cell death through indirect measure of the cell mitochondrial activity,

additional cytotoxic studies need to be conducted to investigate the exact cytotoxic

effects of lead on HCN2 viability. Functional chemokine receptors and chemokines are

expressed by glial cells within the CNSHowever, relatively little is known about the

patterns of chemokine receptor expression and function in neurons (Flynn et al., 2003,

Cartier et al., 2005). Recent studies showed that chemokines and their receptors play an

important role in regulating human neural stem cell (NSC) proliferation and survival. In

addition, there is not much known about the exact role of chemokine receptors in the

CNS development. It has been suggested that chemokines could have both stimulatory

and inhibitory effects on NPC proliferation (Coughlan et al., 2000, Krathwohl and

Kaiser, 2004). The expression of CCR1, CCR2, CCR5, CXCR3, CXCR4 and CX3CR1

has been reported in dividing adult mouse, rat and human NPC, which support our data

in HCN2 (Goczalik et al., 2008, Hesselgesser and Horuk, 1999, Ji et al., 2004, Wu et al.,

2012, Tran et al., 2007). We have observed the expression of all tested chemokine

receptors by HCN2 at a moderate to high level which suggests their involvement in

normal brain development, neurogenesis (i. e. regulating the migration of progenitor cells

in postnatal brain), as well as neuroinflammation. Several studies reported inhibitory

effects of chemokine receptors such as CCR3 and CXCR4 on neuronal precursor

proliferation and on neurogenesis (Krathwohl and Kaiser, 2004). In addition, activation

Page 72: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[51]

of pro-inflammaory chemokine receptors (CCR1, CCR5, CXCR4) have been reported to

play a role in neurodegenerative diseases, which implies their involvement in

inflammatory responses and induce neuronal death (Petito et al., 2001, Kaul et al., 2007).

In contrast, there is evidence showing a beneficial role of chemokine receptors on

neurogenesis during development. Several studies reported migration of neural

precursors to the site of inflammation in response to chemokine followed by injuries,

stoke, etc., to induce neurogeneration.

In this study, we have seen differential changes in expression patterns of chemokine

receptors following lead exposure. Most of the pro-inflammatory chemokine receptors,

such CCR1, CCR2, CCR3, CCR5 and CXCR4, showed down regulation, with one

exception, CXCR1, which significantly increased. Unlike other chemokine receptors,

there is little known about expression of CXCR1 in neurons. Expression of CXCR1

mRNA transcripts has been reported in glutamatergic, GABAergic, and cholinergic

neurons in the rat brain. Recently, Goczalik et al. (2008), reported for the first time the

prominent expression of CXCR1 in healthy human retina neuron. It has also been

suggested that CXCR1 and its ligand IL-8 play an important role as neuromdulator in

CNS development, neuronal survival, modulation of excitability, and neuroimmune

response (Danik et al., 2003). Increased expression of CXCR1 as well as IL-8 in human

retina has been reported in response to inflammation in proliferative viteoretinopathy

(PVR). Thus, during inflammation, elevated levels of IL-8 may activate neuronal

CXCR1/CXCR2 receptors to exert neuroprotective effects.

Among the neuronal chemokine/receptors, fractalkine and its unique receptor play a key

role in both, neuroprotection and neuroinflammation. The chemokine fractalkine, which

is the CX3CR1 ligand, is highly expressed in CNS neurons and is normally produced in a

membrane-bound form, from which the chemokine domain can be released (Harrison et

al., 1998). Over the past decades, it became clear that the interaction between neuronal

fractalkine and microglial CX3CR1 modulates microglia activation. Several studies have

shown the neuroprotective role of fractalkine in neurotoxicity models associated with

neurodegenerative diseases (Mizuno et al., 2003, Cardona et al., 2006). There is evidence

showing the correlation between decline in neurogenesis and reduced fractalkine in the

aged animal hippocampus (Bachstetter et al., 2011). Recent animal studies showed the

beneficial effects of increased fractalkine within hippocampus on NPC activation, which

in turn modulated microglia activation (Vukovic et al., 2012). Although expression of

CX3CR1 is thought to be restricted to microglia and astrocytes, recent findings showed

its expression by neurons which support our finding that fractalkine is expressed in

Page 73: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[52]

HCN2. The expression of CX3CR1 by hippocampal neurons indicates that these

receptors may also be a target for fractalkine produced by microglia and/or astroglia or

adjacent neurons. Despite the known paracrin interaction, recent studies suggested a

neuroprotective effect of fractalkine on neuronal survival and proliferation through

autocrine interaction (Meucci et al., 2000). Interestingly, we have seen a differential

effect of lead on fractalkine and CX3CR1 in HCN2. While lead suppressed the

expression of neuronal CX3CR1, it enhanced fractalkine expression. Thus, fractalkine

may interact with CX3CR1 expressed on neurons and other cells, including-like glia.

In this study, we also observed a significant reduction in expression of β-tubulin III.

In support of these results, it has been reported that cytoskeletal protein class III β-

tubulin isotype, a specific neuronal marker, is abundantly expressed in CNS during fetal

and postnatal development. Indeed, there is evidence indicating during brain maturation

alpha and beta tubulin expression is abundantly decreased (Bond and Farmer, 1983). In

addition, different expression patterns of β-tubulin subclasses during CNS development

has been noted. As exemplified, high expression level of class III β-tubulin was shown in

rat cortical neurons at the birth time which abundantly decreased during development and

maturation. While in dorsal root ganglia, β-tubulin III generally increased in postnatal

development (Jiang and Oblinger, 1992). In vitro and in vivo animal studies showed the

reduction in neurite outgrowth and axonal regeneration in knock-down β-tubulin III (Guo

et al., 2010). In this study, lead also reduced NGF receptor expression. Expression of

both low and high affinity receptors for NGF have been shown by NPC which implicates

their involvement in development and neurogenesis.

In this study,we reported that HCN2 under non-stimulatory condition secret IL-8.

However, no changes in secretion level of IL-8 in the supernatant of HCN2 was detected

following lead exposure. As mentioned previously, HCN2 expressed its receptor,

CXCR1 at non-stimulatory condition, which may implicate the role of this chemokine in

sustaining survival and growth of neuronal precursors, as well as during development.

Lead altered the receptor expression with no changes in secretion levels of IL-8.

We also observed the significant decrease in IL-6 secretion by HCN2 cells in the

presence of lead. It has been reported IL-6 has a neuropoitic effect on the NSC self

renewal and differentiation. On the other hand, in inflammatory conditions, enhanced

expression of IL-6 results in attenuation of NPC proliferation and differentiation. (Lan et

al., 2012). Microglia-derived IL-6 has also been shown to have suppressive effects on

neurosphere generation from adult human retina cells (Balasubramaniam et al., 2009).

Transgenic mice with over-expression of IL-6 showed alteration in hippocampal function

Page 74: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[53]

through changes in excitability of hippocampal neurons and synapses (Nelson et al.,

2012). Recent studies postulate that IL-6 does not have only influences on neurons

directly, but also is an important intercellular messenger in the neuronal-microglia

interaction.

3.7. Conclusion

To our knowledge, this is the first study reporting on the impact of lead exposure over a

short period of time on the human cortical neuronal cell line HCN2. In summary, lead

exposure (10 µM, 24 hous) had no cytotoxic effect on HCN2 cells. However, lead altered

the expresion patterns of chemokine receptors and IL-6 in HCN2. Lead caused changes

of these immune and neurogenesis related molecules and may affect the neuronal

communication with other cells, including neuronal precursors in areas important for

brain remodeling and development, which may finally be responsible for the neurotoxic

effects of lead.

Page 75: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

Chapter 4

A novel in vitro human microglia model:

Characterization of human monocyte-

derived microglia

Page 76: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[55]

4. A novel in vitro human microglia model: characterization of human monocyte-derived microglia

4.1. Introduction to chapter

Microglia are the resident innate immune cells of the brain. As the brain macrophages,

microglia share common characteristics with the monocyte-macrophage lineage such as

expression of surface markers and chemokine/cytokine receptors, antigen presentation

and phagocytic activity. In response to stimuli such as pathogen, injuries, etc, resting

microglia with ramified morphology can become activated with amoeboid morphology.

This activation is usually followed by secretion of pro-inflammatory cytokine sand

chemokines as mediators of the innate immunity. Despite their known contribution to

pathological conditions, microglia also play a key role in CNS development and

neurogenesis through secretion of neurotrophic factors (Kettenmann et al., 2011,

Ransohoff and Perry, 2009, Kim and de Vellis, 2005, Chan et al., 2007).

To date, there is no standardized simple model available to investigate the biology of

human microglia. Consequently, the aim of this study was to establish a new in vitro

microglia model using blood-derived precursor cells. For that purpose, human peripheral

blood monocytes were cultured in serum free medium in the presence of a mixture of

cytokines and chemokines (M-CSF, GM-CSF, NGF and CCL2) to generate monocyte-

derived microglia (M-MG). Monocyte-derived dendritic cells (M-DC) were also

generated as a control population using GM-CSF and IL-4. The human microglia cell

line HMC3 was also used as control.

M-MG were clearly different in morphology, phenotype and function from M-DC, but

shared many properties with HMC3 cells. M-MG acquired a ramified morphology with

primary and secondary processes, comparable to HMC3. They expressed very low levels

of CD45, CD14 and HLA-DR, CD11b and CD11c; but a distinct pattern of chemokine

receptors, including CCR1, CCR2, CCR3, CCR4, CCR5, CXCR1, CXCR3, CX3CR1.

Similar to HMC3, under non-activated condition, the M-MG secreted of IL-8 and IL-6.

In comparison with M-DC, M-MG displayed lower T-lymphocyte stimulatory capacity,

as well as lower phagocytosis activity.

In summary, we have established a new protocol for the generation of human monocyte-

derived microglia, which is is feasible, well standardized and reliable, as it uses well

defined culture medium and recombinant cytokines, but no serum or conditioned

Page 77: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[56]

medium. This model will certainly be very helpful for future studies investigating the

biology and pathology of human microglia.

Declaration of work

All the experimental work were designed and conducted by SE including cell isolation,

cell culture, characterization, flow cytometry studies, functional assays, phagocytosis

tests and statistical analysis under supervision of Prof. Luis Filgueira. Confocal images

were taken with assistance of Mrs. Rasheeda Mohd Zamin. All the experimental work

described in this chapter has been published as manuscript in the Journal of Neuroscience

Methods (Appendix E).

Page 78: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[57]

4.2. Abstract

Microglia as the immune modulator cells of the CNS have the key functional roles both

in maintaining the healthy physiology of the brain and contributing in inflammation

responses especially in neuroinflammation–related pathogenesis. The aim of this study is

to generate the human microglia from blood monocyte .Microglia originate from distinct

sub-population of the monocytic lineage and share expressing various phenotype markers

of monocyte-macrophage lineage such as surface molecules, chemokine/cytokine

receptors, antigen presentation. To generate human microglia, PBMC were cultured in a

serum free condition using a novel mixture of four recombinant human cytokines, i.e. M-

CSF, GM-CSF, NGF and CCL2. These monocyte-derived microglia in presence of

mixture of cytokines acquired ramified morphology of with primary and secondary fine

processes. In context of surface marker expression, M-MG revealed different expression

in compare to monocyte. Microglia like cells showed down-regulation in expression level

of monocytic lineage markers (CD45, CD14, HLA-DR) and chemokine receptors, i.e.

CCR1, CCR2 and while up-regulation was detected in expression of CXCR3 and

CX3CR1. In light of these data, we developed a new in vitro protocol to generate human

microglia representing the specific morphological, phenotypic and functional properties

of the resting microglia. The described protocol is feasible, well standardized and

reliable, as it uses only well defined culture medium and recombinant cytokines, but no

serum or conditioned medium. This protocol will certainly be very helpful for future

studies investigating adult human microglia.

Keywords:Microglia, human peripheral blood monocytic cells, serum-free, characterization

4.3. Introduction

Microglia are resident innate immune cells of the central nervous system (CNS) and play

a crucial role in maintaining the healthy physiological homeostasis. Microglia also

contribute substantially to inflammation in response to injury, toxins, pathogens and

degenerative processes (Streit, 2001, 1996; Howell et al., 2010; Lee et al., 2010; Politis et

al., 2011). Microglia are characterized by a quick response to various stimuli, resulting in

activation with rapid changes in morphology, phenotype and function. Morphological

changes include shortening of cell processes and cellular hypertrophy. To date, there is

no specific microglia marker clearly separating this cell type from other cells of the

monocyte/macrophage lineage. Some studies suggested that primary microglia could be

distinguished from other tissue macrophages according to expression levels of markers

such as CD11b and CD45 (Aloisi et al., 2000; Ford et al., 1995). Under healthy

Page 79: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[58]

condition, resting microglia with typical ramified morphology show low expression

levels of these common myeloid lineage markers (Nimmerjahn et al., 2005). However,

microglia have been shown to share expression of surface markers common to other

immune cells of the macrophage family such as CD45, CD14, MHC-class II, CD68,

immunoglobulin Fc receptors and 2 integrins (Lambertsen et al., 2009; Wirenfeldt et al.,

2005). Expression levels of these surface markers may depend on the inflammatory status

of the CNS and on microglia activation. Similarly, functional inflammatory changes are

dominated by secretion of pro-inflammatory cytokines, chemokines, neurotrophic factors

and up-regulation of corresponding receptors, as well as production of nitric oxide and

reactive oxygen intermediates (Aloisi, 2001; Tambuyzer et al., 2009).

Microglia have recently been shown in a mouse model to originate from a myeloid yolk

sac population during embryogenesis (Ginhoux et al., 2010). In humans, the microglia

origin is still not known. Furthermore, little is known about the turnover rate of microglia

in the healthy CNS. It has been postulated that proliferation of the resident microglia

population is rather low and there may be immigration of bone marrow derived precursor

cells into the CNS. Invasion of bone-marrow derived microglia have been shown to

migrate into the CNS using chimeric irradiated mice (Simard et al., 2006; Zhang et al.,

2007). However, the proportion of bone-marrow progenitor cells in replenishment of

microglia is controversial.

A variety of cytokines have been shown to contribute to microglia development and

differentiation. Colony stimulating factor-1/M-CSF and its receptor CSF-1R play an

important role in the development of macrophage populations (Chitu and Stanley, 2006).

Recently, Ginhoux et al. (2010) reported the constant absence of microglia throughout

life in CSF-1R deficient mice, indicating that M-CSF is essential for microglia

development and differentiation. In addition, granulocyte-macrophage colony

stimulating factor (GM-CSF) also contributes to microglia development and

differentiation (Aloisi et al., 2000; Esen and Kielian, 2007). Both, M-CSF and GM-CSF

have crucial effects on proliferation and survival of primary human fetal and adult

microglia in culture with GM-CSF having a greater impact on proliferation (Esen and

Kielian, 2007; Lee et al., 1994). M-CSF plays a crucial role in the final maturation stage

of microglia. In op/op mice, a model of human osteopetrosis with a functional mutation

the M-CSF gene, the number of microglia are decreased and the cells are smaller and

have impaired activation ability in response to injury (Kalla et al., 2001;Wegiel et al.,

1998). Importantly, GM-CSF or IL-3 does not agile substitute for M-CSF (Blevins and

Fedoroff, 1995).

Page 80: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[59]

Nerve growth factor (NGF), a member of neurotrophins, has also been shown to act on

the proliferation and survival of microglia (Zhang et al., 2003). NGF binds and acts

through the P75 receptor, which has been shown to be expressed in microglia in multiple

sclerosis lesions (Valdo et al., 2002). It has also been shown that microglia express the

NGF receptor TrkA (Tonchev, 2011). NGF induces migration of the microglia cell

through activation of the TrkA (TrkA activation) (De Simone et al., 2007). NGF is

expressed by activated microglia, astrocytes and hippocampal neurons (Friedman, 2000;

Saez et al., 2006; Tonchev, 2011). However, inflammation certainly induces

neurotrophin secretion by human microglia (Heese et al., 1998; Nakajima et al., 2001).

Chemokines have also been shown to play an important role in microglia biology.

Monocyte chemoattractant protein-1 (MCP-1), or CCL2, is one of the prominent

chemokines in the regulation of the microglia migration to the site of inflammation in

experimental models (Leonard et al., 1991; Zhang et al., 2007). CCL2 acts through its

specific receptor CCR2 which is expressed by microglia and other cells of monocytic

lineage, such as macrophages and dendritic cells (Rebenko-Moll et al., 2006). CCL2 acts

also through CCR4, but little information is available about CCR4 expression in

microglia (Craig and Loberg, 2006; Zhang et al., 2006). Studies on normal rat CNS

showed that CCL2 is constitutively expressed by astrocytes and neurons and it can be

found in various brain regions, including the cerebral cortex, the hippocampus and the

hypothalamus (Banisadr et al., 2005). CCL2 is also secreted by astrocytes and neurons

under inflammatory condition (Banisadr et al., 2005; Farina et al., 2007; Tanuma et al.,

2006). It has been shown that CCL2 is secreted by both undamaged and damaged spinal

sensory neurons in rat, resulting in activation of spinal microglia and initiating

neuropathic-like pain (Thacker et al., 2009). Up-regulation of CCL2 expression during

Alzheimer’s disease and multiple sclerosis correlates with activation of microglia and

may contribute to pathogenesis of neurodegenerative diseases (Conductier et al., 2010;

Simpson et al., 2000). In that respect, CCL2 contributes to recruitment of mononuclear

cells into the inflamed CNS, followed by activation of the microglia. Zhang et al. (2007)

showed that CCL2 can induce spinal microglia activation in mice. They also reported in

chimeric mice that CCL2 recruits bone marrow-derived macrophages, which proliferate

and differentiate into microglia in the spinal cord and induce inflammation and

microgliosis after nerve injury. In experimental autoimmune encephalomyelitis (EAE), a

murine model of multiple sclerosis, CCR2 deficient mice do not develop the

mononuclear cell infiltration and inflammation. Indeed, no chemokines increase was

detected in the CNS in that model (Izikson et al., 2000). On the other hand, CCR2

deficiency in a mouse model of Alzheimer’s disease impairs significantly microglia

Page 81: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[60]

accumulation at sites of plaque formation, resulting in a decrease of amyloid-β clearance

and in accelerating early disease progression (El Khoury et al., 2007).

In vitro culture of mouse and human microglia has often been used as a model by various

researchers. Thereby, astrocyte-conditioned medium (ACM) has been used to keep

microglia in culture with a ramified resting morphology. ACM contains M-CSF, GM-

CSF and transforming growth factor (TGF-β), all known to be secreted by astrocytes.

Microglia lose their ramified morphology in the presence of antibodies against the

mentioned cytokines, indicating that M-CSF and GM-CSF are essential for microglia

culturing (Schilling et al., 2001). GM-CSF, even at low concentration in serum-free

medium, keeps microglia at a resting state with morphological ramifications (Fujita et al.,

1996). A recent study reported the proliferative effect of CCL2 on neonatal and

embryonic primary rat microglia in culture without inducing additional changes in

morphology, phenotype and cytokine expression (Hinze and Stolzing, 2011).

Most interestingly, murine bone marrow-derived precursor cells can be differentiated

towards microglia-like cells when cultured in the presence of astrocytes or in mixed glial

cultures. Thereby, M-CSF alone is not sufficient for microglia differentiation, which

indicates the importance of additional astrocyte-derived factors (Noto et al., 2010).

Differentiation of murine bone marrow stem cells toward microglia-like cells has

recently also been successful, using ACM supplemented with GM-CSF (Hinze and

Stolzing, 2011). Culturing fetal and adult human microglia in the presence of GM-CSF

induces functional maturation towards an antigen presenting cell type, especially in adult

microglia. However, GM-CSF does not induce maturation of microglia to acquire the

complete phenotype and function of mature dendritic cells (Lambert et al., 2008; Re et

al., 2002).

In vitro studies on microglia used mainly primary microglia culture from embryonic or

neonatal murine brains (Bassett et al., 2012; Hinojosa et al., 2011). In human, brain-

derived microglia is difficult to obtain for ethical reasons. In addition, only low numbers

of cells are collected. Also, fetal microglia seems to be quite different from adult

microglia. Only few human microglia cell lines have been generated, including HMO6

cells (Nagai et al., 2005) and HMC3 cells (Janabi et al., 1995). These cell lines cannot be

considered as an optimal model for microglia cells due the significant modification in

morphology and function as result of genetic manipulation and long-term culture.

Therefore, a more convenient and more appropriate model for in vitro microglia human

studies is still missing.

Page 82: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[61]

Most importantly, Leone et al. (2006) showed that human blood-derived monocyte,

cultured with astrocytes-conditioned medium (ACM), acquired the ramified morphology

of microglia and expressed substance P, calcium binding protein Iba-1 and dimly MHC

class II, three typical characteristics of microglia. There are also reports showing

successful differentiation of rat blood monocyte towards microglia using ACM

(Schmidtmayer et al., 1994; Sieverset al., 1994). Recently, Hinze and Stolzing (2011)

showed the differentiation of murine bone marrow stem cells towards microglia-like cells

in the presence of ACM with similar phenotypic and functional properties to primary

brain-derived microglia cultures. In the light of these data, we developed a new in vitro

human microglia model using human blood peripheral mononuclear cells, a serum-free

culture condition and a panel of factors, including M-CSF, GM-CSF, NGF and CCL2.

This new human microglia model has then been assessed for morphology, phenotype,

and function.

4.4. Materials and methods

4.4.1. Cell isolation

Human blood mononuclear cells (PBMC) were isolated from buffy coats (100 buffy

coats were used for this study) of healthy donors (Australian Red Cross Blood Service

(ARCBS), Perth, WA, Australia; ethics approval was granted by UWA and ARCBS)

using Ficoll-Paque (Amersham Biosciences, Uppsala, Sweden) as published before

(Meagher et al., 2005). To obtain monocyte (adherent PBMC), the isolated blood cells

were cultured in T25 tissue culture flasks (Sarstedt, Numbrecht, Germany) (2 × 106 to 5 ×

106

cells/ml) using RPMI-1640 Glutamax medium (Invitrogen, Auckland, NZ)

supplemented with 1% antibiotics/antimycotic (10,000 units/ml penicillin G sodium,

10,000 µg/ml streptomycin sulfate and 25 µg/ml Amphotericin B, Invitrogen). It is

important to note that no serum was used in all microglia cultures. After overnight

incubation (humidified air, 37o

C, 5% CO), non-adherent cells (consisting mainly of T-

lymphocytes) were separated by thorough washing with PBS (Invitrogen) and used for

further experiments. The fresh adherent cells, which were mainly monocytes (>90%),

were used for the generation of microglia (M-MG). Dendritic cells (M-DC) and cultured

monocytes, as well as for comparison with the generated cell populations. All

experiments were done with cells from at least 6 different blood donors, if not otherwise

stated.

Page 83: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[62]

4.4.2. Cell culture

To induce the differentiation of M-MG, adherent PBMC (1×106

cells/ml) were cultured

in 6-well tissue culture plates (Sarstedt) using RPMI-1640 Glutamax supplemented with

1% antibiotic/antimycotic (serum-free condition) and a mixture of human recombinant

cytokines, including M-CSF (10 ng/ml; Pepro-Tech, Rocky Hill, NJ), GM-CSF (10

ng/ml; PeproTech), β-nerve growth factor (NGF-β; 10 ng/ml; PeproTech) and CCL2

(100 ng/ml; PeproTech) at standard humidified culture conditions (37oC, 5% CO) for up

to 14 days. Final yield of M-MG was 6-8×106 cells/60 ml of humanBuffy Coats.The cells

were used for experiments and characterization at different time points as indicated.

Monocytes were also cultured in RPMI-1640 Glutamax supplemented with 1%

antibiotic/antimycotic, but no serum, for up to 14 days as control cell population.

Furthermore, to obtain monocyte-derived dendritic cells (M-DC), adherent PBMC were

cultured in 6-well tissue culture plates (Sarstedt) using RPMI-1640 Glutamax, 1%

antibiotic/antimycotic supplemented with 10% fetal calf serum (FCS; Serana,

Bunbury,WA, Australia) in the presence of human recombinant GM-CSF(10 ng/ml;

PeproTech) and IL-4 (10 ng/ml; Pepro-Tech) for up to 14 days (Meagher et al., 2005).

The cells were used for experiments at different time points, as indicated.

Human microglia clone 3 (HMC3) cells, kindly provided by Prof. Karl-Heinz Krause

(University of Geneva, Switzerland) were cultured in T25 tissue culture flask (Sarstedt)

using DMEM/F12 Glutamax medium (Invitrogen) supplemented with 15% FCS and 1%

antibacterial/antimycotic. The cells were passaged and used for experiments when they

reached 70–80% confluence.

4.4.3. Morphological studies

Morphology and viability (using trypan blue dye exclusion: cell viability was >90% for

all experiments) of M-MG, M-DC and HMC3 during the 2 weeks in culture were studied

using an inverted Leica DM IL phase contrast microscope. Images were taken with

Photometrix CoolSNAP Fx digital camera and processed with NIS-elements F3.0

software. HMC3 and M-MG cells were also stained for expression of Iba-1, a well-

known characteristic marker for microglia cells. For that purpose, cells were grown on

glass cover-slips, fixed in 1% paraformaldehyde in PBS/sucrose and incubated with a

polyclonal rabbit anti-Iba-1 (Santa Cruz biotechnology, Santa Cruz, CA) and a donkey

anti-rabbit Alexa Fluor 488 (Invitrogen). Negative control stains were used by omitting

Page 84: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[63]

the primary antibody against Iba-1. Cell nuclei were stained with DAPI (1 µg/ml in PBS,

4,6-diamidine-2-phenylindole dihydrochloride; Roche Diagnostics, Mannheim,

Germany). Imaging was done using confocal microscopy (Leica TCS SP2 multiphoton

confocal microscope).

4.4.4. Flow cytometry

We studied the expression of specific surface molecules on freshly isolated and cultured

monocytes, M-MG, M-DC and HMC3 using flow cytometry. Cells (1×106/ml per

staining condition) were detached using a scraper (Sarstedt) and washed twice with PBS.

Staining was performed according to protocol recommended by BD. A panel of directly

labeled mouse anti-human antibodies, CD45-FITC, CD14-PE, CD40-PE, CD68-APC,

CD80-PE, CD11c-PE,HLA-DR–PERCP, CD11b-AF488, CD11c-PE, CCR1-AF647,

CCR2-AF647, CCR4-PE-Cy7, CCR5-FITC, CXCR1-PE, CXCR3-AF488

(BDPharmingen, San Jose, CA), CCR6-FITC, CCR7-APC (R&A systems, Minneapolis,

MN), NGFR-FITC (Santa Cruz Biotechnology, SantaCruz, CA) were added at the

supplier’s recommended concentration. Corresponding labeled isotype control antibodies

were included in all experiments. A rabbit anti-CX3CR1 polyclonal antibody was used at

the final concentration of 1 µg/ml (AbD Serotec, Oxford, UK), followed by a secondary

donkey anti-rabbit-AF 647 used at the final concentration of 1:200 (Invitrogen). The cells

were incubated with antibodies for 30 min to 1 h at 4o

C, washed with PBS, resuspended

and fixed with 1% formaldehyde in PBS containing 2% sucrose. PBS-2% sucrose

solution were used in combination with fixative to enhance the preservation of cells

morphology following by fixation (quality of fixation) as well as reduce the background

autofluorescence.The mixed samples were measured with a FACSCallibure (BD

Biosciences). Data analysis was performed using FlowJo software version 7.6.3 (Tree

Star, Ashland, OR).

4.4.5. Phagocytosis assay

Phagocytosis was assessed by measuring the uptake of tetra-methyl rhodamine (TMR)-

labeled Staphylococcus aureus (Invitrogen) as published before (Meagher et al., 2005).

Bacterial particles were added to the cells (107 bacteria/ml) at a ratio of 1:1000

(according to manufacturer’s protocol) and incubated for 4 h at 37oC. Cells were then

harvested and fixed with 1% paraformaldehyde in PBS/2% sucrose. Total ingested

fluorescent-labeled bacteria per cell was measured by flow cytometry. To verify that the

bacteria were located intra-cellularly, cells were cultured on glass coverslips, treated with

labeled bacteria, fixed and visualized using confocal microscopy (Leica TCS SP2

Page 85: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[64]

multiphoton confocal microscope). Dendritic cells and freshly isolated monocytes were

used as positive control populations. Untreated cells were used as negative controls.

4.4.6. Mixed leukocyte reaction (MLR)

The T-lymphocyte stimulatory capacity and corresponding antigen-presenting cell (APC)

properties of M-MG, in comparison with M-DC and HMC3, was assessed in MLRs

using a BrdU proliferation assay (Cadosch et al., 2010). Freshly processed non-adherent

PBMC containing mainly (>90%) T-lymphocytes (2 × 105 cells/well) were seeded in a

round bottom 96-well plate (Sarstedt). Allogeneic M-MG, M-DC and HMC3 were used

as antigen-presenting cells (APC) and added at various ratios (1:40 and 1:80) to the T-

lymphocyte (four wells for same condition). T-lymphocytes or APC only were used as

control conditions. To compare the effect of lead on activated T-lymphocytes, PHA was

added to each well at the concentration of 7 µg/well. PHA- activated T-cells with APC

was used as the positive controls. After 5 days in culture, BrdU proliferation assays

(Roche, Mannheim, Germany) were performed according to the supplier’s protocol and

the absorbance of the colorimetric reaction was measured by ELISA reader at 405 nm

(Labsystems Multiskan RC, Helsinki, Finland). Mean proliferation of the different

conditions was statistically analyzed and compared.

4.4.7. Statistics

Data were statistically analyzed through the SPSS statistics 17.0 software. A one-way

ANOVA and univariant linear model were used to compare means. The LSD post hoc

test was also used to determine the significant difference between groups. In all the

analysis p value < 0.05 were considered as statistically significant. Graphs with error bars

represent the standard deviation (SD).

4.5. Results

4.5.1. Morphological and phenotypic changes of monocyte-derived

microglia in culture

Adherent PBMC, representing mainly the monocytic population of blood leukocytes,

were cultured for up to 2 weeks in the presence M-CSF, GM-CSF, NGF-β and CCL2, in

RPMI 1640 medium without addition of serum to create optimally standardized

conditions. The cells were characterized for morphological changes over the period of 2

weeks in culture (Figure 4.1). The well established HMC3 human microglia cells line

was used as comparison. After 5 days in culture, the small (10–15 µm diameters) round-

Page 86: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[65]

shaped and semi-adhered monocytic cells expanded their cell body and developed few

unbranched processes (results not shown). During the second week, the cells reduced the

portion of the cell body and they increased the processes in length and number acquiring

ramified forms. M-MG obtained a ramified morphology after 10–14 days in culture,

including a polygonal or oval cell body and up to 7 primary processes and numerous

secondary process or spines. Over the 2 weeks in culture, the cells doubled in size and

increased in cell number indicating proliferation. After 2 weeks, M-MG displayed a very

similar morphology to the human microglia cell line HMC3 and to previously described

human brain-derived microglia in published studies (Leone et al., 2006).

Figure 4.1: Phase contrast microscopy of monocyte-derived microglia (M-MG) and human

microglia cell line HMC3

Monocyte derived microglia (M-MG) (A) and human microglia cell line HMC3 (B) Images were

taken from cells after 10 days in culture. Bar indicates 100 µM.

Page 87: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[66]

Basic known microglia markers, including expression of Iba-1 and CD68, were also

tested positive, supporting that the described protocol differentiates monocytes towards

M-MG (Figures 4.2 and 4.3).

Figure 4.2: Expression of Iba-1 in HMC3 and M-MG

Iba-1 is a characteristic microglia marker in HMC3(B) and M-MG (D). Corresponding negative

staining controls have been included (A and B). Bar indicates 30 µM.

Page 88: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[67]

Figure 4.3: Differential expression of CD68 in freshly isolated blood monocytes, HMC3 and M-MG.

Flow cytometry dot plots and histograms represent the differential expression of CD68 in freshly

isolated blood monocytes (A), HMC3 (B) and M-MG (C). The red dots/gray line indicates the

negative staining controls using a corresponding isotype control antibody. The black dots/line

indicates the specific test stain for CD68. Note that there is no CD68 expression in monocytes,

whereas two microglia populations clearly express CD68. Bar chart summarizes the mean

florescent intensity of studies groups (D). White bars are representing the isotope control, and

black representing the CD68 expression. M-MG and HMC3 showed significant differences in

expression of CD68 comparing to blood monocyte (P<0.05).

Page 89: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[68]

4.5.2. Expression pattern of specific surface markers

After culture in the presence or absence of the corresponding cytokines cocktails,

monocytes, M-MG, M-DC and the human microglia cell line (HMC3) were tested for

expression of surface markers relevant to human microglia. Figure 4.4 summarizes

expression of all relevant markers for all tested cell types, including CD45, CD14,

MHC class II, CCR2 and CCR4.

Firstly, the common leukocyte antigen CD45 was tested. CD45 is a characteristic

marker expressed by all leukocytes. As expected, CD45 was highly expressed in

monocytes, which did not change CD45 expression levels over the 2 weeks in culture in

serum-free medium without addition of cytokines. M-DC reduced only slightly CD45

expression over the 2 weeks in culture. However, M-MG reduced gradually CD45

expression levels over the 2 weeks in cul-ture reaching very low levels, as expected for

microglia and similar to HMC3 (Janabi et al., 1995; Li et al., 2009). CD14 is known to

be characteristic for monocytes, being expressed at highest level on these cells. As

predicted, M-MG reduced expression of CD14 to very low levels, similar to HMC3.

Activated microglia may process and present antigens on MHC class II. For that

purpose, activated microglia increase MHC classII expression, such as HLA-DR.

Usually, resting microglia express low levels of HLA-DR, whereas freshly isolated

monocytes express high levels of HLA-DR, which is also highly expressed in immature

M-DC. In our study, HLA-DR was clearly down-regulated in M-MG, reaching similar

levels detected in HMC3, indicating that M-MG may represent resting microglia cells.

Chemokines play a crucial role in recruitment and maturation of migratory, bone

marrow-derived microglia cells (Flynn et al., 2003). CCR1, CCR2, CCR5 expression

has been thus reported to be expressed at low levels in microglia (Bajetto et al., 2002;

Skuljecet al., 2011). In our study, monocytes expressed low levels of CCR2, but up-

regulation was seen when cultured in medium with-out cytokines. Interestingly, M-DC

decreased CCR2 expression to undetectable levels, whereas M-MG and HMC3

expressed clearly detectable CCR2 at rather low levels, as expected for microglia.

Expression of CCR4 and its ligand (CCL22 and CCL17) have been reported in the CNS

(Columba-Cabezas et al., 2002). Freshly isolated monocyte expressed CCR4 at very

low levels, while after one week in culture without cytokines, a sub-population (about

25%) expressed CCR4 at higher levels. However, M-DC and M-MG up-regulated

CCR4 expression to clearly detectable levels over time in culture to similar levels as

HMC3.

Page 90: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[69]

Figure 4.4: Expression of surface markers CD45 (a), CD14 (b), HLA-DR (c), CCR2 (d) and CCR4 (e) measured with flow cytometry.

Comparison of freshly isolated monocytes (A), cultured monocytes in the absence of cytokines

(B), monocyte derived dendritic cells (C), monocyte-derived microglia (D) and microglia cell line

HMC3 (E). The cells were cultured for 10 days.

Figure 4.5 summarizes the expression of various chemokine receptors on the different

cell populations that were investigated. Freshly isolated monocytes expressed

intermediate levels of CCR1. Differentiation towards M-MG decreased CCR1

expression to similar levels known for non-activated microglia and also seen in HMC3.

CCR5 expression level was detected at low levels in monocytes, but expressed at higher

levels in both M-MG and HMC3, as expected for microglia. CXCR1 was expressed at

very low levels in freshly isolated monocyte, but was clearly expressed at higher levels

Page 91: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[70]

in M-MG and HMC3. Freshly isolated monocyte did not express neither CXCR3 nor

CX3CR1. However, both these markers were clearly up-regulated in M-MG similar to

HMC3. CXCR3 expression in M-MG was at intermediate levels, while HMC3 showed

higher expression levels. Interestingly, almost similar expression of CX3CR1 was seen

in both M-MG and HMC3. In summary, M-MG and HMC3 expressed very similar

chemokine receptor patterns, supporting the microglia-like properties of M-MG.

Additional markers were also investigated (perosnal communiction), including CD 11c,

CD11b, CD40, CD80, CD83, CCR6 and CCR7. M-MG were negative for CCR6 and

CCR7. CD11c and CD11b were both expressed on M-MG and HMC3, but at much

lower levels than on M-DC. Immature M-DC expressed the dendritic cell markers

CD40 and CD83 at an intermediate intensity level. However, CD80 expression was

undetectable on M-DC. Most important, neither M-MG nor HMC3 expressed any of the

dendritic cell specific surface markers, such as CD83, CD80 or CD40, supporting that

M-MG are clearly different from dendritic cells.

Figure 4.5: Expression of chemokine receptors CCR1 (a), CCR5 (b), CXCR1(c), CXCR3 (d)

and CX3CR1 (e) measured with flow cytometry.

Expression levels in freshly isolated monocytes (A), monocyte-derived microglia (B) and the

microglia cell line HMC3 (C). The cells were cultured for 10 days.

Page 92: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[71]

4.5.3. Phagocytic capacity

Microglia play a major role in phagocytosis and clearance of dead cells and debris,

especially during inflammatory processes caused by injuries or neurodegenerative

diseases. To determine the phagocytic ability, we conducted phagocytosis experiments

(Meagher et al., 2005). M-MG were compared to immature M-DC, potent phagocytic

immune cells (Filgueira et al., 1996). HMC3 were used as microglia control cells.

Fluorescent-labeled S. aureus was used for measuring the phagocytic capacity of the

various cell populations.

Flow cytometry (Figure 4.6) and confocal microscopy (Figures 4.7 and 4.8) were used

for the detection and quantification of bacterial up-take. As expected, M-DC displayed

the highest phagocytosis, similar to HMC3, displayed even lower bacterial up-take.

However, both M-MG and HMC3 showed up-take of bacteria, with clear intracellular

location, as documented through confocal imaging. M-MG and HMC3 both showed

bimodal expression of labled bacteria (Figure 4.6) which may be related to the the sub-

population of cells with lower phagocytic activity.

Figure 4.6: Phagocytic capacity of M-DC (Black line), M-MG (Dotted line) and HMC3 (dashed line) towards TM Rhodamine-labelled S. aureus measured with flow cytometry.

Bacterial particles were added to culture medium at a ratio of 1:1000 (107

bacteria per ml) and

incubated for 4 hours at 37oC. Flow cytometry data represent the different levels of engulfment

fluorescent-labeled bacteria.

Page 93: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[72]

Figure 4.7: Up-take of TM Rhodamine-labeled S. aureus by M-MG using confocal microscopy.

M-MG were incubated for 4 hours with bacteria at a ration 1:1000. Note the intra-cellular

accumulation of red-labeled bacteria in the M-MG cells on the optical confocal section.

Figure 4.8: A selected Z-stack image taken with confocal microscopy showing the up-take of TM Rhodamine-labeled S. aureus by M-MG.

M-MG cells were incubated for 4 h with bacteria at a ration 1:1000. Note the intra-cellular

accumulation of red-labeled bacteria in the M-MG cells on the optical confocal section.

Page 94: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[73]

4.5.4. T-lymphocyte stimulatory capacity of MGM

As innate immune cells of the CNS, microglia are thought to be antigen presenting cells

(APC), able to process and present antigen to T-lymphocytes, activate them and induce

their proliferation. In this study, mixed leukocyte reactions (MLR) were used to

determine the T-lymphocytes stimulatory capacity of M-MG using proliferation assays.

Immature dendritic cells (M-DC) and the microglia cell line HMC3 were used as

comparison. A representative example was shown in Figure 4. 4. As expected, M-DC

induced the most pronounced and significant proliferation of allogeneic T-lymphocyte.

M-MG were also able to induce significant proliferation of allogeneic T-lymphocytes,

but to a much lower degree in comparison to M-DC. No significant T-lymphocyte

stimulatory effect was seen with HMC3, which is in accordance with previous studies

(Fischer and Reichmann, 2001, Janabi et al., 1995, Nagai et al., 2005, Zuiderwijk-Sick et

al., 2007).

Figure 4.9: Mixed leukocyte reactions (MLR)

Testing T-lymphocyte stimulatory capacity of HMC3 (black bars), M-MG (dotted bars) and M-DC

(hatched bars) at different cell ratios (antigen presenting cells: allogeneic T-lymphocytes). The

cells were cultured for 5 days before proliferation was measured using a BrdU incorporation assay.

Error bars represent the +/- 2 SD. Statistically significant differences between control and test

condition are indicated by *, P<0.05.

Page 95: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[74]

4.6. Discussion

Microglia play crucial roles in maintaining homeostasis and contributing to

neuroinflammation in response to any disturbances, such as injuries, toxins and pathogen

(Streit, 2001, Streit, 1996).To date, in vitro studies on microglia have used mainly

primary cells derived from embryonic or neonatal murine and human brains (Hinojosa et

al., 2011, Bassett et al., 2011). This study provides the first simple, standardized and

reliable protocol to generate human microglia-like cells from blood monocytes. The

protocol takes the advantage of a serum free culture condition, as well as a cocktail of 4

well defined recombinant human cytokines, i.e. M-CSF, GM-CSF, NGF and CCL-2. The

cells generated according to the described protocol display most known characteristics of

human microglia, including specific morphology, phenotype and functions.

In our model, the serum-free culture system is essential as it represents the physiological

condition of the extracellular fluid of the CNS, which is equal to the cerebrospinal fluid.

Normal cerebrospinal fluid (CSF) contains extremely low levels of proteins and bioactive

factors and is usually deficient in blood serum proteins (Reiber, 2003, Holman et al.,

2010). The blood-brain barrier prevents trafficking of blood serum proteins and other

factors into the extracellular space of the CNS, as they may damage glial and neuronal

cells through activation, resulting in apoptosis (Nadal et al., 1995, Abbott et al., 2010,

Hooper et al., 2009). For example, in vitro culture of rat microglia in presence of bovine

serum albumin, human or rat serum has been shown to enhance the production of

superoxide (O2−) as one of the mechanisms involved in pathogenesis of neuronal

damages (Elkabes et al., 1998). Knowing about the potential bio-active and even toxic

effects of blood serum, we established therefore this serum free protocol for the

generation of microglia-like cells derived from blood monocytes.

The cocktail of cytokines has been chosen according to the known effects of the

cytokines on microglia in published reports, as stated in the introduction section, and

extensive preliminary experiments. Thereby, the combination of M-CSF, GM-CSF, NGF

and CCL-2 has shown to differentiate M-MG with most similar characteristics to

microglia.

Under healthy condition, resting “surveying microglia” with ramified morphology is

responsible for maintaining the homeostatic condition of the CNS. Using the described

protocol, M-MG obtained a ramified morphology after 10-14 days in culture, including a

polygonal or oval cell body and up to seven primary processes and numerous fine

secondary process or fine spines. Our morphological findings for M-MG correspond to

Page 96: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[75]

the report by Leone et al. (2006), who used a less well-defined protocol. In addition, M-

MG displayed similar morphology to the human microglia cell line HMC3.

Most important was the phenotypic characterization of the cells using a broad panel of

antibodies and flow cytometry. Monocytes cultured over 2 weeks in the presence of the 4

cytokines changed significantly the expression of surface markers towards the known

pattern for microglia. CD45, CD14 and HLA-DR expression levels decreased

substantially in M-MG reaching levels expected for microglia. CD45 has been often used

for discriminating between tissue macrophages and microglia in CNS studies

(Lambertsen et al., 2009, Wirenfeldt et al., 2005). Penninger et al. (2001) states that in

the CNS, microglia express different levels of CD45 depending on their differentiation

state, with decreased expression levels in mature resting microglia. However, CD45

expression increases in activated microglia, e.g. in Alzheimer patients, as this surface

marker seems to play a role in degenerative and inflammatory CNS diseases (Masliah et

al., 1991). CD14 is also regarded as an important LPSreceptor (Triantafilou and

Triantafilou, 2002). Supporting our protocol, Lambert et al. (2008) reported down-

regulation of CD14 in monocyte-derived DC and microglia in presence of GM-CSF, M-

CSF and LPS.

The generation of T cell immunity requires the acquisition and presentation of Ag on

bone marrow-derived APCs. Both dendritic cells and macrophages with high expression

level of MHC class as well as co-stimulatory molecules, can stimulate naive CD8 T cells

in vivo to proliferate, develop effector function, and differentiate into memory cells

(Pozzi et al., 2005). Because of the low expression of MHC class II, B7 and CD40, the

essential molecules necessary for antigen presentation, microglia seem to be potential

APCs that can mount the immune response in the CNS. However, there is not much

known about antigen presenting ability of microglia as well as its role in physiological or

pathological immune responses in the CNS (Ulvestad et al., 1994). Compared with

resting microglia, microglia activation during CNS injury or infection could respond

more promptly to Th1-derived signals and behave as a more potent APC for T cell

restimulation (Aloisi et al., 2000, Streit et al., 1996). In line with previous data, our data

showed non-activated M-MG decreased HLA-DR expression to low levels, similar to

HMC3, but to much lower levels than freshly isolated monocytes or immature M-DC

(Lutz et al., 2000). Furthermore, it was important to discriminate between M-MG and M-

DC, as both may display similar morphological properties. In addition to the differences

in expression levels of MHC class II, CD11b and CD11c, M-MG did not express

characteristic M-DC markers, including CD40, CD80 and CD83. The differences

Page 97: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[76]

between M-MG and M-DC were further supported by the functional tests, such as MLR

and phagocytosis assays. As expected, M-MG displayed much lower T-lymphocyte

stimulatory capacity and bacterial up-take, in comparison with M-DC. In summary, our

data provide clear evidence that our M-MG protocol does not generate M-DC, but a

distinct microglia-like population.

Our study also revealed the expression level of various chemokine receptors, including

CCR1, CCR2, CCR4, CCR5, CXCR1, CXCR3 and CX3CR1 in M-MG and HMC3.

Freshly isolated monocytes showed intermediate expression level of CCR1, low

expression levels for CCR2, CXCR1 and CCR5, and undetectable expression levels of

CCR4, CXCR3 and CX3CR1, as reported previously by others (Geissmann et al., 2003).

M-MG changed expression levels of the various chemokine receptors substantially, in

comparison with freshly isolated monocytes. However, expression patterns of M-MG

were very similar to that of HMC3. M-MG showed down-regulation of CCR1 and CCR2

to still well detectable levels. On the other hand, M-MG displayed significant up-

regulation of CCR4 and CCR5. Interestingly, M-MG expressed CXCR3 and CX3CR1,

which were not detected in monocytes. Up-regulation of CXCR1 was also seen in M-

MG. Our findings are in accordance with the report by Bajetto et al. (2002), who

published that non-activated microglia express CCR1, CCR2 and CCR5 constitutively at

low levels. Several other researchers reported that activated microglia express higher

level of CCR2, CCR3, CCR4, CCR5, CX3CR1 (Simpson et al., 2000b, Eltayeb et al.,

2007, Szczuciński and Losy, 2007). However, little is known about chemokine receptor

expression in human microglia. Our protocol may help in future research to elucidate

regulation of these receptors and chemokine responses in human microglia.

4.7. Conclusion

We developed a new in vitro protocol for the generation of human microglia from blood

monocytes using a serum free culture condition and a novel mixture of four recombinant

human cytokines, i.e. M-CSF, GM-CSF, NGF and CCL2. Detailed characterization of

M-MG revealed a cell population representing resting microglia with their specific

morphological, phenotypic and functional properties. The described protocol is easy to

handle, well standardized and very reproducible, as it uses only well defined culture

medium and recombinant cytokines, but no serum or conditioned medium. This protocol

will certainly be very helpful for future studies investigating adult human microglia.

Page 98: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[77]

Chapter 5

Lead modulates chemokine receptors

expression pattern and induce

interleukin-8 in human microglia

Page 99: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[78]

5. Lead modulates chemokine receptors pattern and induce interleukin-8 in human microglia

5.1. Introduction to chapter

In the previous chapter, we showed differentiation and characterization of human

microglia derived from blood monocytic cells in the presence of a novel mixture of

cytokines. In this chapter, we investigated the effect of lead exposure on an in vitro

human monoycte-derived microglia model assessing morphology, phenotype and

function with the main focus on chemokine receptor expression pattern.

Lead is an environmental toxin with detrimental known effects on the functional and

development of CNS even at the low concentration. Despite the wealth of data on the

neurotoxic effect of lead in children, mechanism underlying its toxicity at the cellular and

molecular levels still need be studied. Microglia cells, the resident macrophages of the

brain, play important role in haemostasis of the CNS as neuroprotective cells (secretion

of anti-inflammatory cytokine, chemokine and their receptors, neurotrophic factors, etc).

Microglia activation which usually occurs in response to stimuli (toxin), has been shown

to be involved in many neuroinflammatory processes. Although recent animal studies

showed that lead may cause microglia activation and enhance hippocampal injuries

through changes in the secretion of pro-inflammatory cytokines, there is no evidence

showing the exact role of human microglia in lead induced toxicity or inflammation.

Therefore, we were interested to study whether exposure to low concentration of lead

over short period of time could affect the survival and function of the human microglia;

in particular, immune properties of these cells by alteration in chemokine receptors

expression pattern. Our results indicate that low level of lead exposure has no cytotoxic

effect on the morphology or viability of monocyte-derived microglia. General down

regulation in the expression of chemokine receptors expression was observed with

significant reduction in the expression of CCR1, CCR2, CCR3 and CXCR1, as well as

CD68. In addition, lead exposure induced increased secretion of interleukin-8 (IL-8), a

chemokine that is unique to humans and has recently been shown to contribute to brain

remodelling and memory loss. In summary, lead exposure can modulate

neuroinflammatory properties in human microglia. Declaration of work: All the

experiments were done by SE including cell culture, morphological studies, flow

cytometry, cytokine assay under supervision of Prof. Filgueira. A manuscript has been

prepared and submitted to Journal of Neurotoxicology (in revision at time of submission

of thesis).

Page 100: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[79]

5.2. Abstract

Lead (Pb) is a widely spread environmental pollutant with potentially detrimental effects

on major organ systems, including the central nervous system. Lead is therefore

considered a major health concern for humans. Several recent studies have confirmed the

neurotoxic effects of lead on development and function of the brain, even at low blood

concentrations (<10 µM). Thereby, lead has been shown to influence synaptogenesis and

neurogenesis, to negatively affect learning and memory, as well as to activate glia cells.

However, the molecular and cellular mechanisms involved in lead-induced neurotoxicity

in humans are still not well understood, especially for exposure to low lead

concentrations and for the contribution of microglia to lead toxicity. Microglia are the

resident immune cells of the CNS with a prominent role in physiology and pathology of

the brain. Over the past few years, environmental toxins, including lead, have been

reported to activate microglia in animal studies. Activated microglia have been shown to

contribute to neuroinflammation and neurodegenerative diseases. However, little is

known about the role of human microglia in lead induced neurotoxicity.

In this study, we investigated the effects of lead on human microglia in vitro at

concentrations, expected to represent in vivo concentrations found during lead

intoxication. For that purpose, a new human microglia model was applied. Human

microglia cells were exposed to 1-100 µM of lead acetate for 1 hour to 5 days.

Subsequently, changes in morphology, phenotype, cytokine secretion patterns and

function were investigated. Our results indicate that lead is taken up by microglia and

accumulates inside the cells, as visualized with specific fluorescent probes, using

fluorescence microscopy and flow cytometry. Lead exposure induced minor, but

significant morphological changes at higher concentrations (>50 µM), but had no

influence on morphology at lower concentrations (10 µM). Interestingly, lead was not

cytotoxic and did not influence microglia viability at lower concentrations. However,

lead had substantial effects on the expression patterns of surface markers that are

involved in inflammation, cell activation and migration, with significant reduction in

expression of CCR1, CCR2, CCR3 and CXCR1, as well as CD68. In addition, lead

exposure induced increased secretion of IL-8, a chemokine that is unique to humans and

that has recently been shown to contribute to brain remodeling and memory loss. In

summary, our results indicate that human microglia are significantly affected by lead

exposure, even at lower concentrations, resulting in discrete cell activation and

significant changes in chemokine receptor and surface marker expression, as well as

increased secretion of IL-8. We can therefore conclude that lead exposure, even at lower

Page 101: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[80]

concentrations, has a significant influence on microglia function, with possible

substantial consequences for homeostasis, tissue remodeling and immune responses in

the central nervous system.

Keywords: human microglia, in vitro model, M-MG, lead, IL-8, chemokine receptors

5.3. Introduction

Since humans started to process and use metals for their commodities, lead has widely

spread and become a persistent environmental pollutant. With that in mind, regulations of

how to deal with lead mining, processing and applications have reduced environmental

lead exposure in a number of countries, including Australia, Germany, Mexico, Sweden

and USA. Blood lead levels have declined on average below officially accepted toxic

concentrations in those populations (Canfield et al., 2003; Kasten-Jolly et al., 2011;

Koller et al., 2005; Sharma et al., 2011). However, lead exposure and corresponding

accumulation in people still happens at lower concentrations. Therefore, recent studies

have focused on mechanisms involved in low-level lead-induced toxicity (Bellinger,

2008; Dietert and Piepenbrink, 2006; Gidlow, 2004; Lanphear et al., 2000).

Lead is known to induce neurotoxicity even at low levels, resulting in irreversible,

detrimental effects on development, growth and cognitive functions (Finkelstein et al.,

1998). Lead enters the CNS by crossing the BBB. Foetuses and children are more

vulnerable than adults, as their BBB is not fully formed. In addition, their brain is more

susceptible to toxins, as it is still developing and growing. Lead seems though to interfere

with neuronal recruitment and differentiation (Baranowska-Bosiacka et al., 2013).

Several human and animal studies have reported the significant negative effects of lead,

even at lower blood levels (<2 µg/dl or <0.1 µM), on the cognitive function of

adolescents (Lucchini et al., 2012). Various published studies have reported that lead

exposure results in loss of hippocampal neurons, uncontrolled secretion of

neurotransmitters, alteration in neuronal growth rate, demyelination of nerve fibers,

proliferation and activation of astrocytes, as well as in breakdown of the BBB, causing a

deficit of learning and memory, and the decline of the IQ (Liu et al., 2012). It has been

shown that lead may substitute calcium and zinc resulting in its many toxic actions

(Marchetti, 2003). However, the molecular mechanisms in lead-induced neurotoxicity are

still not well understood.

Using animal models, lead exposure has been shown to potentially contribute to

pathogenesis of neurodegenerative diseases, such as Alzheimer’s disease, by inducing

glial cell activation and expression of the amyloid precursor protein (Monnet-Tschudi et

Page 102: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[81]

al., 2006). Lead has also been shown to modify cytokine gene expression patterns in the

murine CNS (Kasten-Jolly et al., 2011). In addition, lead-induced neurotoxicity has been

correlated with oxidative stress by generating reactive oxygen species (ROS), as well as

membrane lipid peroxidation (Adonaylo and Oteiza, 1999).

Interestingly, proliferation of T lymphocytes has also been observed in vitro after lead

exposure, indicating that lead has also an influence on immune responses (McCabe et al.,

2001). However, little is known about the effects of lead on human microglia.

Microglia are the innate resident immune cells of the CNS and play a crucial role in

maintaining the healthy and physiological homeostasis of the CNS. Resting microglia

with their branched processes form a dense cellular network, survey actively the nervous

parenchyma and respond rapidly to any disturbances, such as pathogens, toxins and

injuries, by retracting the processes and forming amoeboid activated microglia cells

(Block et al., 2007). Interestingly, the view of microglia function has been challenged

over the past decades. It has been recognized that activated microglia play an important

role in neuroinflammation and neurodegenerative diseases, even at early stages. Thereby,

microglia-derived soluble immune factors play a crucial role, including cytokines and

chemokines, as well as their receptors. Cytokines contribute to immune modulation in the

CNS, but they have also been reported to play an important role in neuronal development

and survival, by influencing neuronal activities, cell-cell interaction and neuronal-

endocrine communication (Boulanger, 2009; Deverman and Patterson, 2009). It has been

well documented that most CNS cells, including neurons, astrocytes and microglia, are

producer and targets of cytokines (Hopkins and Rothwell, 1995). In the healthy brain,

various cytokines, such as neurotrophins and pro- and anti- inflammatory factors, have

been shown to be constitutively expressed by various cell types (Elkabes et al., 1996).

Despite detection of cytokine mRNA under normal conditions, resting microglia have

been reported to secrete cytokine protein only at low or undetectable levels.

In turn, once microglia are activated, expression of various pro-inflammatory cytokines

are up-regulated, including Tumor Necrosis Factor (TNF)-α, Interleukin (IL)-1β, and IL-

6, contributing to neuroinflammation and neuronal cell death (Hanisch, 2002; Szelényi,

2001). In line with neurotoxic effects of activated microglia, several neurotrophic and

anti-inflammatory cytokines (IL-4, IL-10, TGF-β) have also been shown to be secreted

by those cells (Aloisi, 2001). Microglia, as well as astrocytes and neurons, also express a

large number of chemokines and chemokine receptors with potential effects on

development and homeostasis of the CNS, mainly through regulating cell migration (Cho

and Miller, 2002; Etemad et al., 2012; Flynn et al., 2003).

Page 103: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[82]

During microglia activation, up-regulation of chemokine receptors have been shown in

several neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease,

human immunodeficiency virus-associated dementia and multiple sclerosis (Wyss- Coray

and Mucke, 2002). Pro-inflammatory chemokines may also induce neuronal cell death

directly, or through microglia activation (Cartier et al., 2005).

To date, little is known about lead accumulation in microglia. However, astrocytes have

been shown to take up substantial amounts of lead, even more than neuronal cells, which

is enhanced in a neuron-glia co-culture models (Tiffany-Castiglioni, 1993; Tiffany-

Castiglioni and Qian, 2001). Recently, low–level, but long-term lead exposure has been

shown to activate rat microglia, resulting in memory deficiency (Liu et al., 2012). Lead is

well known for inducing deficits in spatial memory and learning (Carpenter et al., 1994).

However, the mechanisms for this phenomenon are still not well understood.

Interestingly, increased IL-8 secretion by microglia induces impairment of long-term

potentiation, responsible for memory deficiency (Xiong et al., 2003). Unfortunately, little

is known about IL-8 effects in the CNS, as this chemokine is unique to humans, but not

found in rodents (Edman et al., 2008). However, human peripheral blood mononuclear

cells (PBMC) have been shown to up-regulate expression of IL-8 when exposed to low-

level of lead (Gillis et al., 2012).

In this study, we investigated the effects of lead exposure on human microglia in vitro,

focusing on morphology, viability, chemokine receptors expression patterns and cytokine

secretion.

5.4. Material and methods

5.4.1. Cell culture

We used a novel human in vitro microglia model established by our group (Chapter 4).

Briefly, to generate human microglia, Human blood mononuclear cells (PBMC) were

isolated from buffy coats (100 buffy coats were used for this study) of healthy donors

(Australian Red Cross Blood Service (ARCBS), Perth, WA, Australia; ethics approval

was granted by UWA and ARCBS). The Monocytes (adherent PBMC) were seperated

from non-adherent cells (consisting mainly of T-lymphocytes) through plastic adherent

method (as mentioned in Chapter 4). Adherent PBMC (1×105

cells/ml) were cultured in

RPMI 1640 Glutamax medium and 1% antibacterial/antimycotic solution (Gibco),

supplemented with M-CSF (10 ng/ml; PeproTech, Rocky Hill, NJ), GM-CSF (10 ng/ml;

PeproTech), β-nerve growth factor (NGF-β; 10 ng/ml; PeproTech) and CCL2 (100

Page 104: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[83]

ng/ml; PeproTech), at standard humidified culture conditions (37oC, 5% CO2) for up to

14 days.

5.4.2. Lead exposure

Lead acetate [Pb(CH3CO2)2·3H2O] (Sigma-Aldrich, St. Louis, MO) was prepared at the

concentration of 100 mM as stock solution under sterile conditions. In order to study the

effect of lead on human microglia, lead was directly added at the concentration of 1, 5,

10, 25, 50 and 100 µM to the cells (depending on the experimental design) at the culture

days, 7-10 for different time period, including 1 hour, 24 hours, 3 and 5 days. To ensure

that lead effects were not caused by possible, even low endotoxin contamination, LPS(10

ng/ml, E. coli serotype 0111:B4, Sigma -Aldrich).were included in positive control

experiments, clearly showing completely different results.

5.4.3. Morphological studies

Microglia were cultured in the presence or absence of 10 µM lead for 24 hours.

Morphological appearance and changes of cell shape were documented by using an

inverted Leica DM IL microscope, equipped with phase contrast and various adequate

fluorescence filters. Images were taken with a Photometrix Cool SNAP Fx digital camera

and processed with the NIS- elements F3.0 software. Sytox green, a nucleic acid stain

(Invitrogen) was applied to exclude the dead cells in morphological quantification (Luo

et al., 2001). Cells were incubated with 1 µM Sytox Green for 30 min at 37 oC. Five

random optical fields were selected. Images were taken with fluorescent filter and bright

field microscopy. After overlaying the images, a total of 100 live cells were counted. In

repeated independent experiments, we also quantified the cell numbers with branching

changes by staining microglia with an antibody against Iba-1, a macrophage/microglia

marker, using a standard immunostaining protocol (Blomster et al., 2011).

Immunostaining was performed with microglia cultured on cover slips, fixed with 1%

paraformaldehyde in PBS, containing 2 % sucrose. After permeabilization with 0.01%

Triton X100 in PBS, cells were incubated overnight at 4°C with rabbit anti- Iba-

1(1:100,Wako Pure Chemical Industries), with subsequent incubation for 1 hour with

Alexa Fluor 488-conjugated donkey anti-rabbit antibody (1:200, Invitrogen) and DAPI (1

µg/ml, Roche, Manheim, Germany). Slides were analyzed using fluorescent microscopy

as described above and images were taken. According to previous studies, microglia with

less than two branches were scored as activated (Kauppinen et al., 2008).

Page 105: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[84]

5.4.4. Viability assays

To assess the effects of lead on viability of human microglia, MTS assays were carried

out (Cell Titer 96 AQueous Assay, Promega, Madison, WI). Briefly, cells were seeded in

96 well plates (5x105 cell/ml, 200 µl per well) and cultured to reach optimal confluence.

Then cells were incubated with various lead concentrations (0, 1, 10 and 50 µM,

quadruplets for same condition) for 24 hours. Thereafter, 20 µl of Cell Titer 96®

AQueous One Solution (MTS) was added directly to each well. Cells were incubated for

4 hours at 37oC and the absorbance of the colorimetric reaction was measured by a plate

reader (Labsystem Multiscan RC) at 490 nm. Viability of lead-treated microglia was

compared with the non-treated control and statistically analyzed using the Student’s t-test

and ANOVA (Vairano et al., 2004).

An additional method, namely the propidium iodide dead cell exclusion test, was also

used for testing microglia viability exposed to lead (Kauppinen et al., 2008). For that

purpose, cells were treated with different concentration of lead (as mentioned above) for

24 hours. Then, 2 µg/ml propidium iodide was added to the culture medium. Morphology

and viability of the cells were quantified using fluorescent microscopy. For

quantification, five randomly selected fields were chosen in each well and at least 100

cells were tabulated. Dead cells were labeled with propidium iodide and seen in orange

color under the fluorescent microscope. In addition, the samples were also assessed with

FACSCallibure (BD Biosciences, Mountain View, CA). Data analysis was performed

using FlowJo software.

5.4.5. Detection of up-take and intracellular accumulation of lead

We investigated the up-take and intracellular accumulation of lead by microglia with the

specific lead detection fluorescent probe Leadmium Green AM (Invitrogen) using flow

cytometry. In addition to Leadmium, we studied the expressdion level of DCF Newport

Green (Molecular Probes, Eugene, OR) as less specific lead detector. Human microglia

were incubated overnight with different concentrations of lead (5, 10, 25, 50 and 100

µM). Then, the cells were incubated with 1 µM Leadmium Green AM dye in saline for

30 minutes to 2 hours. To test the DCF Newport Green, in separate experiment, cell were

also cultured with 1 µM DCF Newport Green dye in saline for 30 minutes to 2 hours.

Cells were analyzed by flow cytometry using the 488nm excitation and measuring

fluorescent emission at 520nm. Non-lead treated cells were included in the experiment as

negative controls (Zeller et al., 2010). FlowJo software was used for further analysis of

data. Representative histograms for each experimental group were overlayed and mean

Page 106: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[85]

fluorescent intensity (MFI) were compared statistically using the Student’s t-test.

Experiments with cells from different donors were repeated five times. Another

fluorescent probe, which is less specific, Newport Green DCF (Invitrogen), was also used

to detect lead up-take by human microglia.

5.4.6. Reactive oxygen species

We studied the effect of lead on ROS production by human microglia using the

fluorescent dyes Dihydroethidium (DHE) to detect superoxide and BODIPY® 581⁄591

C11 to detect lipid peroxidation. Cells were incubated with different concentrations of

lead, including 0, 5, 10, 25 and 50 µM for 24 hours. Five separate experiments with cells

from different blood donors were performed. Subsequently, each sample was separately

incubated with DHE (1 µM) or BODIPY (2 µM) for 90 minutes. Cells also were cultured

with LPS (10 ng/ml, E.coli, Sigma-Aldrich) for 24 hours as positive control. Mean

fluorescence intensity of the samples was measured using the FACS Canto flow

cytometer (BD, Bioscience). After analyzing the flow cytometry data with FlowJo

software, they were statistically compared to controls using Student’s t-test, whereby

mean fluorescence intensity (MFI) of each group were calculated and compared for DHE

or Bodipy (Kauppinen et al., 2008; Yang et al., 2007).

5.4.7. Expression of surface markers

We studied the effect of lead (10 µM) on the expression patterns of specific surface

molecules in human microglia using flow cytometry. Cells were divided into two groups,

non-treated and lead-treated, which were exposed to lead for 24 hours. After detaching

and washing twice with PBS, the cells were stained with direct labeled antibodies, or

using primary unlabeled and corresponding labeled secondary antibodies.

A panel of directly labeled mouse anti-human antibodies was applied at the supplier’s

recommended concentration. The antibodies were directed against the following antigens

and containing corresponding label: CD45-FITC, CD14-PE, CD40-PE, CD80-PE, CD68-

AF647, HLADR–PERCP, CD11b-AF488, CD11c-PE, CCR1-AF647, CCR2-AF647,

CCR3-PE, CCR4-PE-Cy7, CCR5-FITC, CCR6-AF647, CCR7-PE, CCR9-fitc, CXCR1-

PE, CXCR3-AF488, CXCR4-PE and CXCR5-FITC (all from BD Bioscience).

Corresponding labeled isotype control antibodies were included in all experiments, as

well as non-treated cells. A rabbit anti-CX3CR1 polyclonal antibody was used at the

final concentration of 1 µg/ml (AbD Serotec, Oxford, UK), followed by a secondary

donkey anti-rabbit-AF 647 used at the final concentration of 1:200 (Invitrogen). Cells,

Page 107: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[86]

which were not labeled with the primary antibody, were used as negative control.

Corresponding excitation and emission wavelengths of the used fluorochrome were

FITC: 494, 519; PE: 496/546,578; PerCP: 482, 678; APC: 650, 660; Alexa Fluor -488:

495, 519; Alexa Fluor -647: 650, 668. The cells were incubated with antibodies for 30

minutes, washed with PBS, resuspended and fixed with 1% formaldehyde in PBS,

containing 2% sucrose. The fixed samples were measured with a FACSCallibure (BD

Biosciences). Data analysis was performed using FlowJo software.

5.4.8. Cytokine production

The cytometric bead array (CBA) human inflammatory kit (BD Biosciences) was used

for the quantitative measurement of cytokine production and secretion under control and

lead-exposed conditions. For that purpose, human microglia were incubated with and

without lead at various concentrations for 24 hours, as explained above. The following

day, 50-100 µl of the medium was collected and stored at -20oC. The assay was

performed according to the manufacturer’s instruction using 50 µl of the human

inflammatory cytokine standards and sample dilution. Cytokine concentrations were

measured using the FACSCanto flow cytometer (BD Bioscience). FCAP Array, version

3, software was used to analyze the data. Measurements were done in duplicates and

repeated once with 5 independent experimental repeats.

5.4.9. Statistics

Data were statistically analyzed with the SPSS statistical software (version 17.0). A one-

way ANOVA and univariant linear model were used to compare means. The

LSD/Dunnett post-hoc test also used to determine the significant difference between

groups. In all the analysis p value<0.05 were considered as statistically significant. Bars

in graphs represent the standard error (SED).

5.5. Results

5.5.1. Detection of lead uptake and intracellular accumulation by

human microglia

To study lead uptake and intracellular accumulation in human microglia, we used the

fluorescent probe Leadmium Green and flow cytometry. Leadmium Green has been

reported to be a very sensitive and specific probe for lead detection at nM levels. The

cells were incubated with lead at 5, 10, 25, 50 and 100 µM for 24 hours. Then, cells were

incubated with 1 µM Leadmium Greenand 1 µM DCF Newwport Green dyes for 30

Page 108: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[87]

minutes and mean fluorescence intensity (MFI) was calculated using flow cytometry.

Non- lead treated cells were included in each experiment as negative control. Lead

uptake was detectable as green fluorescence signal in the microglia cells from 5 µ

concentrations onwards in a concentration-dependentmanner (Figure 5.1 C). Mean of

fluorescence intensity ± SD of the Leadmium for the M-MG cultured with different

concentration of lead, 0 to 100 µM respectively were 3.2 ± 0.07,5.88 ± 0.19, 8.51 ± 0.32,

10.36 ± 0.05, 14.64 ± 2.3 and 30.38 ± 1.56. Similar results but at lesser extent were

obtained when using a different, less specific fluorescent probe, Newport Green DCF

(Figure 5.1 D). These data clearly indicate that human microglia are able to take up and

accumulate lead intracellularly.Mean of fluorescence intensity ± SD of the Newport

Green for the M-MG cultured with different concentration of lead, 0 to 100 µM

respectively were 4.26 ± 0.8, 4.68 ±0.45, 5.17 ±0.42, 7.34 ±1.4, 10.36 ±0.5 and 15.7

±0.87.

Figure 5.1: Lead uptake by human microglia.

These figures show uptake and accumulation of lead in human M-MG using the Leadmium and

DCF Newport Green using flow cytometry (n=5). Figures A and B are the representative FSC:SSC

dot plots of M-MG in absent/presence of 10 µM lead from one representative experiment out of

five with similar results. Histograms compare the relative expression of Leadmium (C) and DCF

Newport Green (D) to the corresponding amount of intracellular lead. the absence (a) or presence

of various concentration of lead, including 5 µM (b), 10 µM (c), 25 µM (d), 50 µM (e) and 100 µM

(f).

Page 109: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[88]

5.5.2. Effect of various lead concentration on human microglia cell

morphology and activation

Microglia react to toxic effects with quick morphological changes and activation,

resulting in a less branched and amoeboid appearance. Accordingly, we studied the effect

of lead on morphological changes in human microglia using live cell imaging. As

mentioned before, five random optical fields were selected. One hundred live cells from

each experiment were quantified and scored as activated, when displaying less than two

branches. In separate experiments, cells were stained with Iba-1 as microglia marker and

number of green fluorescent labeled branches was counted. Under control conditions (no

lead exposure) and at exposure of up to 10 µM lead, microglia showed a resting (non-

activated) morphology with usually more than four branched processes (Figure 5.2. A).

However, morphological changes were clearly seen from 50 µM lead concentration

onwards, causing the cells to retract their processes and to become more round shaped,

which suggests changes towards the activated form (Figure 5.2. B, C). Quantification

showed that more than 75% of the cells displayed ramified morphology in the control

group (0 µM lead). Only at lead concentrations above 25 M, cells showed significant

morphological changes compared to the control group (Figure 5.2. D). No significant

changes in morphology (size of the cell body, number of branches) were detected after

administration of the lower lead concentrations (<10 µM) for 24 hours. The

morphological changes documented with microscopy were confirmed and quantified

using the more sensitive method of fluorescent microscopy (data not shown; Kauppinen

et al., 2008; Minami et al., 2012).

Page 110: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[89]

Figure 5.2: Effect of lead exposure on morphology of microglia.

Morphological appearance of human microglia exposed to various concentrations of lead 0 µM

(A), 10 µM (B), 50 µM (C) for 24 hours as documented with phase contrast light microscopy

(representative of 5 experiments, bar indicates 100 µm). Quantification of branched and round

shaped morphology is shown in the bar chart in D as percentage of total live cells. Cells were

scored as activated/round shaped when displaying less than two branches. * indicates significance

at P<0.05, n=5. Error bars indicate SEM.

Page 111: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[90]

5.5.3. Effect of various concentration of lead on human microglia

viability

We investigated whether lead affected the viability of microglia cells. For that purpose,

human microglia were treated with various concentrations of lead (1, 10 and 50 µM) for

24 and 72 hours. Then, we assessed the viability of the cells using the MTS test (Figure

5.3. A). Viability of the non-treated cells (control group) was considered as 100%

(baseline) and the experimental groups were correspondingly compared to these controls.

Results showed that lead had no effect on viability at the concentration of 1 and 10 µM.

To assess microglia survival rate, we also calculated the ratio of dead to live microglia

using fluorescence microscopy, after exposure to 5, 10, 25, 50 µM lead for 24 hours.

Statistical analysis showed no significant differences between the lead-treated

experimental groups and the control cells at lower lead concentrations. Moderate increase

in number of dead cells was only seen at the concentration of 50 M, which was not

statistically significant. Overall, these data indicate that low lead concentrations (<10

µM) have no effect on the viability of human microglia.

Figure 5.3: Viability of human microglia after lead exposure.

A) Bar charts represent the percentages of live cells after exposure to various lead concentrations

(0, 1, 10 and 50 µM) for 24 hours (black bars) and 72 hours (white bars). Viability of the cells was

assessed by a MTS colorimetric test. Viability of negative control group (non-lead treated) was

used as baseline and normalized to 100%.Difference between two groups (lead-treated/non-treated

cell) were statistically analyzed using the Student’s t-test and Dunnett’s test (n=20, * indicates

significance with P<0.05). B) Stacked columns represent the fraction of dead (black) to live

(Green) microglia in the presence of various concentration of lead (0, 5, 10, 25 and 50 µM) for 24

hours. Dead cells were labeled with 2 µg/ml propidium iodide and counted using fluorescent

microscopy. Ratio of the dead to live cells is shown as percentage. Difference between two groups

(dead and live cells) were statistically analyzed using the Student’s t-test and Dunnett’s test (n=20,

P<0.05).

Page 112: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[91]

5.5.4. Effects of lead exposure on reactive oxygen species production

and lipid peroxidation in human microglia

Many studies have shown that activated microglia release reactive oxygen species (ROS)

in response to environmental stimuli, such as toxins. To study whether lead exposure

induced ROS production in human microglia, we used the fluorescent probes

Dihydroethidium (DHE) as superoxide detector and BODIPY® 581⁄591 C11 to measure

lipid peroxidation. Lead treated and untreated cells were stained with these fluorescent

probes. Propidium iodide and SYTO green labelling was included to determine live and

dead cells. Fluorescent intensity was measured using flow cytometry.Cells also cultured

overnight with LPS (10 ng/ml) as positive control. Only live cells were included in the

analysis. Figure 5.4. A. and B.shows the location of cells in FSC:SSC axis in absent and

presnt of lead treatmnet (10 μM lead). Figure 5.4. C is representative histograms from

one experiment out five experiments.. Overall, microglia treated with 5 µM lead showed

moderate increase of ROS production followed by a decrease of ROS towards 50 µM

lead exposure. Mean of fluorescence intensity ± SD of the DHE for the M-MG cultured

with different concentration of lead, 0 to 50 µM as well as LPS respectively were 151.22

±9.76, 176.6 ± 7.76, 159.44 ± 8.87, 144 ± 14.57, 151.2 ± 9.88 and 356.8 ± 52.2. The

mean ± SD of the isotype control was also 3.52 ± 0.67. Similarly, no effect of lead on

lipid peroxidation in the human microglia was seen at the tested lead concentrations

(Figure 5.4 D). One can conclude that low lead concentrations have no significant effect

on ROS production and lipid peroxidation in human microglia. Mean of fluorescence

intensity ± SD of the BODIPY for the M-MG cultured with different concentration of

lead, 0 to 50 µM as well as LPS respectively were 4.44 ± 0.29, 5.06 ± 1.06, 4.124 ± 0.85,

4.6 ± 0.77, 5.492 ± 0.87 and 163.6 ± 15.8. The mean ± SD of the isotype control was also

3.24 ± 0.82.

5.5.5. Effect of lead exposure on expression of chemokine receptors and

surface molecules in human microglia

Chemokines and chemokine receptors are considered to play a key role in the initiation

of inflammation and cell migration in the context of CNS development and remodelling.

As reported previously, most of the chemokine receptors are expressed by microglia, due

to their involvement in immune responses (Chapter 4; Flynn et al., 2003). We were

interested in investigating whether lead may affect the pattern of chemokine receptors

expression in human microglia. After 7-10 days in culture, monocyte-derived microglia

were exposed to 10 µM lead for 24 hours and compared with non-treated control cells.

Page 113: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[92]

Figure 5.5 shows representative histograms for the expression pattern of chemokine

receptors in lead treated cells as compared with control cells, including CCR1, CCR2,

CCR3, CCR4, CCR5, CCR9, CXCR1, CXCR3, CXCR4 and CXCR5. Figures 5.6. A and

5.6. B summarize the expression pattern of all relevant markers tested in all experiments

(n=30). Overall, the presence of lead changed expression of most chemokine receptors.

However, only CCR1, CCR2, CCR3 and CXCR1 expression was found to be

significantly decreased in comparison with the non-treated microglia. Down-regulation

was also found for CCR5, CCR6, CXCR4 and CXCR5, but which was not statistically

significant (P 0.86, 0.45, 0.45 and 0.67). However, CCR4, CCR9 and CXCR3 were up-

regulated, although this was not statistically significant. The cells expressed CCR7 at

very low level and no changes were seen after lead exposure.

Figure 5.4: Effect of lead on ROS production and membrane peroxidation in human microglia.

Cells were incubated for 24 with various lead concentration (0, 5, 10, 25 and 50 µM) before they

were incubated for 30 minutes with DHE (1 µM) for ROS detection, or BODIPY (2 µM) for

detection of membrane peroxidation. LPS (10 ng/ml) were used as positive control. Fluorescence

intensity was quantified with flow cytometry. A) Filled histograms represent DHE treated cells,

whereas the open curve to the left represents cells not labeled with DHE. B) Summary of changes

as mean fluorescent intensity of the DHE and BODIPY (n=5). Mean ± SD of the MFI were

calculated for all samples and statistically analyzed using the Student’s t-test.

.

Page 114: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[93]

Figure 5.5: Effect of lead exposure (10 µM, 24 hours) on chemokine receptors and surface marker expression patterns in human microglia as measured with flow cytometry.

Examples of one representative experiment. Black histograms for lead-untreated control cells and

white histograms for lead treaded cells. Blue histograms are the isotype controls. Cells were

cultured for 7-10 days (n=30). Shifting black comparing to white histogram to the right is

representing the increase in expression level of marker in presence of lead and vice versa. Double

picks in CCR5 and CXCR1 are showing sub-populations in M_MG express different levels of

these markers.

Page 115: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[94]

Figure 5.6: Comparison between expression pattern of surface markers in the absence and presence of lead in human microglia

Comparison between control cells (white bars) and lead-treated cells for the expression of various

surface markers. B) Comparison between control cells (white bars) and lead-treated cells 10 µM

lead, 24 hour, for expression of various chemokine receptors. Data represent the mean florescent

intensity. Analysis of the significant differences between two groups was done by ANOVA and

LSD/Dennett’s test. (* indicates, n=30).

A

B

A

Page 116: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[95]

5.5.6. Effect of lead on cytokine secretion patterns in human microglia

Microglia activation has always been associated with the expression of cytokines, which

contribute to neurotrophic and neuroinflammatory events. Cytokines may also contribute

to the modulation of microglia activation (Vila et al., 2001).We used a cytometric bead

array for human inflammatory cytokines to determine whether lead exposure induced

changes in inflammatory cytokine expression patterns in human microglia. Cells were

cultured in the presence of different concentration of lead (0, 10, 50, 100 µM) for 24

hours. Culture medium supernatants were collected and used for the measurement of a

panel of inflammatory cytokines, including IL-1β, IL-8, IL-6, IL-12p70, TNF- and IFN-

(Figure 5.7). Of the six evaluated cytokines, only IL-8 was found to increase two folds

which was statistically significant (P0.036), when human microglia were exposed to

10µM lead, namely from 4730 pg/ml (lead-untreated control cells) to 8986 pg/ml (10 µM

lead). Furthermore, highest lead exposure to 100 M not only induced significant

increase in IL-8 expression (37569 pg/ml, P=0.000) but also significant enhancement

was observed in expression of IL-6, TNF- and IL-1β respectively (406, 846 and 1099

pg/ml, P<0.05). Slight increase in expression of IL-6 was also seen following exposure to

10 µM lead, however this increase was not statistically significant.Expression of all other

cytokines was at low detectable levels and was not influenced by lead. It is noteworthy

that production of all tested cytokines was significantly stimulated by LPS (10 ng/ml),

which was used as a positive control.

Page 117: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[96]

Figure 5.7: Effect of lead on cytokine secretion in human microglia.

Cells were challenged with lead (0, 10, 50, 100 µM) or LPS 10 ng/ml for 24 hours. Negative

control, no lead or LPS treatment was also included in experiment. Various cytokines were

measured in the culture supernatants using a cytometric bead assay and flow cytometry. Data were

analyzed by FCAP software. Final concentration of cytokines are shown as pg/ml (n= 15, *

indicates P<0.05).

0.00

500.00

1000.00

1500.00

2000.00

2500.00

3000.00

3500.00

4000.00

4500.00

No Pb Pb 10 µM

Pb 50 µM

Pb 100 µM

LPS

IL-1β

(p

g/m

l)

* p=0.000

* p=0.000

0.00

500.00

1000.00

1500.00

2000.00

2500.00

3000.00

3500.00

4000.00

4500.00

No Pb Pb 10 µM

Pb 50 µM

Pb 100 µM

LPS

1L-

10

(p

g/m

l)

* p=0.000

0.00

500.00

1000.00

1500.00

2000.00

2500.00

3000.00

3500.00

4000.00

4500.00

No Pb Pb 10 µM

Pb 50 µM

Pb 100 µM

LPS

TNF-

(p

g/m

l)

* p=0.000

* p=0.000

* p=0.048

0.00

5000.00

10000.00

15000.00

20000.00

25000.00

30000.00

35000.00

40000.00

45000.00

No Pb Pb 10 µM

Pb 50 µM

Pb 100 µM

LPS

IL-8

(p

g/m

l)

*p=0.036

* p=0.000

* p=0.000

0.00

500.00

1000.00

1500.00

2000.00

2500.00

3000.00

3500.00

4000.00

4500.00

No Pb Pb 10 µM

Pb 50 µM

Pb 100 µM

LPS

IL-6

(p

g/m

l)

* p=0.020

* p=0.000

Page 118: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[97]

5.6. Discussion

Lead has been used widely for many decades for the production of various commodities

(paint, gasoline, utensil, electronics, etc.) and it will continue to be used in the future for

many kinds of technological purposes, due to its unique properties (Luo et al., 2012).

Therefore, lead exposure and toxicity is still a major global health problem, especially in

lead mining and processing countries, where workers and corresponding local residents

are affected. The CNS is one of the main targets of lead toxicity. During development

and growth, the brain is most susceptible to toxic effects of lead, which correlates with

deficiency in neurogenesis and remodeling of the brain. These pathological changes in

the brain result in neurocognitive and functional deficits that are induced by lead even at

low blood concentration (< 10 µM) (Liu et al., 2012). Immune and inflammatory effects

of lead on the nervous system have also been reported (Waterman et al., 1994). In that

respect, one has to consider microglia as an important cell population, representing the

innate immune system in the CNS. Microglia are the resident macrophages of the CNS.

They act as the first line of defense in the brain and play a crucial role in health and

disease, including infections, trauma and neurodegenerative diseases. Activated

microglia can either induce or inhibit neurogenesis, depending on the condition and local

tissue response. Neurogenesis is of course important in the developing and growing CNS,

but adult neurogenesis contributes also to some important areas and functions in the

adult, such as the olfactory system, the forebrain and memory function in the

hippocampus (Lazarini et al., 2012; Morrens et al., 2012). Interestingly, using modern

imaging technologies, structural changes of the brain could be visualized in long-term

lead exposed workers, which could also be correlated to functional deficits (Bleecker et

al., 2007; Jiang et al., 2008). However, little is known about the molecular and cellular

mechanisms related to lead neurotoxicity.

In this study, we investigated the effects of low level lead exposure on human microglia,

using a novel human in vitro model (Chapter 4). We showed that at low concentration

(<10 µM), lead has only minor effect on morphology and viability of microglia.

However, functional and phenotypic tests indicate that lead induces specific changes in

chemokine receptors expression and secretion of IL-8.

Lead has a special and detrimental effect on the CNS during development and growth,

where cell proliferation, maturation and migration play a crucial role. Various studies

reported the inhibitory effect of lead on proliferation of the oligodendroglia and neuronal

Page 119: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[98]

precursors. Most interestingly, an in vitro study showed that low level exposure of lead

(0.01-10 µM) has no effects on rat neuronal stem cells viability, but results in a

concentration-dependentdecrease of proliferation of the cells. Reduction in neurons and

oligodendrocytes numbers, and a significant increase in astrocyte numbers has also been

reported following lead exposure (Huang and Schneider, 2004). However, there is no

clear evidence on apoptotic effects and decreased viability of astrocytes and microglia

after lead exposure. In contrast, astrocytes have been reported to be resistant to cell death

and morphologic changes following lead exposure (Opanashuk and Finkelstein, 1995).

Nevertheless, activation of astrocytes and microglia has been found (Garber and Heiman,

2002). These published reports are in concordance with our findings. Our results indicate

that lead, up to concentrations of 50µM, does not affect the survival of the human

microglia. In this context, it is worth mentioning that many lead neurotoxicity studies

have used very high lead concentrations in the range of 100µM and above, showing

substantial cytotoxicity. However, it is unlikely that such high lead concentrations are

reached in vivo. Therefore, we suggest that lead neurotoxicity is rather due to changes in

cell recruitment, turnover and function, and not due to increased cell death.

It has been well documented that resting microglia with ramified morphology are

activated by various stimuli, including toxins. Thereby, the cells change their

morphology, by shortening and losing branches, and by hypertrophy of the cell body,

resulting in a more mobile and migratory, “ameboid” microglia (Nakajima and Kohsaka,

2001; Liu et al., 2012). Loss of microglia ramification in the presence of LPS has been

published (Nakamura et al., 1999). Other factors and conditions have also been shown to

activate microglia, including exposure to interferon gamma (Kloss et al., 2001) and

astrocytes-conditioned medium (Bohatschek et al., 2001). Changes in morphology of the

macrophages in other organs have also been reported in response to lead intoxication

(Sengupta and Bishayi, 2002). However, our study indicate that low level lead exposure

(up to 10 µM) does not significantly change microglia morphology, and therefore does

not significantly activate the cells. Only exposure to higher lead concentrations (50 µM

and above) resulted in a significant morphological change, indicating substantial

microglia activation. The mechanisms of this transformation are still unknown. Elevation

of Ca2+

and cAMP, or inhibition of G-proteins and phosphatases have been postulated as

the possible mechanisms (Kalla et al., 2003). However, more research is required to

elaborate a better understanding of microglia activation in the context of lead

neurotoxicity.Oxidative stress has been postulated to play an important role in neuronal

cell death and pathogenesis of neurodegenerative diseases (Halliwell, 1992; Thomas et

al., 2007). Oxidative stress may induce cell death through increased production of ROS

Page 120: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[99]

and enhanced lipid peroxidation, resulting in substantial functional changes of cell

membranes and membranes of the organelles (Moreira et al., 2005). Activated microglia

has been shown to be an important source of ROS and have therefore been implicated in

the pathology of neurodegenerative diseases (Schilling and Eder, 2011). In addition,

mitochondrial alteration and ROS production has been correlated with microglia

activation (Verri et al., 2012). Disturbance in the pro-oxidative and anti-oxidative

balance in the brain has been considered as one of the possible mechanisms involved in

lead induced neurotoxicity. Previous studies have thus reported the enhanced regional

lipid peroxidation in the rat brain, especially the hippocampus, after low levels of lead

exposure (Villeda- Hernández et al., 2001). However, most studies have used rodent

models (Adonaylo and Oteiza, 1999, Qin et al., 2005, ), which are known to produce high

amounts of ROS, whereas humans produce much lower amounts, if at all. With this in

mind, the results of our study are reasonable, as they indicate that lead exposure does not

induce increased production of ROS and subsequent lipid peroxidation in human

microglia. The difference in ROS production between humans and rodents may thus be

responsible for discrepancies in the literature.

Cytokines and chemokines form an important part of an immune responses, regulating

the auto- and paracrine communications with other cells in various tissues, including the

CNS, where these factors support immune defense, repair and recovery processes. It has

been postulated that there is a constitutive base-level production of cytokines and

neurotrophic factors in the healthy CNS, supporting homeostasis and remodeling.

However, there has been more emphasis on the role of cytokines in causing damage in

the context of inflammation and degenerative diseases. Several studies have reported a

key role for pro-inflammatory cytokines, such as IL-1β, IL-6, tumor necrosis factor-

and transforming growth factor-β (TGF-β), in the pathogenesis of stroke, Parkinson's and

Alzheimer's disease (Mathias et al., 2004). Importantly, microglia have been shown to be

an important source of cytokines and chemokines in the CNS, as well as playing a role in

inflammation and degenerative diseases (Hanisch, 2002).Therefore, we investigated

whether lead exposure induced production of inflammatory cytokines by microglia.

However, no significant secretion of those cytokines was found, including IL-6, IL-10,

TNF-, IFN- and IL-12, independent of lead concentration. This indicates that lead-

exposed human microglia may not react with increased production of inflammatory

cytokines following lead exposure. However, lead induced increased secretion of IL-8,

an inflammatory chemokine. Chemokines are chemoattractant factors controlling

migration of cells of the immune systems. However, chemokines play also an important

role in the developing brain, controlling migration and differentiation of neural cells. In

Page 121: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[100]

addition, chemokines also contribute probably to cell replacement in the adult brain in

areas where tissue remodeling takes place. Additionally, chemokines contribute to cell

activation, control of metabolic functions and cell survival, as well as induction of

programmed cell death (Bacon and Harrison, 2000). In the CNS, various chemokines and

their receptors have been shown to play a role, including CCL2 (MCP-1)/CCR2, CCL3

(MIP-1α)/CCR1/CCR5, CCL4 (MIP-1β)/CCR5, CCL5 (RANTES)/ CCR1/CCR3/CCR5,

CXCL-1 (GRO-α)/CXCR2, CXCL8 (IL-8)/CXCR1, CXCL10 (IP-10)/CXCR3, CXCL12

(SDF-1)/CXCR4 and CX3CL1 (fractalkine)/CX3CR1 (Cartier et al., 2005; de Haas et al.,

2007). Among those chemokines, only CCL2, SDF-1 and fractalkine seem to be

constitutively expressed by neurons and astrocytes. However, expression of other

chemokines has also been reported in response to inflammation. IL-8 is one such

inflammatory chemokine, found only in humans, but not in rodents. IL-8 is produced by

astrocytes and microglia in the context of inflammation (Aloisi et al., 1992; Ehrlich et

al., 1998). The role of IL-8 and its receptors play a crucial role in the recruitment of

neutrophils (Horuk et al., 1997), bone marrow stromal cells (Wang et al., 2002), in cell

adhesion, neuronal protection and brain development (Goczalik et al., 2008). In humans,

IL-8 binds to two receptors, CXCR1 and CXCR2. CXCR1 has been shown to be

important in cell activation, while CXCR2 mediates the chemoattractant properties

(Semple et al., 2010). Interestingly, several in vitro studies have shown that IL-8 has also

a neurotrophic effect (Araujo and Cotman, 1993). However, little is known about the

role of Il-8 in the healthy brain and whether IL-8 is produced by resting microglia. On the

other hand, it is well known that activation of microglia induces IL-8 secretion (D'Aversa

et al., 2008). Our study clearly shows that human microglia produce detectable amounts

of Il-8 in our non-activated in vitro model. Furthermore, exposure to lead increased

significantly IL-8 production, both at low and high lead concentrations. This is consistent

with a recent study that showed increased IL-8 expression in peripheral blood monocytes,

when challenged with lead at the concentration of 10 µM and above (Gillis et al., 2012).

Interestingly, IL-8 has also been shown to be produced by microglia, astrocytes and

neurons in human Alzheimer disease, upon amyloid-β (Aβ) or pro-inflammatory

cytokine activation. Moreover, IL-8 has an inhibitory effect on Aβ-induced neuronal

apoptosis in vitro, along with an increase secretion of brain derived nerve growth factor

(Ashutosh et al., 2011). With this in mind, it is tempting to speculate that lead may affect

susceptible CNS regions, including the hippocampus, by inducing production of IL-8 in

microglia, which then interferes with cellular homeostasis and cell turnover, resulting in

the known effects of decreased neuronal density and corresponding functional

deficiencies.

Page 122: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[101]

Chemokine receptors contribute to communication between neurons and glia cells,

including microglia. Most important, their action is not restricted to neuroinflammation,

but they play a role in cellular and molecular homeostasis (Bajetto et al., 2001). E.g. the

chemokine receptors CXCR3, CXCR4 and CCR3 play a role during brain development

and growth (Van Der Meer et al., 2001). Several studies have reported the contribution

of various chemokine receptors to the pathogenesis of neurodegenerative diseases, such

as Alzheimer’s disease, Parkinson’s disease, multiple sclerosis, stroke and HIV-

associated dementia (Heneka et al., 2010; Mines et al., 2007). Interestingly, it has been

reported that knocking out CCR2 expression enhances microglia recruitment in a mouse

model of Alzheimer’s disease. Although in our study lead decreased expression of most

chemokine receptors, only CCR1, CCR2, CCR3 and CXCR1 were significantly reduced

in microglia. On the other hand, increased in expression level of CCR4, CCR9 and

CXCR3 was observed upon lead exposure. Interestingly, among the tested markers,

CCR5 and CXCR1 have shown to be expressed by M-MG at two different levels (two

picks in histograms) in both control and lead-treated cells. In accordance with our data, it

has been reported that that human monocytic consist of two principal subsets with

different expression level of CXCR1 and CCR5 (Geissmann et al., 2003). Therefore, our

results may suggest existence of two sub-populations of monocyte-derived microglia

with high and intermediate expression level of both markers. It is noteworthy to mention

that both sub-population showed decreased expression in response to lead exposure.

However, the role of these changes in expression levels and the molecular mechanisms

driving these changes are still not understood.

Accumulating evidence suggests that the diverse biological activity of macrophages is

mediated by functionally distinct subpopulations that are phenotypically polarized by

their microenvironment and by exposure to inflammatory mediators. M1 macrophages

exhibit potent microbicidal activity, and release IL-12, promoting strong Th1 immune

responses. M1 macrophages are activated by type I cytokines, IFNγ and TNFα, or after

recognition of pathogen associated molecular patterns or PAMPs (e.g., LPS, lipoproteins,

dsRNA, lipoteichoic acid) and endogenous “danger” signals (e.g., heat shock proteins,

HMGB1). M1 exert anti-proliferative and cytotoxic activities, which is due in part to the

release of reactive oxygen and nitrogen species and pro-inflammatory cytokines (e.g.,

TNFα, IL-1, IL-6). M1 population is thought to contribute to macrophage-mediated

tissue injury. In contrast, M2 macrophages support Th2-associated effector functions. M2

macrophages release IL-10 and exert selective immunosuppressive activity, and inhibit

T-cell proliferation. M2 macrophages also play a role in the resolution of inflammation

through phagocytosis of apoptotic neutrophils, reduced production of pro-inflammatory

Page 123: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[102]

cytokines, and increased synthesis of mediators important in tissue remodeling,

angiogenesis, and wound repair (Benoit et al., 2008, Laskin, 2009). In the context of lead

poisoning, it seems that expression of certain chemokine receptors as well as quick

changes towards M1/M2 phenotype is affected in human microglia, which may

contribute to changes in cell migration, activation, turnover and function in the CNS and

finally be responsible for the neurotoxic effect of lead

5.7. Conclusion

In summary, low level lead exposure is not cytotoxic to human microglia, in the sense

that it does not affect viability, nor does it substantially activate the cells. However, lower

level lead exposure has functional and immune modulatory effects on the human

microglia, by increasing expression of IL-8 and changing significantly expression

patterns of various chemokine receptors. Those changes may be the relevant events in

acute and chronic lead toxicity by interfering with cell recruitment, cell replacement and

tissue remodeling in various areas of the CNS

Page 124: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[103]

Chapter 6

Lead alters the chemokine receptors

expression pattern of the human

microglia cell line (HMC3)

Page 125: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[104]

6. Lead alters the chemokine receptors expression pattern of the human microglia cell line (HMC3)

6.1. Introduction to the chapter

In the previous chapters, effect of lead exposure on human monocyte-derived mciroglia

was studied. Alteration in the expression pattern of chemokine IL-8 and pro-

inflammatory chemokine receptors was reported following by lead exposure (Chapters

4 & 5). Due to the important role of human microglia as the brain immune effectors

during inflammation, as well as neuroprotection against toxicity, the effect of lead

exposure was also investigated using the human microglia cell line HMC3 as an

additional estbalished human microglia model. HMC3 is an immortalized microlgia cell

line derived from human fetal brain (Janabi et al., 1995). There is not much known about

HMC3 characteristics and function. However, expression a number of chemokine

receptors has been reported by HMC3 (Flynn et al., 2003). Therefore, changes on

morphology, phenotype and function of HMC3 were investigated after exposure to

different concentration of lead acetate (1, 10 and 50 µM) for 24 hours. The main focus of

this chapter was to establish the underlying similarities in response to lead exposure

between the established human microglia cell line HMC3 with the human monocyte

derived microglia (M-MG) model.

Result showed that lead had no toxic effect on the morphology and viability of HMC3 at

concentrations below 10 µM. In addition, there was general down regulation pattern in

expression of chemokine receptors as well as surface molecules. Reduction in

stimulatory effect of HMC3 on activated PBMC was seen in presence of lead. Lead

exposure also correlated with changes in the secretion of IL-8 and IL-6 by HMC3.

In summary, the results showed in M-MG are valid, as they have been repeated and

shown to be similar, using the established HMC3 cells line. These findings also suggest

the immunomodulatory effect of lead on human microglia, through changes in the

chemokine/receptor expression patterns, which may affect CNS development,

remodeling and function.

Declaration of work:

All the experiments in this study were designed and conducted by SE under the

supervision of Prof. Luis Filgueira. The full draft of the manuscript has been prepared for

submission to an international peer reviewed journal.

Page 126: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[105]

6.2. Abstract

It has been well documented that lead even at low concentrations has neurotoxic effects

on the development and function of the brain. Its detrimental effects on the CNS can

vary from activation of glia cells, induced neuronal death, changes in synaptogenesis to

impaired learning and memory function. Microglia cells, the resident macrophages of the

brain, play an important role in homeostasis of the CNS as neuroprotective cells

(secretion of anti-inflammatory cytokine, chemokine and their receptors, neurotrophic

factors, etc). Microglia activation which usually occurs in response to stimuli (toxin), has

been shown to be involved in many neuroinflammatory processes. Involvement of glia

activation in lead-induced toxicity has been reported in animal studies. However, there is

no evidence showing the exact role of human microglia in lead induced toxicity of

inflammation.

Therefore, the aim of this study was to investigate the effects of low level of lead

exposure (10µM, 24 hrs) on a human microglia cell line at the cellular and functional

level. To obtain this aim, HMC3 were exposed to different concentration of lead 1-100

µM for 24 hours. Then, changes on its morphology, viability, chemokine and cytokine

expression pattern as well as its function as brain immune cells were studied.

Results showed that lead at lower concentration <10 µM had no toxic effects on the

viability and morphology of HMC3. Furthermor, non-activated HMC3 are able to uptake

lead and accumulate inside even at low level of lead exposure, as visualized with specific

fluorescent probe, using fluorescence microscopy and flow cytometry. In addition,

general down-regulation in the expression pattern of chemokine receptors and surface

molecules involved in microglia differentiation, activation and inflammation was

observed. Lead attenuated the stimulatory effects of HMC3 on activated T-cell

proliferation which implicating changes in antigen presenting ability in presence of lead..

Furthermore, lead at concentration of 1 and 10 µM significantly increased in the

secretion of IL-6 and IL-8, respectively. Several studies have reported the

neuromodulatory effect of IL-6 and IL-8 on CNS development and function.

In summary, our results indicate lead, even at low concentrations, has a significant

influence on the characteristics and function of the human microglia cell line HMC3.

These changes may be responsible for some of the effects seen in the context of lead

neurotoxicity.

Keywords: Human Microglia, Human microglia cell line HMC3, Chemokine receptor, lead

Page 127: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[106]

6.3. Introduction

Lead is an potent neurotoxin, which causes long lasting neurobehavioral deficits and

cognitive impairments in children even at the very low doses (Zurich et al., 2002).The

developing nervous system is more vulnerable to toxic effects of lead (Bressler et al.,

1999b, Kasten-Jolly et al., 2012, Kasten-Jolly et al., 2011b, Struzynska et al., 2007b).

There is evidence showing that lead exposure alters the immune responses in the brain in

a way that resembles neuroinflammation. It has been known that lead accumulates more

in glia cells (astrocytes, microglia) than in neurons (Tiffany-Castiglioni and Qian, 2001).

Lead-induced astrocyte activation has been shown to increase the BBB permeability,

which makes lead entre to the brain easier (Dyatlov et al., 1998). It has been suggested

that lead potentiates chemokine and cytokine secretion, as well as glutamate induced

oxidative stress by astrocytes. However, there is not much known about the role of

microglia in lead induced neurotoxicity in human. Recent published animal studies

suggest that lead exposure can activate microglia and increase secretion of some

inflammatory cytokines, such as IL-1 β and TNF-, as well as hippocampal neuronal

injury. It has also been suggested that lead neurotoxicity may be mediated by microglia

activation (Liu et al., 2012). Recent studies showed a correlation between learning,

memory deficits and microglia activation (Tanaka et al., 2011). Several studies have

shown a correlation between increasing level of blood lead and the etiology of

neurodegenerative diseases, such as Alzheimer’s and Parkinson’s disease, and

amyotrophic lateral sclerosis (Liu et al., 2006, Kamel et al., 2002, Coon et al., 2006), Liu

et al., 2006). Several studies have reported the negative effects of lead on the nervous

system, especially motor neuron related disorders and cognitive reduction in

occupational lead exposed workers (Karimooy et al., 2010, Khalil et al., 2009, Zheng et

al., 2011). Recent studies suggest that long-term exposure lead may be responsible for

neurological deficiencies even at low concentrations (Baranowska-Bosiacka et. al.,

2012). Thus, there is growing interest in studying the effects of lead on the CNS, not

only in children but also in adults. According to the harmful effects of lead on memory,

learning and cognitive function, great attention has focused over the past decade on the

hippocampus as a target for lead toxicity, particularly in young children (Marchetti,

2003). However, underlying mechanisms of lead toxicity are still not well known.

Hence, understanding the mechanisms of lead neurotoxicity may provide a basis for

developing a new therapeutic strategy, aimed at preventing vital behaviour abnormalities

induced by lead poisoning.

Page 128: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[107]

There is evidence of toxic effects of lead on the immune system (Fischbein et al., 1993).

Enhanced proliferation of T-cells has been reported previously, following lead exposure

at low concentrations (McCabe et al., 2001). In addition, it has been shown that exposure

to lead strongly enhances the susceptibility of rodents to endotoxin shock and parasitic

infections, by affecting the immune responses.

Microglia cells are the resident macrophages of the brain. As the immune cell of the

CNS, they play a key role in maintaining the homeostasis and function of the brain

through secretion of a variety of modulatory molecules, neurotrophic factors, and

through phagocytic ability and other immune-related functions during development and

adult life. In response to environmental stimuli, such as toxins or pathogens, “surveying”

microglia with ramified morphology rapidly turn into the amoeboid “activated” form.

Along with this morphological and functional alteration, microglia undergo several

changes at the cellular and molecular level, including changes in chemokine and

cytokine secretion, and surface molecules expression. Activated microglia act as the

effective phagocytes and antigen presenting cells in the CNS. It has been shown that

microglia phagocytosis play a critical role during neuronal development and

differentiation, as well. Furthermore, microglia express diverse membrane receptors in

order to recognize pathogen, toxins endogenous danger molecules and initiate immune

responses through presenting the antigen to immune cells (Block et al., 2007). Microglia

also can acquire the reactive or over activated form, which contributes to neurotoxicity

and neuroinflammation in the CNS. Microglia’s reactive phenotype displays a high

phagocytic and antigen presentation ability, as well as secretion of several pro-

inflammatory cytokine and reactive oxygen molecules (Tremblay et al., 2011,

Kettenmann et al., 2011, Ransohoff and Perry, 2009). There are still controversies about

turnover and proliferation capacity of the microglia during life span. Particular concern is

about the differentiation of the blood monocytes into the microglia in order to replenish

the resident population (Boche et al., 2013). There is published evidence that monocyte

migrate to the brain and become microglia during neurodegenerative diseases (Simard

and Rivest, 2004).

Most of the current data on microglia physiology and function has been obtained from

animal studies. Difficulties in obtaining human brain tissue samples, insufficient number

of cells in primary culture, as well as changes in its characteristics over time, are some of

the reasons restricting most of the studies to investigating microglia in animal models

instead of using human cells. Furthermore, there are only few established human

microglia cell lines that are commercially available. The human microglia cell line

Page 129: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[108]

HMC3 is one of the few well established human microglia cell lines that are available for

research (Janabi et al., 1995). HMC3 cells show typical microglia characteristics,

including the constitutive expression of Iba-1, CD14, as well as IL-6, and IFN- induced

expression of CD11b, CD68 and MHC class II (Li et al., 2009).

Chemokines are chemoattractant cytokines, well known for their ability to recruit

inflammatory cells from the blood to the CNS and to enhance inflammatory responses.

However, recent studies have also shown their role in the development and migration of

neuronal precursor cells, as well as in mediating the microglia-neuronal interaction.

Chemokines and chemokine receptors have been detected under physiological and

pathological condition in brain tissues, as well as in neuronal and glia cultures (Bajetto et

al., 2002) (Xia and Hyman, 1999, Glabinski et al., 2002, Rezaie et al., 2002). Elevated

levels of chemokines and their reciprocal receptors have been found during

neuroinflammation and in neurodegenerative diseases (Simpson et al., 2000a, Cartier et

al., 2005). Despite the important role of these molecules and their receptors in mediating

brain inflammation, the expression patterns and functions of chemokines and their

receptors under physiological conditions in human microglia are still not fully

understood. In vitro studies showed that human microglia express several chemokine

receptors under non- stimulatory and under inflammatory conditions (Chapter 4, Flynn et

al., 2003).

In the context of lead and inflammation, recent animal studies showed the involvement

of glia activation in lead-induced neurotoxicity and impaired cognitive function. It has

been reported that astrocytes sequester lead ions, maintaining ionic balance in the

nervous system thus protecting neurons from lead toxicity (Selvin-Testa et al., 1991,

Raunio and Tahti, 2001)Activation of astrocytes with increased in their differentiation

have been also reported in rat model of lead-toxicity (Struzynska et al., 2007b).

Activation of astroglia may often lead to loss of the buffering function and contribute to

pathological processes. In addition, recent in vitro and in vivo studies in rat reported that

lead could cause microglia activation and hippocampal neuronal injuries through lead-

induced inflammation (Liu et al., 2012). All these studies implicate the significant

involvement of glia cells in both, protection and immune responses during lead toxicity.

However, there is no reported data by other researchers about the role of human

microglia in lead induced neurotoxicity/neuroinflammation. Therefore, we investigated

the effect of lead exposure on the human microglia cell line HMC3 to answer the main

question, whether human microglia are activated by lead and how these changes could

affect microglia functions as immune cells of the brain. In this study, HMC3 were

Page 130: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[109]

exposed to different concentrations of lead over 24 hours. Changes in morphology,

phenotype and function were investigated. We also studied whether microglia as the

brain phagocytes are able to engulf and sequester the lead. Knowledge about the

influence of lead on human microglia cell focusing on immune modulators will help to

better understande the role of cell-cell communication in lead-induced neuronal

dysfunction and degeneration.

6.4. Material and methods

6.4.1. Cell culture

In this study, we used human microglia clone 3 (HMC3) cells, kindly provided by Prof.

Karl-Heinz Krause (University of Geneva, Switzerland). Cells were cultured in T25

tissue culture flask (Sarstedt) using DMEM/F12 Glutamax medium (Invitrogen)

supplemented with 5% FCS and 1% antibiotics/antimycotics (Invitrogen) at standard

humidified culture conditions (37oC, 5% CO2). The cells were passaged and used for

experiments when they reached 70–80% confluence.

6.4.2. Lead exposure

Lead acetate (Pb(CH3CO2)2 3H2Os (Sigma-Aldrich) was prepared at the concentration of

100 mM as stock solution under sterile conditions. In order to study the effect of lead on

HMC3, lead was directly added to the medium at the concentration of 5,10, 25, 50 and

100 µM (depending on the experimental design). Cells after 7 days in culture,were

challenged with lead for 24 hours before measurements were done.

6.4.3. Morphological studies

HMC3 cells were cultured in the presence or absence of 10 µM lead for 24 hours.

Morphological appearance and changes were documented by using an inverted Leica

DM IL phase contrast and fluorescent microscope. Images were taken with a

Photometrix Cool SNAP Fx digital camera and processed with the NIS- elements F3.0

software. Sytox green, nucleic acid stain (Invitrogen) was applied to exclude the dead

cells in morphological quantification (Luo et al., 2001). Cells were incubated with 1 µM

Sytox Green for 30 minutes at 37°C. Five random optical fields were selected. Images

were taken with fluorescent filter and at bright filed. After overlaying the images, a total

of 100 live cells were counted. In repeated independent experiments, we quantified the

numbers of branches in microglia stained with an antibody against Iba-1, a macrophage

and microglia marker, using a standard immunostaining protocol (Blomster et al., 2011).

Page 131: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[110]

Immunostaining was performed with microglia cultured on cover slips, fixed with 1%

paraformaldehyde in PBS, containing 2% sucrose. After permeabilization with 0.01%

TritonX100 in PBS, cells were incubated overnight at 4°C with rabbit anti-Iba-1

(1:100,Wako Pure Chemical Industries), with subsequent incubation for 1 hour with

Alexa Fluor 488-conjugated donkey anti-rabbit antibody (1:200, Invitrogen) and DAPI

(1:400, Roche). Slides were analysed using fluorescent microscopy as described above

and images were taken. According to previous studies, microglia with less than two

branches were scored as activated (Kauppinen et al., 2008).

6.4.4. Detection of up-take and intracellular accumulation of lead

We investigated up-take and intracellular accumulation of lead by microglia with the

fluorescent probe Leadmium Green AM (Molecular Probes, Invitrogen, Carlsbad, CA)

using flow cytometry. HMC3 were incubated with different concentrations of lead (5, 10,

25, 50 and 100 µM) overnight. Then, M-MG were incubated with 1uM Leadmium Green

AM dye in saline for 30 minutes to 2 hours. Cells were analyzed by flow cytometry on

FACSCalibure flow cytometery (BD Biosciences, San Jose, CA) using the 488nm

excitation and measuring fluorescent emission at 520 nm. Non-lead treated cells were

included in the experiment as a negative control (Zeller et al., 2010). FlowJo software

was used for further analysis of the flow cytometry data. Representative histograms for

each experimental groups were overlayed and mean fluorescent intensities (MFI) were

compared using the Student’s t-test. The experiments were repeated for five times with

the same results (n=5). Another fluorescent probe, which is less specific, New port Green

DCF (Molecular Probes, Invitrogen, Carlsbad, CA), was also used to detect the lead

uptake.

6.4.5. Viability assays

To assess the effects of lead on the viability of HMC3, MTS assays were carried out

(Cell Titer 96 Aqueous assay, Promega). Briefly, cells were seeded in 96 well plates at

the concentration of 2x104 cells/200 µl/well and cultured to reach the 70-80%

confluence. After incubation of cells with 0, 1, 10, 50 µM of lead for 24 hours at 37oC,

20 µL of cell titer 96® AQueous One Solution (MTS) was added directly to each well.

After 4 hours incubation, absorbance of the colorimetric reaction was measured by a

plate reader (Labsystem Multiscan RC) at 490 nm. Viability of lead-treated cells was

compared the non-treated control cells and analysed using the Student’s t-test and

ANOVA (Vairano et al., 2004).

Page 132: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[111]

6.4.6. Expression of surface markers

We studied the effect of lead (10 µM) on the expression pattern of chemokine receptors

in HMC3. Cells were incubated with 10 µM lead for 24 hours. After detaching and

washing twice with PBS, cells were stained with direct labelled antibodies, or using

primary unlabelled and corresponding secondary labelled antibodies. Non-lead treated

cells were included in all tests. In number of experiments, cells were exposed to IFN-γ

(10 ng/ml, Sigma-Aldrich) and dexamethasone (1 µM, Sigma -Aldrich) for 24 hours. A

panel of directly labelled mouse anti-human antibodies , CD45-FITC, CD14-PE, CD40-

PE, CD11c-PE, HLADR–PerCP, CD11b-AF488, CD11c-PE, CCR1-AF647, CCR2-

AF647, CCR4-PE-Cy7,CCR5-FITC, CXCR1-PE, CXCR3-AF488, CXCR5-FITC (BD

Pharmingen, San Jose, CA), CCR6-FITC and CCR7-APC (R&A systems, Minneapolis,

MN) were added at the supplier’s recommended concentration. Excitation and emission

wavelengths of the used fluorochrome were resspectively: FITC: 494, 519; PE:

496/546,578; PerCP: 482, 678; APC: 650, 660; Alexa Fluor-488: 495, 519; Alexa Fluor -

647: 650, 668. Corresponding labeled isotype control antibodies were included in all

experiments. The cells were incubated with antibodies for 30 minutes at 4oC, washed

with PBS and resuspended and fixed with 1% formaldehyde in PBS containing 2%

sucrose. The fixed samples were measured with a FACSCallibure (BD Biosciences).

Data analysis was performed using FlowJo software version 7.6.3 (TreeStar, Ashland,

OR).

6.4.7. Mixed leukocyte reaction (MLR)

The T-lymphocyte stimulatory capacity and corresponding antigen-presenting cell

(APC) properties of HMC3, in the presence and absence of lead (10 µM) compared to

phytohaemagglutinine (PHA) (7 µgr/ml, Sigma-Aldrich) activated T-lymphocytes, were

assesses in MLRs using a BrdU proliferation assay (Cadosch et al., 2010). Freshly

processed non-adherent PBMC containing mainly (>90%) T-lymphocytes (2x105/well)

were seeded in a round bottom 96–well plate (Sarstedt). Allogeneic HMC3 were used as

antigen-presenting cells (APC) and added at the ratio (1:80) to the T-lymphocyte (4 wells

for same condition). T-lymphocytes with APC only was used as the control condition. To

compare the effect of lead on activated T-lymphocytes, PHA was added to each well at

the concentration of 7 µg/well. PHA- activated T-cells with HMC3 was used as the

positive controls. After 5 days in culture, BrdU proliferation assays (Roche, Mannheim,

Germany) were performed according to the supplier’s protocol and the absorbance of the

Page 133: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[112]

colorimetric reaction was measured by ELISA reader at 405nm (Labsystems Multiskan

RC). Mean proliferation of the different conditions was analyzed and compared.

6.4.8. Phagocytosis assay

Phagocytosis was assessed by measuring the uptake of tetramethylrhodamine (TMR)-

labelled Staphylococcus aureus (Invitrogen) as published before (Meagher et al., 2005).

To compare the effect of lead with IFN- and LPS on phagocytic activity of HMC3 and

its activation, cells were incubated overnight with 10 and 50 µM lead acetate, IFN- (10

ng/ml, Sigma-Aldrich)or LPS (10 ng/ml, E. coli serotype 0111:B4, Sigma -Aldrich).

. Next day, bacterial particles were added to the cells (107/ml) at a ratio of 1:1000

(according to manufacturer’s protocol) and incubated for 4 hours at 37oC. The cells were

then harvested and fixed with 1% paraformaldehyde in PBS containing 2% sucrose.

Total ingested fluorescent-labeled bacteria per cell were measured by flow cytometry.

6.4.9. Cytokine measurement

The cytometric bead array (CBA) human inflammatory kit (BD Biosciences) was used

for the quantitative measurement of cytokine production and secretion under control and

lead-exposed conditions. For that purpose, HMC3 were incubated with 1 and 10 µM

lead for 24 hours. To see whether HMC3 were responsive to IFN- and LPS activation,

cells were incubated with these activators at the concentration of 10 ng/ml for 24 hours.

The following day, 50-100 µl of the medium was collected and stored at -20oC. The

assay was performed according to the manufacturer’s instruction using 50ul of the

human cytokine standards and sample dilution. Cytokine concentrations were measured

using the FACSCanto flow cytometer (BD Bioscience). FCAP Array, version 3, software

was used to analyze the data. Measurements were done in duplicates and repeated once

with 3 independent experimental repeats.

6.4.10. Statistics

Data were analyzed with the SPSS statistical software (version 17.0). A one-way

ANOVA and univariant linear model were used to compare means. The LSD/Dunnett

post-hoc test also used to determine the significant difference between groups. In all the

analyses p value<0.05 was considered statistically significant. Bars in graphs represent

the standard error (SED).

Page 134: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[113]

6.5. Results

6.5.1. Intracellular detection of lead uptake in human microglia cell

line HMC3

Phagocytic ability of microglia play an important role in maintaining the brain

homoestasis in the healthy brain through clearance of debris and death cells. Low

phagocytic activity of HMC3 has been reported previously (Janabi et al., 1995).

Therefore, to detect the lead uptake by HMC3 and its intercellular accumulation, we used

the fluorescent probe Leadmium Green, which has been shown to be very sensitive and

specific probe for lead detection at nM levels. For this purpose, HMC3 were incubated

with different concentrations of lead (5, 10, 25, 50 and 100 µM) for 24 hours at 37oC.

Following incubation with 1µM Leadmium green for 30-60 minutes, the mean

fluorescent intensity (MFI) of the Leadmium was measured using flow cytometry and

compared to the non-lead treated cells as negative control (Figure 6.1). Increase in

intracellular amount of lead has been seen in a concentration-dependentmanner from 5

µM onwards. A major increase in green fluorescent intensity was seen from 10 µM to 25

µM. Mean of fluorescence intensity ± SD of the Leadmium Green for the HMC3

cultured with different concentration of lead, 0 to 100 µM respectively were 5.92 ± 0.96,

10.78 ± 0.95, 18.96 ± 0.58, 32.90 ± 1.84, 35.83 ± 2.24 and 40.02 ± 2.65.

Figure 6.1: Lead uptake by human microglia cell line HMC3

These figures show uptake

and intracellular accumulation

of lead in human microglia

using flow cytometry (n=5).

Figures A and B are the

representative FSC:SSC dot

plots of M-MG in

absent/presence of 10 µM lead

from one representative

experiment out of five with

similar results Histograms

compare the relative expression

(MFI) of fluorescent probes,

Leadmium (C) to the

corresponding amount of

intracellular lead. the absence

Page 135: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[114]

(a) or presence of various concentration of lead, including 5 µM (b), 10 µM (c), 25 µM (d), 50

µM (e) and 100 µM (f).

6.5.2. Effect of low level lead exposure on the morphology and

viability of HMC3

Quick changes in morphology from ramified to ameboid in response to stimuli is one of the

main characteristic of microglia. Under non-stimulatory condition (no lead) HMC3 showed

the typical resting morphology of microglia with long branches (Figure 6.2 A). Exposure to

10 µM for 24 hours lead did not have any significant effect on HMC3 morphology

regarding size of the cell and/or number and size of branches. However, exposure to higher

concentration of lead caused changes in morphology towards hypertrophy in cell body and

shortening of the branches. HMC3 treated with LPS, known microglia activation factor,

showed similar morphology to 50 µM lead which suggesting lead could induce activation in

HMC3 at higher concentrations of lead.

The effects of lead on the viability of the HMC3 at different concentrations and incubation

periods were studied using the MTS colorimetric assay. Cells were cultured with 1, 10 and

50 µM lead for 24 and 72 hours. Non-lead treated cells and IFN- stimulated HCM3 were

included in experiments as negative and positive controls, respectively. Cell viability were

determined by optical density (OD) measured at 490 nm. The measured optical densities

from different experimental condition are shown as percentages. The viability of non-lead

treated control cells was considered as 100%. The bar chart summarizes the effects of

different concentrations of lead 1, 10 and 50 µM over 24 hours and 72 hours (Figure 6.2. E).

at both exposure times, a slight viability increase was seen in at 1 µM, which was followed

by a slight decrease at 10 µM. In general, lead at lower concentrations (1 and 10 µM)

showed no significant effects on viability of HMC3 after 24 or 72 hours. However,

treatment with 50 µM lead acetate for 24 and 72 hours showed significant reduction in

viability (P<0.05) (n=20).

Page 136: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[115]

Figure 6.2: Effect of lead on morphology and Viability of HMC3.

Morphological changes of HMC3 exposed to different concentrations of lead 0 µM (A), 10

µM (B), 50 µM (C) for 24 hours. Cells were also cultured with LPS (10 ng/ml) for 24 hours to

obtain activated morphology (D). Images were taken with the phase contrast light microscopy

(n=5, bar indicates 100 µm). Arrows (red) show the morphological changes in HMC3 such as

hypertrophy in cell body as well as shortening of the branches E) Bar charts represent the

percentages of live cells after exposure to various lead concentrations (0, 1, 10 and 50 µM) for

24 hours (black bars) and 72 hours (white bars). Viability of the cells was assessed by a MTS

colorimetric test. IFN- was used as HMC3 activation marker (positive control).Viability of

negative control group (non-lead treated) was used as baseline and normalized to 100% (n=20,

* indicates significance with p < 0.05).

Page 137: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[116]

6.5.3. Effect of of lead exposure on expression of chemokine

receptors and surface molecules in HMC3

Chemokine and chemokine receptors play a key role in modulating the inflammatory

responses in the CNS. Previous studies have shown the microglial expression of different

chemokine receptors (Flynn et al., 2003). To study the influence of lead exposure on

chemokine and surface molecules expression patterns, cells were incubated with 10 µM

lead for 24 hours. Following staining, flourscent intenisty of tested markers were

measured using the flow cytometry. General down-regulation pattern were seen in the

expression level of studied chemokine receptors in HMC3 treated with 10 µM lead

(Figure 6.3). Among the tested chemokine receptors, CCR1, CCR2, CCR5, CXCR1 and

CXCR5 showed the siginificant down-regulation (P= 0.009,0.038, 0.042, 0.015 and

0.05) in lead tretaed HMC3 compating to the non-treated group (Figure 6.4). Although,

CCR9 expression level showed increase in lead-treated HMC3, this changes was not

statically significant.

In additon to chemokine receptors, we also compared the expressions pattern of surface

markerc in HMC3 before and after lead exposure. Similar to chemokine recetors, overall

down–regulation was observed among surface molecules following lead exposure

(Figure 6.4). Significant attenuation in the expression of CD45, HLA-DR and CD11b

and CD54 was observed (P=0.05,0.04, 0.028 and 0.042). Interstingly, lead showed no

significant effect on expression of CD68 as an activation marker, while CD11c

expression was up-regulated in lead treated HMC3 (P=0.022).

Studies showed that microglia during activation and inhibition expressed different level

of chemokine and chemokine receptor in distinct patterns. In order to identify whether

lead behaves as an activator or a suppressing agent on microglia, we compared the

chemokine receptor expression pattern in HMC3 exposed to lead with IFN-γ and

dexamethasone, well-known inflammation stimulator and inhibitors (Figure 6.5).

Therefore, cells were cultured with 10 µM lead, IFN-γ and dexamethsone for 24 hours.

The different expression level of chemokine receptors were compared with non-treated

control group. Most of the markers were responsive to IFN-γ and dexamethsone

stimulation , up- or down regulation of expression, respectively. Interstingly, lead did not

behave similar to IFN-γ nor dexamethasone in term of chemokine receptor expression

pattern.

Page 138: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[117]

Figure 6.3: Effect of lead exposure (10 µM, 24hrs) on chemokine receptor expression patterns in human microglia HMC3.

Examples of one representative experiment. Black histograms for lead-untreated control cells

and white histograms for lead treaded cells. Blue histograms are the isotype controls. Cells

were cultured for 7-10 days (n=20). Shifting black comparing to white histogram to the right is

representing the increase in expression level of marker in presence of lead and vice versa.

Page 139: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[118]

Figure 6.4: Lead alters the expression of surface markers in HMC3

A) Comparison between control cells (white bars) and lead-treated cells (black bars) for the

expression of various surface markers. B) Comparison between control cells (white bars) and

lead-treated cells 10 µM lead, 24 hour, for expression of various chemokine receptors. Data

represent the mean florescent intensity. Analysis of the significant differences between two

groups was done by ANOVA and LSD/Dennett’s test. (* indicates, n=20).

A

B

Page 140: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[119]

Figure 6.5: Lead showed distinct immunomodulatory effects on the surface molecule

expression in HMC3.

This diagram summarizes the comparison between stimulatory, inhibitory effects of IFN-

dexamethasone on the expression pattern of surface molecules in HMC3 and 10 µM lead-

treated HMC3. Non-lead treated HMC3 were included as a control. Data represents flow

cytometry measurements. Relative expression of marker were compared using the ANOVA

and LSD/Dennett’s test (n-=10).

CD11C CD68 CCR1 CCR2 CCR4 CCR5 CCR6 CCR7 CCR9 CXCR1 CXCR3 CXCR5

Page 141: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[120]

6.5.4. Effects of lead on phagocytic activity of HMC

Microglia are potent phagocytic cells of the brain and play a major role in the clearance

of dead cells and debris, especially during inflammatory processes caused by injuries or

neurodegenerative diseases. Phagocytic activity of microglia is also crucial in the

elimination of neurons during development. Phagocytic activity is usually enhanced

following microglia activation.and considered as one of the microglia activation marker.

Therfore, to determine the effect of lead on microlgia phagocytic ability and to find out

whether lead could enhance the microglia activation or not, we studied the different

concentration of lead (10 and 50 µM) on the phagocytic ability of HMC3 (Figure 6.6).

Changes on phagocytic ability of lead-treated HMC3 cells were also compared to IFN-

and LPS activated cells as positive controls. Non- treated HMC3 were considered as a

negative control. Fluorescent-labelled S. aureous was used for measuring the phagocytic

capacity of the various cell populations. Flow cytometry was used for the detection and

quantification of bacterial up-take. Our result showed that lead at the concentration of 10

µM did not change the phagocytic activity of HMC3. As we expected, lead at higher

concentration (50 µM) enhanced the bacterial engulfment similar to IFN- and LPS.

Figure 6.6: Effects of lead on phagocytic activity of HMC3.

Bar chart summarizes the phgocytic activity of HMC3 treated with different concentration of

lead (1, 10 and 50 µM) towards TM Rhodamin-lableled S. aureus measured with flow

cytometry. LPS and IFN- were used as the positive controls. Non-treated cells as negative

control were also included. Bacterial particles were added to culture medium at a ratio of

1:1000 (107 bacteria per ml) and incubated for 4 hours at 37

oC. Mean fluorescent intensity

represents the different levels of bacterial engulfment (P<0.05, n=7).

Page 142: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[121]

6.5.5. Influence of lead on microglia T-cell stimulatory capacity

Microglia as the resident immune cells of the brain have the unique capacity to process

and present antigens to T-lymphocytes which leads to activate and stimulation of T-cell

proliferation, as well as initiate the immune responses. To undestand whether lead could

enhance microglia antigen presentation ability through T-cell stimulation, we studied the

effect of lead exposure on T-lymphocyte stimulatory capacity of HMC3 using the mixed

leukocyte reactions (MLR) assay. In this study, non–activated and PHA- activated T-

cells were used as negative and positive controls, respectively. Co-culture of HMC3 and

T-cells were exposed to lead for 24 hours. To understand whether lead has more

stimulatory effect on activated T-cells, we included PHA and non-PHA treated T-cells in

one experimental condition. Optical density of culture was measured at 420 nm. The

higher number of cells represents the higher capacity in stimulation of T-cells. Optical

density of PHA-activated group was considered as 100%, which means the maximum

expected T-cell stimulation. A representative experiment is shown below (Figure 6.7).

Lead–treated HMC3 did not stimulate non-activated T-cells proliferation and showed

significant differences comparing to PHA-activated group (P=0.042). Regarding the

effect of lead on T-cell stimulation, result showed that lead–treated HMC3 had no

stimulatory effects on non-activated T-cells proliferation; however, lead-treated HMC3

significantly suppressed T-cell proliferation in PHA-activated experimental conditions

(P=0.036). In summary, low-level of lead exposure not only had no significant

stimulatory effect on T-cell proliferation by HMC3, it could attenuate antigen

presentation ability of microglia in response to activated T-cells.

6.5.6. Effect of lead on cytokine secretion in HMC3

Microglia activation has always been associated with the expression of cytokines, which

contribute to neurotrophic and neuroinflammatory events. Cytokines may also contribute

to the modulation of microglia activation (Vila et al., 2001).We used a flow cytometric

bead array for human inflammatory cytokines to determine whether lead exposure

induced changes in inflammatory cytokine expression patterns in human microglia. Cells

were cultured in the presence of different concentration of lead (1, 10µM) for 24 hours.

Cells were also incubated with HMC3 activators, IFN- and LPS (10 ng/ml) as a positive

control. Next day, culture medium supernatants were collected and used for the

measurement of a panel of inflammatory cytokines, including IL-1β, IL-8, IL-6, IL-

12p70, TNF- and IFN- (Figure 6.8). Of the six evaluated cytokines, only IL-8 and IL-6

showed changes in secretion level. It is worth to mention that HMC3 secreted cytokine at

the very low level. Interestingly, we found a significant increase in the secretion of IL-8

Page 143: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[122]

in a concentration-dependnet manner in the presence of lead (P=0.02, 0.04). When

HMC3 was treated with lead 10 µM, they showed only significnat decrease in IL-6

secretion (P=0.042). A slight increase was also showed in the presence of lead 1 µM,

however, it was not significant. HMC3 were responsive to stimulation with IFN- (10

ng/ml) followed by significant increase in secretion of both IL-6 and IL-8 (P=0.002,

0.000). LPS-treated HMC3(20 ng/ml) only showed increase in IL-8 expression

(P=0.026).

Figure 6.7: Influence of lead on microglia T-cell stimulatory capacity.

Comparing the T-lymphocyte stimulatory capacity of HMC3 in lead treated cells to PHA-

activated ones. Antigen presenting cells, allogenic T-cell were in cubated with HMC3 for 5

days at the ratio of 1:80 Cells. Non-treatd and PHA-activated cells were included as negative

and positive controls, respectively. Proliferation was measured using a BrdU incorporation

assay. Error bars represent the standard errors. No significant differences were seen between

control and test condition.

Page 144: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[123]

Figure 6.8: Effect of lead on cytokine secretion in HMC3

Cells were challenged with lead (0, 1 and 10µM), LPS 20 ng/ml and IFN- (10 ng/ml) for 24

hours. Negative control, no lead was also included in the experiment. Various cytokines were

measured in the culture supernatants using a cytometric bead assay and flow cytometry. Data

was analyzed by FCAP software. Final concentration of cytokines are shown as pg/ml (n= 10,

* indicates P<0.05).

Page 145: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[124]

6.6. Discussion

Despite the important role of microglia in the health and pathology of the CNS,

especially their involvement in inflammation-mediated neurotoxicity, there is not much

known about the role of human microglia in lead-exposed brain. Previously, we showed

the effects of lead exposure on human microglia using the novel in vitro model generated

from human blood monocytes (Chapter 4). In this study, we used the human microglia

cell line HMC3 as an additional human microglia model to investigate the effect of lead

on their morphology, phenotype and function, as well as to elucidate underlying

similarities between the HMC3 and M-MG models.

Lead is a widely spread persistent metal with potent toxic effects on human health.

Despite significant exposure reduction in humans, lead is still a major global health

concern. The developing central nervous system is one of the main targets of lead

toxicity. In children, neurotoxic effects of lead exposure can vary from changes in

neurocognitive function, in low-level exposures, to a potentially fatal encephalopathy in

acute lead poisoning, depending on the amount and length of exposure (Needleman,

2004b). In recent years, several studies have focused on the immunotoxic capacity of

lead. As a result, lead has been recognized as a new group of immunotoxicant with

potential capacity in shifting the immune function, even at the low to moderate levels of

exposure. It has been shown that lead could dramatically alter the function of the

immune system, even with moderate changes to immune cells (Dietert and Piepenbrink,

2006). There are evidences demonstrating a role of inflammation and inflammatory

mediators in pathogenesis of several CNS diseases (Lucas et al., 2006). In addition,

recent studies suggest the role of inflammatory responses in lead-induced neurotoxicity.

In that respect, microglia as the representative of the innate immune subsystem in the

CNS play a key role in inducing and modulating the immune responses in lead toxicity

(Struzynska et al., 2007b).

Microglia comprise 5-20 % of the glia population in CNS and their density varies in

different region of the brain (Denes et al., 2008). Under non-stimulatory condition,

microglia monitor surrounding environments and maintain homeostasis by removing

dead and injured cells, as well as by enhancing neuronal survival through secreting

neurotrophic and anti-inflammatory factors (Streit, 2002, Block et al., 2007). Microglia

activation, in response to injuries, trauma, toxin, etc., is followed by secretion of pro- and

anti-inflammatory cytokines, chemokines, expression of surface molecules, antigen

presentation, as well as phagocytosis in the CNS. It is becoming more accepted that,

although microglia activation is necessary and crucial for host defense and neuron

Page 146: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[125]

survival, over-activation of microglia could result in deleterious and neurotoxic

consequences.

During activation, microglia change their morphology from ramified to amoeboid

appearance, which usually is followed by an increase in the size of the cell body and by

shortening the branches. Microglia activation with changes in morphology under

inflammatory conditions in the presence of LPS in vitro has been reported (Sheng et al.,

2011). In addition, changes in morphology of microglia and astrocytes, following

activation, have also been reported in a number of lead-exposed animal models

(Struzynska et al., 2007b, Gillis et al., 2012). However, up to concentration of 50 µM,

lead had no toxic effects on the morphology or viability of the M-MG cells. Similarly,

we did not observe any changes in morphology or viability at lower concentration of lead

in HMC3. It seems that there is no clear evidence about negative effects of lead on

viability and proliferation of glia cells in CNS. A recent study has shown the broad

variability in size of microglia in low lead exposed animal (Sobin et al., 2013).

Interestingly, recent studies reported that treatment with LPS (100 ng/ml) and IFN- (10

ng/ml) could induce ramified morphology in a rat microglia cell line (Sheng et al., 2011).

However, in the presence of 50 µM or more lead, the size of HMC3 increased in a time-

dependent manner, similar to LPS treated cells, which is supported by published studies

(Espinosa-Oliva et al., 2009, Kim et al., 2000, Nakamura et al., 1999).

In the previous chapter, we showed that lead could alter the expression pattern of

chemokine receptors in M-MG model, general down regulation in expression of pro-

inflammatory chemokine receptors (Chapter 5). Chemokine receptors play an important

role in neuron-microglia communication, neuronal development as well as

neuroinflammation (Cartier et al., 2005, Mines et al., 2007). Therefore, novel therapeutic

strategies attempt to reduce or prevent the extent of chemokine/receptor expression by

microglia as well as other CNS cells to provoke neuroprotective processes. There is not

much known about the expression of chemokine receptors in healthy CNS in particular

non-activated microglia. Expression of a number of chemokine receptors by HMC3 has

been also reported (Flynn et al., 2003). Among the chemokine receptors, CCR1, CCR2,

CCR5, CXCR1 and CXCR5 showed higher levels of expression than others which may

implicate the involvement of these molecules in the natural function of microglia such

maintain homeostasis or cell turnover. Therefore, we were interested to investigate the

effect of lead exposure on chemokine receptor expression pattern in HMC3. We were

also wondering, if HMC3 would show similar responses to lead exposure. Our results

Page 147: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[126]

showed HMC3 expressed most of the chemokine receptors at low-medium level under

non-stimulatory condition similar to M-MG.

Following lead exposure, general down-regulation in expression patterns of chemokine

receptors were observed. Among the tested receptors, expression of CCR1, CCR2,

CCR5, CXCR1 and CXCR5 in lead-treated HMC3 showed the significant decrease

(P<0.05). Expression of CCR1, CCR2 and CXCR1 showed significant down-regulation

in lead-treated M-M. Recent evidence has shown involvement of CXCR1 and CXCR5 in

neurogenesis (Edman et al., 2008, Kizil et al., 2012). A distinct expression pattern of

CCL2/CCR2 has been documented during different stages of embryonic brain

development implicating a role for this chemokine network in development and

neurogenesis (Rezaie et al., 2002). On the other hand, enhanced expression of chemokine

receptors, CCR1, CCR2 and CCR5 have been demonstrated in neurodegenerative

diseases, such as Alzheimer’s disease, multiple sclerosis and Parkinson’s disease

(Eltayeb et al., 2007, Bagaeva et al., 2006). Therefore, it seems that lead alters the

expression of certain chemokine receptor in human microglia which may affect the cell

regeneration, migration and differentiation.

In this study, we also compared the effects of lead on chemokine receptor expression

pattern with dexamethasone and IFN- as well-known immune suppressant and

stimulants. We observed that lead did not behave similar to either of them, suggesting

lead has different effects on immune responses than dexamethasone and IFN-.

In the context of expression of surface molecules, in line with published data (Flynn et

al, 2003) and our M-MG model, HMC3 expressed CD45, CD11b, CD11c and CD68 ,at

low levels. Among the surface markers, CD54, adhesion molecules, showed the highest

expression level in HMC3 control cells. In the presence of lead (10 µM, 24 hours),

significant reduction in tested surface markers, including CD45, HLA-DR, CD11b,

CD11c and CD54, was seen (P<0.05). However, a similar decrease in expression of

surface markers was seen in lead-treated M-MG model.CD54, inter-cellular adhesion

molecules 1 (ICAM)-1, which has shown to be expressed by microglia in CNS, play

important role in trafficking leukocyte in the brain. Increased in expression of CD54

followed by microglia activation has been previously reported (Zuckerman et al, 1998,

Huber et al., 2006). In addition, it has been reported that CD54 plays an important role in

the antigen presentation ability of microglia together with MHC molecules and other co-

stimulatory molecules such as CD40 (Aloisi et al., 2000, Ma et al., 1999). Our results

showed that lead had no significant stimulatory effects on antigen presentation ability of

Page 148: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[127]

HMC3 and M-MG Decreased expression of HLA-DR, as well as co-stimulatory

molecules (CD40 and CD54) were observed.

Microglia are known as the phagocytes of the CNS (Nakajima and Kohsaka, 2001).

Increased phagocytic activity has been demonstrated by activated microglia which could

play both beneficial and detrimental during health and pathology (Rogers et al, 2001,

Fricker et al, 2012). HMC3 under basal condition showed low level of phagocytic

activity. Our result showed no influence of low level lead exposure on phagocytic

activity. However, at higher concentrations, significant increased phagocytosis was

observed, similar to that of LPS- and IFN--activated HMC3. In the context of effect of

lead on phagocytic activity of microglia, HMC3 and M-MG model showed a similar

responses (data not shown).

Using Leadmium Green as an specific lead detector probe, our result showed that HMC3

could take up lead intracellularly and accumulate inside the cell, similar to the M-MG

shown in the previous chapter. Otherwise, there is no other study showing lead uptake by

human microglia cells in vitro. However, lead uptake by astrocyte has been previously

reported. Similar to astrocytes, microglia may exert a neuroprotective mechanism against

toxic effects of lead on neuron especially during development by engulfing lead and

storing it intracellularly (Tiffany-Castiglioni and Qian, 2001).

We also compared the secretion of cytokines (IL-6, IL-8, TNF-, IFN-, IL-1β and IL-

12) in the absence and presence of lead in the HMC3 with M-MG models. HMC3 at the

non-stimulatory conditions secreted low detectable amount of IL-8 and IL-6. We have

observed both cytokines in supernants of non-tretaed M-MG. However, these cells

secreted IL-8 at the higher level than HMC3. In concordance with previous data, HMC3

exposed to different concentration of lead (1 and 10 µM) showed significant up-

regulation of IL-8 secretion, similar to M-MG. IL-8 is a chemokine secreted only in

human and shown to play an important factor in intercellular communication between

glia and neurons (Puma et al., 2001). IL-8 secretion by microglia has been previously

shown, which supports our findings in both M-MG and HMC3 (Ehrlich et al., 1998).

Interestingly, we also observed significant down-regulation in IL-6 secretion in HMC3

after lead exposure. IL-6 expression by non-activated human microglia has previously

reported (Janabi et al, 1995, Nagai et al, 2005) which supports our findings. Positive

effects of IL-6 has been reported on neurogenesis in the dentate gyrus of adult mice and

on memory performance of the animal (Baron et al., 2008).

Page 149: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[128]

6.7. Conclusion

In summary, findings of this study demonstrated that lead has similar effects on HMC3

and M-MG in the context of morphology, phenotype and function. These similarities

between M-MG and human microglia cells derived from the human fetal brain supports

M-MG as an novel, but valid in vitro model representing the characteristics of human

microglia. Like lead-treated M-MG, lead at low concentration (<10 µM) had no toxic

effects on the viability of HMC3. Lead-treated HMC3 showed no sign of activation

regarding the changes of its morphology or function. However, lead has a significant

influence on expression of chemokine receptors and markers important for microglia

proliferation, differentiation and activation. Similar to M-MG, HMC3 could take-up lead

under non-stimulatory conditions, accumulate lead intracellularly substantially, that may

cause the possible alteration in intracellular signaling and cellular functions. All these

results together suggest that despite not having direct toxic effects on human microglia,

lead exposure, even at low levels, may cause indirect negative effects on cell -cell

communication, which plays a key role on neuronal remodeling and synaptic activity.

In conclusion, one can postulate that both microglia models are equivalent and valid

to investigate lead toxicity in vitro in humans.

Page 150: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[129]

Chapter 7

Differential effects of lead on CX3CR1 and

TLR4 expression in human microglia

in vitro

Page 151: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[130]

7. Differential effects of lead on CX3CR1 and TLR4 expression in human microglia in vitro

7.1. Introduction to chapter

From the previous studies in chapters 5 and 6, lead had the functional and

immunomodulatory effects on human microglia. Lead exposure altered significantly the

expression patterns of various chemokine receptors in human in vitro microglia models.

In this chapter, we focus on the effects of lead on CX3CR1 and TLR4 expression pattern

in human microglia due to their important role in modulating inflammatory responses.

Over the past decade, several studies have shown the neurotoxic effects of lead exposure,

even at low concentration, on the impairment of learning and memory function

especially in children. Microglia as the immune effectors of the CNS are actively

involved in initiating the immune responses, secreting variety of cytokine and

chemokines as well as having normal role in the development and function of the brain.

The exact role of microglia in lead-induced neurotoxicity is still unknown. However, it

has been suggested that microglia activation may involve in lead-induced

neuroinflammation and neurotoxicity.

It has been known that chemokine and their receptors play a key role in modulating the

immune responses. There is evidence showing the interaction between fractalkine

(CX3CL1) and the fractalkine receptor (CX3CR1) regulates microglial activation in the

CNS. Numerous studies have revealed that the CX3CL1/CX3CR1 axis plays a direct role

in neurodegeneration/ or neuroprotection depending upon the CNS insult. CX3CR1

deficiency has been shown to be involved in impairment of hippocampal cognitive

function. Toll like receptor 4 (TLR4), the innate immune receptor, is expressed on

microglia and mediates the neuroinflammation. Numerous studies have shown TLR4-

dependent activation of microglia in neurodegenerative diseases. Developmental role of

TLR4 in learning and memory has also been demonstrated. In addition, a recent study

showed that lead could alter the susceptibility to different pathogens through TLR4

activation in murine macrophages.

In this study, first we investigated the differential expression of CX3CR1 and TLR4 in

non-treated M-MG and HMC3. Then, after 24 hours exposure to 10 µM lead acetate, the

changes on expression patterns of mentioned markers were measured using flow

cytometry. Results showed different expression level of CX3CR1, low and intermediate,

Page 152: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[131]

in M-MG derived from different individuals with dissimilar responses to lead exposure.

Previous studies have reported the polymorphisms in CX3CR1 gene. However, the

asscoaition between polymorphic variant of CX3CR1 and neuroinflammtion/

neurodegeeration remain incompletely defined. Human microglia cell line HMC3

expressed CX3CR1 at the low level. Lead has different effect on the expression pattern

of CX3CR1. Lead increased slightly the expression CX3CR1 in cells with low

expression level, however, CX3CR1 expression in the intermediate group down

regulated after lead exposure. These data may suggest the differential responses of

CX3CR1 to lead. We have observed the different responses to TLR4 as well which could

be possibly associated with different individual susceptibility to lead exposure. This

highlighted again that lead could altered the microglia function and inflammatory

responses through changing expression pattern of immune receptors.

Declaration of the work:

All the experiments were designed and conducted by SE involving cell culture, surface

marker expression (FACS) and statistical analysis under supervision Prof. Luis Filgueira

. SE was responsible for study design, experiments, data analysis and writing of the

manuscript. Dr. Marc J Ruitenberg contributed to study design, data analysis and editing

of the manuscript. Prof. Luis Filgueira contributed to study design, data analysis and

editing of the manuscript. A manuscript has been submitted to the Journal of

Neuroinflammation.

7.2. Abstract

Lead is a well-known environmental pollutant and neurotoxic heavy metal. Lead

neurotoxicity results in learning and memory impairment by probably altering

neurogenesis and neuronal turnover. Microglia cells have been proposed to contribute to

lead toxicity by promoting an inflammatory environment and interfering with neuronal

turnover. However, the exact mechanisms of how lead affects microglia phsyiology and

function at cellular and molecular levels is still not well understood. Due to the

importance of both receptors, CX3CR1 and TLR4, in modulation of neuroinflammation

towards neuroptotction or neurotoxicty; in this chapter, we hypothesized that lead may

alter inflammatory responses through changes on expression pattern of CX3CR1 and

TLR4 in human microglia. To conduct this aim, a newly established human monocyte-

derived microglia and the microglia cell line HMC3 were used as human in vitro

mcirolgia models. The cells were exposed to various lead concentrations (1µM to 100

µM). In addition,dexamethasone, as an immune suppressive agent, as well as interferon-

Page 153: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[132]

gamma, a known immune stimulatory agent were used to comapre the expression pattern

of CX3CR1 and TLR4 in presnce of lead. Expression levels of CX3CR1 and TLR4 was

measured using flow cytometry.

According to our results, CX3CR1 showed dimorphic expression in microglia cells

derived from different individuals, having either CX3CR1low

or CX3CR1intermediate

expressing cells. However, the human HMC3 microglia cell line belongs to the

CX3CRlow

expressing population. We showed that lead increased significantly CX3CR1

expression in CX3CR1low

cells, whereas it decreased significantly in CX3CR1intermediate

cells. TLR4 was expressed at low levels in all microglia populations. Intretsigly,

increased in TLR4 expression only was seen in the CX3CR1intermediate

microglia cells.

Dimorphic expression of CX3CR1 in human microglia from different individuals.is

supporting the presence of functionally relevant genetic polymorphisms in humans. In

addition, differential expression of CX3CR1 in the HMC3 cell line and the monocyte-

derived microglia may be possibly related to the different state of microglia

differentiation.

Furthermore, lead influences up-regulation of TLR4 expression in a CX3CR1

polymorphic dependent manner, indicating, that regulation of CX3CR1 and TLR4 must

be interconnected. Finally, our study indicates that lead interferes with regulation of

CX3CR1 and TLR4 expression. The detailed molecular mechanisms remain to be

investigated.

Keywords: human microglia, lead, CX3CR1, TLR4.

7.3. Introduction

Microglia are the resident immune cells of the central nervous system (CNS) and form a

dense cellular network throughout the nervous tissue. Thus, microglia contribute to the

first line of defence in the CNS. They play a key role in the homeostasis of the brain.

They are actively involved in removing cell debris and dead cells, including damaged

neurons. They also contribute to neurogenesis by producing neurotrophic factors (Aloisi,

2001, Ransohoff and Perry, 2009). They are activated in response to inflammatory

stimuli, following exposure to toxins or pathogens. Thereby, they change their dendritic

morphology towards round-shaped amoeboid cells, able to migrate, proliferate and

release inflammatory factors, as well as to increase the phagocytic activity. Several of

these functions are influenced or controlled by chemokines and corresponding receptors

Page 154: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[133]

expressed by microglia and the surrounding glia and neuronal cells (Bajetto et al.,

Kettenmann et al., 2011, Kim and de Vellis, 2005).

Chemokines are chemoattractant cytokines that play a vital role in leukocyte recruitment

and activation. The effect of chemokines is mediated by specific G-proteins-coupled

receptors. Interaction between the chemokine ligand and the receptor activates

downstream intracellular cascades, which act on regulation of gene expression,

modulation of cell cycle and changes in cell function. Chemokines and their receptors

have been shown to be expressed in all tissues and organs, including the CNS (Bacon

and Harrison, 2000). In the CNS, chemokines and their receptors contribute to attraction

and migration of neural stem and progenitor cells, to cell survival and differentiation,

cytokine production, myelination, neuro-vascularization and neuroprotection (Jaerve and

Müller, 2012). Fractalkine receptor (CX3CR1) is one such chemokine receptor. CX3CR1

is unique as it has fractalkine as its only, but very specific ligand. CX3CR1 has been

shown to control microglia activity and to contribute to migration and proliferation of the

cells. CX3CR1 deficiency results in microglia dysfunction, increased pathogen

susceptibility and increased neuronal cell death, as shown in CX3CR1 deficient animal

models (Lee et al., 2010). Importantly, CX3CR1 is constitutively expressed by microglia

in the CNS. On the other hand, fractalkine is expressed by neurons under healthy and

inflammatory conditions. Fractalkine has also been shown to be expressed by microglia

and astrocytes, especially following stimulation by inflammatory factors, including

tumor necrosis factor- or IL-1 β (Mizuno et al., 2003). Fractalkine can be found in two

forms, a membrane-bound and a soluble variant. The membrane form contributes to cell

adhesion, while the soluble form, produced from cleavage of the membrane bound form,

acts as a chemotactic agent and helps to recruit inflammatory cells (Chapman et al.,

2000). The membrane-bound form of fractalkine is cleaved by metalloproteases on

neurons upon cell activation (Hundhausen et al., 2003). Fractalkine has been reported to

be up-regulated and a neuroprotective factor in the context of neurodegenerative diseases

(Mizuno et al., 2003). Healthy neurons secrete fractalkine to modulate microglia

activation. On the other hand, interaction between neuronal fractalkine and its receptor

on microglia maintain microglia in their non-activated state (Gemma et al., 2010). The

neuroprotective properties of fractalkine are due to preventing neuronal cell death and

reducing the secretion of pro-inflammatory cytokines, including IL-1 β, IL-6 and tumour

necrosis factor-, as well as to enhance expression of iNOS by activated microglia

(Jones et al., 2010, Mizuno et al., 2003). Unfortunately, the exact role of CX3CR1

function in the brain remains controversial. In addition to the neuroprotective effects, the

fractalkine/CX3CR1 axis has also been shown to contribute to neurodegeneration.

Page 155: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[134]

Disruption of this axis may activate the microglia and induce secretion of pro-

inflammatory cytokines. Lack of microglial expression of CX3CR1 has been shown to

be neurotoxic and to induce neuronal cell death in models of systemic inflammation,

Parkinson’s disease and amyotrophic lateral sclerosis (Cardona et al., 2006). On the other

hand, in models of ischemic stroke and Alzheimer’s disease, deficiency in CX3CR1

expression reduced inflammation and neuronal cell loss, implicating its role in the

various aspects of microglia functions (Denes et al., 2008, Fuhrmann et al., 2010).

However, the fractalkine/CX3CR1 axis seems to play a vital role in the crosstalk

between neurons and microglia.

Microglia express various other membrane receptor proteins for the interaction with

neurons and other cells of the CNS (Noda and Suzumura, 2012). Some of those receptors

belong to the toll-like receptor (TLR) family, which are essential in molecular pathogen

recognition and may also contribute to phagocytosis. TLR signaling is initiated by

specific ligands, mostly related to the recognition of pathogen associated molecular

patterns, such as bacterial LPS and other microbial and viral products. After binding of

the ligand to the receptor, the cells are activated by a down-stream cascade, which results

in production of pro-inflammatory cytokines and chemokines, as well as expression of

corresponding surface receptors. TLR4 is an important member of the TLR family. In

addition to binding LPS, TLR4 mediates strong activation of microglia, by enhancing

phagocytosis, including uptake of β amyloid, and production of pro-inflammatory

molecules. On the other hand, deficiency in TLR4 expression or in function results in

reduced microglia activity, as shown in TLR4 knock out animal models (Song et al.,

2011). TLR4 is therefore considered to be a crucial component of the innate immune

response in the brain (Jack et al., 2005). TLR4 expression has been shown to be

exclusively expressed by microglia (Tanga et al., 2005). Microglia has thus been shown

to be the most responsive cells to LPS in brain (Eklind et al., 2001). In addition to the

exogenous LPS, TLR4 is also able to bind endogenous ligands, known as molecules

representing the danger-associated molecular pattern (DAMP), such as heat shock

proteins, S100 protein and high mobility group box 1 protein (HMGB1), which are

released by injured and necrotic cells (Kakimura et al., 2002). TLR4 is unique, as its

activation induces production of interferon-β and CXCL-10. Moreover, TLR4 has been

shown to play a role in auto-regulatory apoptosis in activated microglia, through

interferon-regulatory factor 3 (IRF-3) activation and interferon-β production (Jung et al.,

2005).

Page 156: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[135]

Despite the known roles of CX3CR1 and TLR4 in microglia activation, when broadly

investigated separately, no previous report has shown the direct or indirect interaction

between TLR4 and CX3CR1 under neuroinflammatory condition in human

microglia. However, there are evidences showing the activation via TLR4 can

regulate the CX3CL1/CX3CR1 expression in mouse microglia/macrophages

(Boddeke et al. 1999, Pachot et al., 2008). Furthermore, it has been reported that

activation of CX3CR1 can also potentiate the TLR4 activation through NF-kB

dependent signaling pathway in mouse macrophages (Donnelly et al., 2011, Ishida et

al., 2008).

Lead has been commonly accepted as a potent neurotoxin, especially in the developing

brain. However, the detailed mechanisms are still not well understood. A recent animal

study has reported the up-regulation of fractalkine in the lead exposed immature rat

brain, suggesting an inflammatory component in lead toxicity (Struzynska et al., 2007b).

Interestingly, CX3CR1 deficient mice have shown increased expression of IL-1 β in

response to LPS, with enhanced susceptibility to neurotoxins (Cardona et al., 2006,

Ransohoff et al., 2007). However, there is not much known about the effect of lead on

CX3CR1 expression in microglia. Other recent studies also suggest inflammatory effects

of lead by targeting TLR4 (Luna et al., 2012). However, the exact mechanisms are still

not known.

In this study, we investigated the effects of lead on expression of CX3CR1 and TLR4 in

human microglia using a novel human in vitro microglia model and the human microglia

cell line HMC3.

7.4. Material and Methods

7.4.1. Cell culture and experimental setting

In this study, we used a novel human in vitro microglia model, established by our group

(Etemad et al., 2012). Briefly, human peripheral blood monocytes were cultured in

RPMI 1640 with Glutamax and 1% antibiotics/antimycotic (all from Invitrogen,

Mulgrave, Victoria, Australia), supplemented with macrophage colony-stimulating factor

(M-CSF; 10 ng/ml; PeproTech, Rocky Hill, NJ), granulocyte-macrophage colony-

stimulating factor (GM-CSF; 10 ng/ml; PeproTech), β-nerve growth factor (NGF-β; 10

ng/ml; PeproTech) and CCL2 (100 ng/ml; PeproTech), at standard humidified culture

conditions (37oC, 5% CO2) for up to 14 days. Cell preparation of 30 blood donors were

used for this study

Page 157: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[136]

We also used the human microglia cell line HMC3, derived from human fetal brain

tissue (kindly provided by Prof. Karl-Heinz Krause, University of Geneva, Switzerland).

HMC3 cells were cultured in T25 tissue culture flask (Sarstedt,) using DMEM/F12 with

Glutamax (Invitrogen) supplemented with 5% FCS and 1% antibiotics/antimycotic. The

cells were passaged and used for experiments when they reached 70–80% confluence.

In order to study the expression pattern of CX3CR1 and TLR4 under different

experimental conditions, the human microglia and HMC3 were treated with lead acetate

(1µM, 10µM, 50µM and 100µM, Sigma-Aldrich, St Louis, MO). The cells were also

exposed for 24 hours to immune stimulatory factors, such as LPS (10 ng/ml, Sigma-

Aldrich) and human recombinant interferon-gamma (10 ng/ml, PeproTech), as well as to

the immune-suppressant dexamethasone (1µM, Sigma-Aldrich).

7.4.2. Flow cytometry

Expression patterns of CX3CR1 and TLR4 in human microglia under various

experimental conditions was studied using flow cytometry. The cells were detached

using a scraper (Sarstedt) and washed twice with PBS. Cells (1x106 cells/ml) were

stained according to a protocol recommended by BD and used previously (Etemad et al.,

2012). Cells were incubated for 1 hour at 4oC with the primary antibody and blocking

agent (10% FCS), washed with PBS and incubated for 30 minutes with secondary

antibody. Following the final wash, cells were fixed with 1% formaldehyde in PBS

containing 2% sucrose. The fixed samples were measured with a FACSCalibure (BD

Biosciences). Data analysis was performed using the FlowJo software version 7.6.3

(TreeStar, Ashland, OR).

A rabbit anti-CX3CR1 polyclonal antibody (AbD Serotec, Oxford, UK; 1 /100) and

mouse monoclonal anti-TLR4 antibody (Santa Cruz Biotechnology, Santa Cruz, CA;

1:100) were used, followed by a secondary donkey anti-rabbit AF647 and anti-mouse

AF488 antibody at the final concentration of 1:200 (Invitrogen).

7.4.3. Statistical analysis

Data were statistically analyzed with the SPSS 17.0 software. Student t- test and one-way

ANOVA were used to compare means. The Dunnett post-hoc test was also used to

determine the significant difference between groups. In all the analysis P<0.05 was

considered as statistically significant. Graphs with error bars represent the standard error

(SED).

Page 158: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[137]

7.5. Results

7.5.1. Differential expression of CX3CR1 in human microglia in vitro

We studied the expression of CX3CR1 in human microglia cells using two in vitro

models, monocyte-derived microglia cells (M-MG) and the microglia cell line HMC3.

Cells were cultured under standard protocols. It is noteworthy that M-MG were cultured

in serum free media, while HMC3 were culture in presence of 5% FCS. Once the cells

reached the optimal density, they were stained with anti-CX3CR1 antibody and the

fluorescence intensity was measured by flow cytometry. As expected, CX3CR1 was

expressed by non-lead treated, control M-MG. In addition, we observed that non-

activated, control M-MG from different human cell donors expressed CX3CR1 at

various levels, either low (n=18, MFI=19.87, CX3CR1low

) (Figure 7.1 A) or intermediate

expression levels (n=12, MFI=28.44, CX3CR1intermediate

) (Figure 7.1 B). The difference in

expression levels was statistically significant (P=0.027). However, the cells from the

different donors had the same morphological appearance (data not shown). We also

studied CX3CR1 expression in HMC3 cells, which was at low levels (n=20, MFI=17.15)

when the cells were non-activated (Figure 7.1 C), similarly to the CX3CR1low

M-MG and

statistically different from expression levels of CX3CR1intermediate

. CX3CR1 expression

levels of cells derived from low expressing (n=18) and intermediate (n=12) expressing

M-MG, as well as from HMC3 (n=20) were averaged and compared (Figure 7.1 D).

Page 159: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[138]

Figure 7.1: Expression levels of CX3CR1 in human M-MG and HMC3 cells.

Depending on cell donor, CX3CR1 is expressed at low levels (A: CX3CR1 low) or

intermediate levels (B: CX3CR1 intermediate). Interestingly, CX3CR1 expression in the

microglia cell line HMC3 is at low levels (C), similar to the CX3CR1 low M-MG. In (D),

CX3CR1 expression levels of cells derived from low expressing (n=18) and intermediate

(n=12) expressing M-MG, as well as from HMC3 (n=20) were averaged and compared.

7.5.2. Differential effects of lead on CX3CR1 expression in human

microglia

To study the effect of lead on CX3CR1 expression, M-MG and HMC3 were cultured in

the presence of various concentrations of lead for 24 hours. Then, the cells were stained

and fluorescence intensity was measured using flow cytometry (Figure 7.2). We

observed two different responses in M-MG from the different cell donors after lead

exposure. M-MG with primarily lower expression showed up-regulation of CX3CR1

after exposure to lead (Figure 7.2. A). However, lead reduced expression of CX3CR1 in

M-MG with primarily intermediate expression levels (Figure 7.2. B). As we expected,

HMC3 with low expression level of CX3CR1 at non-stiumulatory condition showed

significant ehancement in CX3CR1 expression following exposure to lead, similar to

CX3CR1low

M-MG responses (n=20; from 17.154 to 38.942; P=0.009) (Figure 7.2. C).

Page 160: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[139]

Figure 7.2. D summarizes the mean fluorescent intensity of lead- treated M-MG in

comparison with control in both responder populations and HMC3 cells. D. Lead

increased significantly expression of CX3CR1 in CX3CR1low M-MG (n=18; MFI: from

19.875 to 51.71; P=0.029), while CX3CR1intermediate

M-MG decreased CX3CR1

expression after lead exposure, although not significantly (n=12; MFI: from 28.44 to

18.9; P=0.05).

Figure 7. 2: Effect of lead on expression of CX3CR1 in human M-MG and HMC3 cells.

The cells were cultured for 24 hours in the presence of 10 µM lead. Histograms show

representative examples of CX3CR1 expression in M-MG (A and B) and HMC3 cells (C) after

lead exposure (white) in comparison with staining control (grey). Bar charts (D) compare lead

treated (black) with non-treated control (white) M-MG cells of basal low or intermediate

CX3CR1 expression, as well as HMC3 cells. Analysis of the significant differences between

two groups was done by ANOVA and LSD/Dunnett’s test. P<0.05.

Page 161: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[140]

7.5.3. Effect of immune modulators on CX3CR1 expression in

HMC3 cells

To investigate whether the dichotomy in CX3CR1 expression and corresponding

response to lead exposure was unique or representing a basic pattern of response in

human microglia, we included treatment of HMC3 cells with interferon-gamma as

immune activator and dexamethasone as an immune suppressant. Interestingly,

dexamethasone did not influence CX3CR1 expression in HMC3 cells, whereas

interferon-gamma significantly increased CX3CR1 expression (P=0.000), similarly to

lead (Figure 7. 3).

Figure 7. 3: Comparison of various stimuli on CX3CR1 expression in HMC3 cells.

The cells were incubated for 24 hours in the presence of lead (Pb), dexamethasone (Dexa) or

interferon-gamma (IFN-). Interestingly, lead and interferon-gamma increased significantly

CX3CR1 expression in HMC3 cells.

7.5.4. Effect of lead on TLR4 expression human microglia

TLR4 as an important class of innate immune receptors, is expressed on microglia and

play key role in recognition of endogenous ligands as well as invaded pathogen. TLR4

medaites inflammtory respomses and participate both in development and in responses

associated with CNS injury.Therefore, expression of TLR4 was measured in M-MG and

HMC3 using flow cytometry. Under non-activated condition, both CX3CR1 low and

intermediate M-MG cell sub-pupulation as well as HMC3cell expressed TLR4 at low

levels (Figure 7.4 A-C. Following lead exposure, CX3CR1intermediate

M-MG cells showed

siginificant increse in expression of TLR4 (MFI: from 17.89 to 30.7775;

P=0.048),whereas no changes was seen in TLR4 expression level in CX3CR1low

M-MG(

MFI from 15.68 to 1.48, P=0.35) nor HMC3 cells was seen (MFI: from16.136 to

Page 162: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[141]

13.821,P=0.45)(Figure 7.4 D-F). Expression levels of cells derived from low expressing

(n=18) and intermediate (n=12) expressing M-MG, as well as from HMC3 (n=20) were

averaged and compared (Figure 7.4 G).As expected, dexamethasone decreased

significantly TLR4 expression in HMC3 cells, whereas interferon-gamma increased

significantly TLR4 expression in HMC3 cells

Figure 7. 4: Effect of lead on expression of TLR4 in human M-MG and HMC3 cells.

The cells were cultured for 24 hours in the presence of 10 µM lead. Histograms (whites) show

representative examples of TLR4 expression in two M-MG sub-population and HMC3 cells in

absence (A-C) and presence of lead (D-F) and comparing the changes to iso type control

(gray).Bar charts (G) compare lead treated (black) with non-treated control (white) M-MG

cells of basal low or intermediate CX3CR1 expression, as well as HMC3 cells. Interestingly,

only CX3CR1intermediate

M-MG increased significantly TLR4 expression after lead exposure.

Analysis of the significant differences between two groups was done by ANOVA and

LSD/Dunnett’s test.

Page 163: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[142]

7.6. Discussion

In this study, we investigated the effects of lead exposure on expression of CX3CR1 and

TLR4 in microglia using two human in vitro cellular models.

Fractalkine is expressed by neurons and astrocytes as a membrane-bound form for cell

adhesion and as a soluble form for chemotaxis. CX3CR1 is expressed mainly by

microglia in the CNS. The fractalkine-CX3CR1 axis mediates neuron-microglia

interactions, and contributes to the control of cell activation, proliferation, and migration

(Hatori et al., 2002, Zujovic et al., 2000). In microglia, fractalkine/CX3CR1 interaction

maintains the cells in a resting functional state (Rogers et al., 2011). Following neuronal

injury, decreased fractalkine expression levels is therefore associated with enhanced

microglia recruitment and activation (Rogers et al., 2011). However, little is known

about the regulation of CX3CR1 expression in human microglia. We have previously

shown that the human microglia cell line HMC3 express CX3CR1 at low levels (Etemad

et al., 2012). In the present study, we observed that human microglia expressed CX3CR1

at low and intermediate levels depending on the cell donor, which suggests heterogeneity

of CX3CR1 expression in human microglia depending on genetic polymorphism. In this

regard, previous studies have identified two single nucleotide polymorphisms (SNP) in

the CX3CR1 gene in humans (Chan et al., 2005). It is not known, whether these

polymorphisms influence expression levels of CX3CR1. However, CX3CR1

polymorphisms are associated with increased risk of macular degeneration (Raoul et al.,

2010), HIV-1related AIDS progression in children (Singh et al., 2005) and diverse

survival of patients suffering from glioblastoma (Rodero et al., 2008). It has also been

published that the two CX3CR1 variants may have differential effects on microglia

function, including cell migration and adherence (Donnelly et al., 2011b). Interestingly,

CX3CR1 expression levels in human macrophage populations also depends on

differentiation and functional status (Geissmann et al., 2003), indicating that CX3CR1

may be functionally regulated in human microglia, depending on activation. However,

our study confirms the existence of dimorphic CX3CR1 expression in humans. Now, it

was of interest to investigate, whether lead influences CX3CR1 expression levels in

CX3CR1low

and CX3CR1intermediate

microglia. Interestingly, lead somehow increased

significantly CX3CR1 expression levels in CX3CR1low

microglia, but decreased

CX3CR1 expression levels in CX3CR1intermediate

microglia. These results indicate that

lead has an influence on the regulation of CX3CR1 expression in human microglia cells,

which can be seen as an inflammatory effect of lead, similar to interferon-gamma, as

confirmed by our results in HMC3 ells. However, it remains to be further investigated of

Page 164: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[143]

how and at what levels lead interferes with the regulation of CX3CR1 expression, and

whether the molecular interference of lead with CX3CR1 expression is different for the

two polymorphisms.

In this regard, recent studies showed the involvement of three polymorphic genes

related to lead toxicity (ALA-dehydratase, Vitamin D receptor gene, Hemochromatosis

gene), as well as sex differences in altering lead poisoning susceptibility in humans and

in animal models (Onalaja and Claudio, 2000). It has been suggested that these genetic

polymorphisms can potentially influence the accumulation of lead in the human body

and make some individuals more vulnerable to lead intoxication (Todd et al., 1996,

Sobin et al., 2009). Moreover, Schneider et al. (2011) reported that exposure to lead

results in significant changes in expression of various genes in the rat hippocampus and

various sex-dependent responses of the brain to lead exposure (Schneider et al., 2011).

These reports support our conclusion that lead interferes with the regulation of CX3CR1

expression and that further studies have to investigate the detailed mechanisms of this

interference. Considering the potential phenotypical and functional differences in

microglia between human individuals, one has to be careful in using only one given

microglia cell line, e.g. HMC3, which represents only one genetic make-up of only one

human individual. Consequently, we recommend to use our recently established new

human monocyte-derived microglia model for corresponding microglia studies to

consider genetic differences between human individuals with relevant functional impact.

When it comes to the discussion about the function of the fractalkine-CX3CR1 axis in

lead neurotoxicity, one has certainly to consider the controversial reports about the

function of this axis, as it includes neuroprotective and neuroinflammatory components.

However, all studies have been done in animal models and only little is known about this

axis in humans (Cardona et al., 2006). In the context of lead toxicity, knowledge about

the role of fractalkine-CX3CR1 axis remains also very scarce and further studies are

required (Struzynska et al., 2007b, Liu et al., 2012).In this study, we also investigated the

TLR4 expression pattern in human microglia models following lead exposure.

Interestingly, significant up-regulation in TLR4 expression was only seen in microglia

cells sub-population expressing the CX3CR1 at intermediate levels. These data indicate

that TLR4 expression is somehow connected to CX3CR1 expression in human microglia

as well as postulate the involvement of TLR4 in CX3CR1 dependent polymorphic

susceptibility against lead toxicity in a human population.

In that respect, the many different TLRs expressed on microglia and the TLR4 localized

on the surface of microglia is fisrt line of host defence against invading microorganisms

Page 165: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[144]

(Lehnardt et al., 2003). Microglia TLR4 is up-regulated upon brain inflammation and.

also mediates recognition of endogenous danger signals derived from stressed or necrotic

cells (Kakimura et al., 2002). Intriguingly, microglia TLRs tend to trigger a very

standard cytokine and chemokine response, irrespective of the type of TLR agonist they

meet. Which by microglia may be to recruit the assistance by other cells rather than to

immediately mount a destructive response towards invaders (van Noort and Bsibsi,

2009).

Although previous studies have reported that TLR4 activation, can regulate

CX3CL1/CX3CR1 expression and, vice versa, through activation of NF-kβ signaling

pathway (Boddeke et al., 1999, Donnelly et al., 2011, Ishida et al., 2008 and Pachot et

al., 2008), there is still not much known about interaction between TLR4 and CX3CR1

in the context of lead-induced neuroinflammation/neurotoxicity.

Since the shown reciprocal interaction has not been reflected in this study, our findings

may suggest the distinct effect of lead on expression pattern of TLR4 and CX3CR1 as

well as involvement of other signaling pathways in modulation of this interaction.

Therefore, more research is needed to identify the mechanisms behind the connection

between TLR4 and CX3CR1 regarding modulation of inflammatory responses in

microglia.

7.7. Conclusion

CX3CR1 is differentially expressed in human microglia, probably depending on genetic

polymorphism. Correspondingly, lead has differential effects on expression of CX3CR1

in microglia from different human individuals. Furthermore, lead influences up-

regulation of TLR4 expression in a CX3CR1 polymorphic dependent manner, indicating,

that regulation of CX3CR1 and TLR4 must be interconnected. Finally, our study

indicates that lead interferes with regulation of CX3CR1 and TLR4 expression. The

detailed molecular mechanisms remain to be investigated.

Page 166: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

Chapter 8

General discussion

Page 167: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[146]

8. General discussion and future directions

8.1. Summary of key findings

Lead has been known for a while as a potent neurotoxicant with severe detrimental

manifestations on the development and function of the CNS. Several human and animal

studies have reported significant effects of lead, even at lower blood lead levels (<2 µg

dl/0.1 µM), on learning ability and memory, as well as on cognitive functions of adults

(Olympio et al., 2009, Wang et al., 2012). Children and foetuses are two groups that are

highly susceptible to lead toxicity. It is evident that blood lead levels over 10

µg/dl/0.48µM (threshold limit) have negative effects on children’s brain development

and immune function (CDC, 2005, Finkelstein et al, Koller et al., 2004, Lanphear et al,

2000, Lidsky and Schneider, 2003).

CNS cells are the primary target of lead toxicity, especially during development. One of

the target cell populations are neurons, respectively neuronal precursor cells. They were

investigated in chapter 3.

In addition to neurons, involvement of glia cells in lead induced toxicity has been shown

in vitro and in vivo. Lead was shown to activate glia cells and to induce indirectly

neuronal cell death in animal models of lead toxicity. Astrocytes have been the major

glia population investigated in the past in the context of lead toxicity. They were shown

to take-up and store lead intra-cellularly, a phenomenon resembling the lead-

sequestering ability for mature astroglia in vivo (Tiffany-Castiglioni and Qian, 2001).

Microglia were investigated in chapters 4, 5, 6 and 7. Microglia as the immune cells in

the brain play an important role during health and pathology of the CNS (Kim and De

Vellis, 2005, Tremblay et al., 2011). As the brain macrophages, they share common

characteristics with other monocyte/macrophage lineages, including expression several

surface markers, expression of chemokine and chemokine receptors, secretion of

cytokines, antigen presentation and phagocytosis (Guillemin and Brew, 2004,

Kettenmann et al., 2011, Rock et al., 2004). Under healthy condition, microglia

constantly screen the microenvironment and modulate neuronal health and activity

through secretion of various neurotrophic factors (Nimmerjahn et al., 2005, Ransohoff

and Perry, 2009). Within the CNS, microglia are the first line of defense that respond

rapidly to any type of brain injury with changes in morphology and function towards

activation. Microglia activation is followed by secretion of several pro-inflammatory

cytokine and chemokine which can directly damage cells and lead to neuronal cell death

Page 168: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[147]

(Kreutzberg, 1995, Kettenmann et al., 2011, Ransohoff and Perry, 2009, Tambuyzer et

al, 2009). Thus, microglia are known to be the key cellular mediator of the

neuroinflammatory responses that can induce both, neuroprotection or neurotoxicity,

depending on the insult. Involvement of microglia activation has been recently reported

in animal model of lead-induced neurotoxicity, with enhanced secretion of pro-

inflammatory cytokines, as well as with induced neuronal injury (Liu et al., 2012).

However, there is not much known about the role of human microglia in lead induced

neurotoxicity/neuroinflammation. To investigate the role of human microglia in lead

toxicity in human, a new model was required, which was developed in the context of this

thesis (Chapter 4). Then the effect of low level of lead exposure (10 µM, 24 hours) was

investigated using the new human microglia model (Chapter 5). The results were then

confirmed using an established human microglia cell line (Chapter 6). Finally, two

important mechanisms were further investigated with these two microglia models,

namely fractalkine/CX3CR1 and TLR4 (Chapter 7).

The main aim of the first experimental chapter of this thesis (Chapter 3) was to

investigate the influence of lead on human neuronal precursor cells in vitro. The human

cortical neural cell line HCN2 was used as an in vitro human experimental model for this

study. Interestingly, lead was not really toxic to HCN2 cells, at least at lower

concentrations, which are probably those found in humans exposed to toxic lead levels.

With this in mind, it was of interest to investigate phenotypical and functional changes in

HCN2 cells. Since there were not many reported studies about the cellular characteristics

of HCN2 cells, further characterization was primarily needed. We showed that the non-

differentiated HCN2 cells represent the characteristics of neuronal precursor cells

(NPCs) in the context of morphology, as well as expressing neuronal precursor marker

(nestin, vimentin, β-tubulin III). Interestingly, expression of β-tubulin III was clearly

reduced in lead treated cells. Decrease in gene expression level of β-tubulin III in cortical

neurons with increasing postnatal development and differentiation has been reported

(Jiang & Oblinger, 1992). Lead driven differentiation was also supported by

corresponding morphological changes. This indicates that lead may drive differentiation

of HCN2 cells towards more mature neurons, something which may also happen in lead

exposed children and adults. In children, where brain development is still ongoing and

neuronal precursors differentiating in various brain regions contribute to it, lead may

have a more general detrimental effect on brain development by interfering with cell

differentiation. In adults, where neuronal precursors are mainly required in the

hippocampus, the memory function seems to be affected by lead toxicity. The

mechanisms of how lead influences directly HCN2 differentiation remains to be further

Page 169: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[148]

investigated. Interestingly, lead suppressed the secretion of IL-6 in HCN2, which may be

a possible mechanism for blocking differentiation (Monje et al., 2003, Nakanishi et al,

2007). In vitro animal models have also reported the suppressive effects of IL-6 on

differentiation of neuronal stem cells into neurons, as well as diminished hippocampal

neurogenesis.

In the context of phenotype, HCN2 expressed a number of chemokine receptors, such as

CCR1, CCR2, CCR3, CCR5, CXCR4, CX3CR1, implicating the involvement of

chemokine signaling in neuronal development, as well. Previous studies also showed that

chemokines, such as CCL2, play a key role in migration of NPCs towards the site of

injury during neuroinflammation (Belmadani et al., 2006). Following lead exposure,

expression of the mentioned chemokine receptors were down-regulated in HCN2 cells.

However, a significant increase in expression of CXCR1 and fractalkine was observed.

Changes of these receptors and factors may play a major role in lead neurotoxicity

resulting in the well -known clinical effects. Summarizing, these findings suggest that

lead may affect the development and function of human NPCs through changes in

chemokine signaling pathways, cytokine secretion and metabolic differentiation

pathways.

For this study, we also needed a good model representing the characteristics of human

microglia in vitro, that would consider polymorphism and diversity usually seen in

humans. In the second experimental chapter (Chapter 4), we developed a novel protocol

to generate human blood monocyte-derived microglia model. Detailed characterization

of M-MG revealed a cell population representing resting microglia with their specific

morphological, phenotypic and functional properties. The described protocol is easy to

handle, well standardized and very reproducible, as it uses only well defined culture

medium and recombinant cytokines, but no serum or conditioned medium. This protocol

was very helpful for investigating the influence of lead on human microglia.

To date, a number of animal and human models of microglia and microglia-like cell

lines have been used to examine neuroinflammatory phenomena. These models include

primary microglia cultures, and immortalized microglia cell cultures, which were either

retrovirus transformed or non-retrovirus transformed. These culture models share

similarities, but show also crucial differences that must be considered when choosing an

appropriate model for neurodegenerative research. Most of the common microglia

models used in several studies were derived from rodents (mice and rats). Difficulties in

obtaining human brain tissue samples, limited number of cells in primary culture, as well

as changes in their characteristics after isolationhave been some of the reasons restricting

Page 170: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[149]

microglia studies to animal models, instead of using human cells. In the context of

human immortalized microglia cell cultures, there are few established human cell lines

available for research. The human microglia cell line HMC3 is one of the few, well-

established human immortalized microglia cell lines (Janabi et al., 1995). HMC3 cells

were used in this study as an experimental model (Chapters 6 and 7). However, HMC3

are just representing one human, fetal individual, and results may be biased due to that

fact. Investigating the influence and effects of lead on human microglia has been the

main focus of this thesis (Chapters 5 and 6), with different aspects in mind, including

morphology, viability, surface marker expression, chemokine receptor expression

pattern, as well as cytokine secretion. For that purpose, monocyte-derived microglia and

the microglia cell line HMC3 have been used as human in vitro microglia models. In

both in vitro models, similar changes were observed in responses to lead exposure. Our

finding showed that lead had no cytotoxic effects on viability or morphology of M-MG

and HMC3 in culture. We also showed that lead at the low levels had no significant

impact on the phagocytic ability or the T- lymphocyte stimulatory capacity. Both

microglia cell types showed the ability to take-up lead in a concentration-dependent

manner. With these facts in mind, it is tempting to postulate that lead may not be really

cytotoxic to human microglia at lower concentrations, which are probably found in vivo

in exposed humans. However, there were substantial changes in other functions and

phenotype in microglia exposed to lead. In the context of phenotype, there was a general

down regulation of chemokine receptors. This indicates that lead may disrupt or change

migratory properties of microglia and the physiological interaction between microglia

and their neighbouring cells, including neuronal precursor cells. This interaction may be

further affected by lead-dependent changes in chemokine and cytokine secretion by

microglia and other cell types. In that context, one has to mention the increase in IL-8

secretion by M-MG and the enhanced secretion of IL-6.

Changes in these cytokines may well affect brain remodeling and recruitment of

neuronal precursor cells in the developing brain and in the adult hippocampus. In

summary, these studies provide a basis in understanding of the impacts of low-level of

lead exposure (<10µM, 24 hours) on human microglia in culture and changes in immune

properties of microglia as one of the possible involved mechanisms.

Finally, the last experimental chapter (Chapter 7) focused on the impacts of lead

exposure on expression of CX3CR1 and TL4 in human microglia. These two membrane

proteins pay a cruicial role in inflammatory responses, including inflammation in the

brain.

Page 171: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[150]

TLR4 is one of toll-like receptors, which is expressed by microglia in CNS and plays a

key role in the recognition of danger molecules secreted by injured CNS upon exposure

to pathogens, toxins, mechanical damage, etc (Rock et al., 2004, Olson and Miller,

2004b). TLR4 is one of the LPS receptors in conjunction with CD14 and its activation

triggers the complex intracellular signaling cascades, which cause induction of

cytokines and other inflammatory mediators (Laflamme et al., 2003, Qin et al., 2005,

Triantafilou and Triantafilou, 2002). TLR4 has been reported to induce microglia

activation and cytokine production in several neurodegenerative diseases (Heneka et al.,

2010, Lehnardt et al., 2003). Recent studies suggest that lead could alter the immune

susceptibility of macrophages to pathogen probably through interfering with the

binding of LPS to TLR4 (Luna et al., 2012). The mechanisms of how lead influences

changes of TLR4 expression remains unknown and needs to be further investigated.

Our findings showed that lead has differential effects on expression of CX3CR1 in

microglia from different human individuals. Interestingly, polymorphism has been

shown in humans for CX3CR1 (Garin et al., 2003, Singh et al., 2005). This polymorphic

property may be responsible for the dichotomy in response to lead in different

individuals. However, a correlation between lead response and CX3CR1 polymorphism

has to be investigated through future experiments.

Furthermore, lead influences up-regulation of TLR4 expression in a CX3CR1

polymorphic dependent manner. On the other hand, activation of macrophages via

TLR4 can regulate fractalkine and CX3CR1 expression (Boddeke et al., 1999, Pachot

et al., 2008). In addition, it has been shown that CX3CR1 can also increase NFkB

dependent transcription signaling via TLR4-dependent inflammatory responses

implicating the prominent regulatory role of CX3CR1 and TLR4 signaling during

inflammation (Donnelly et al., 2011a, Ishida et al., 2008). In summary, these data

emphasizes the importance of CX3CR1 and TLR4 in lead toxicity.

As lead showed significant increase in expression of IL-8 and CX3CR1 in human

microglia as well as their reciprocal receptors/ligands, CXCR1 and fractalkine in human

NPCs, the following paragraphs will focus on the importance of IL-8/CXCR1 and

fractalkine/CX3CR1 signaling pathways in the microglia-neuron interaction. In addition,

possible outcomes of lead interference on neurogeneration or neuroinflammation will be

discussed.

Page 172: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[151]

8.2. Neuronal remodeling and lead toxicity

There is evidence showing that lead exposure even at low concentration can cause

learning and memory impairments in animals and humans (Al-Saleh et al., 2009, Liu et

al., 2012). The hippocampus is a brain center for learning and memory (Bird and

Burgess, 2008). Long-term potentiation of the hippocampal excitatory synaptic

transmission is thought to be one of the underlying mechanisms for hippocampus-

dependent learning and memory (Bliss and Collingridge, 1993). Remodeling of brain

circuits, including the formation, modification and elimination of synaptic structures,

occurs throughout life as animals adapt to their environment. Until recently, known

mechanisms for experiment-dependent synaptic plasticity were focused on neuron-

astrocytes interaction (Stevens, 2008). However, microglia have been shown to be active

even under physiological condition and their processes actively in contact with dendritic

spines (Tremblay, 2011). Recent evidence shows that microglia may phagocytize

synaptic elements and modify synaptic plasticity during early postnatal development, as

well as after brain injury or during inflammation (Tremblay, 2011, Tremblay and

Majewska, 2011). Over the past decade, several studies suggested that lead interferes

with neuronal remodeling as one of the possible underlying mechanism in lead-induced

learning and memory deficits. It has been postulated that lead may specifically affect the

synaptic transmission; however, the molecular targets for lead are still unknown

(Bressler et al., 1999a, Goldstein, 1990b). In vitro studies suggested that exposure to lead

could cause changes in protein kinases activation in synapses and disrupted the normal

developmental sequence. Adhesion molecules also play key role in of synaptic plasticity.

Changes in adhesion molecules expression has been shown in lead induced neurotoxicity

studies (Davey and Breen, 1998, Hu et al., 2008, Hu et al., 2011). In general, the ability

of lead to substitute for other polyvalent cations such as calcium and zinc is regarded as

the main toxic mechanisms in CNS. These interactions allow lead to affect different

biologically significant processes, including metal transport, energy metabolism,

apoptosis, ionic conduction, cell adhesion, inter- and intracellular signaling, diverse

enzymatic processes, protein maturation, and genetic regulation. Membrane ionic

channels and signaling molecules seem to be the most relevant affected molecules

(Garza et al., 2006, Nemsadze et al., 2009, Rai et al., 2010, Xu et al., 2006b). Taken

together, lead could alter the neuronal remodeling process through changes in the

microenvironment of the brain. It also could affect signaling pathway, interfere with

communication of neuron with other cells (microglia, astrocytes) in the brain, and disrupt

Page 173: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[152]

normal development without producing signs of cell death. In the future, it will be

important to provide better understanding of the role of microglia in neuronal synaptic

modification during lead poisoning, and examine whether microglia function, such as

phagocytosis or secretion of immune modulators proceeds differently in health and

disease.

8.3. Why microglia are important in lead-neurotoxicity?

The importance of microglia as the immune cells of the CNS in degenerative diseases

makes them an interesting target for therapeutic approaches. The involvement of

microglia in neurotoxicity induced by heavy metal such as cadmium (Yang et al., 2007),

manganese (Crittenden and Filipov, 2008), cobalt (Mou et al., 2012) has been previously

shown using the in vitro and in vivo animal models. Several studies reported that such

metals could trigger microglia activation followed by increased in number and size of

glia cells, enhanced secretion of pro inflammatory cytokine such as TNF and IL-6, more

expression of oxidative stress and nitiric oxide (NO) (Kauppinen et al., 2008, Chen et al.,

2006). Beside activation, microglia cell death also has been reported in rat methyl

mercury induced neurotoxicity(Nishioku et al., 2000). Nuclear factor kappa B (NF-κB)

and p38 mitogen-activated protein kinase (MAPK) signaling pathways has been

suggested as the main involved signaling pathway in metal -induced cytokine

production in activated microglia.

In the context of lead toxicity as a heavy metal, animal studies revealed induced

astrocyte activation as the CNS glia cells following lead exposure. Activation of

astrocytes may lead to loss of buffering function and induced neuronal cell death. It has

also been showed that astrocytes could accumulate lead ions intracellularly as possible

neuronal protection from lead toxicity (Struzynska et al., 2007). Astrocyte activation by

lead has been associated with an increased number of cells and activation markers such

as GFAP as well as increased secretion of pro-inflammatory cytokine (Selvin-Testa et

al., 1991). There is evidence showing the toxic effects of lead on both microglia and

astrocytes in immature culture of rat brain (Zurich et al., 2002). Interestingly, recent in

vitro and in vivo animal study showed that lead can cause microglia activation, which

can up-regulate the level of IL-1β, TNF-α and iNOS, these pro-inflammatory factors

may cause hippocampal neuronal injury as well as long-term potentiation (LTP) deficits.

However, to our knowledge no previous studies investigated the effect of lead exposure

on human microglia in vitro.

Page 174: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[153]

On the other hand, learning and memory impairment as well as cognitive deficits are

one of the manifestations of lead toxicity in children. Over past decades, several studies

has focused on role of microglia in cognitive function of the brain (Ziv and Schwartz,

2008). Detrimental effects of microglia activation on learning and memory has been

reported (Cagnin et al., 2001). Inhibiting the activation can improve the cognitive

impairment in a murine model of human immunodeficiency virus (HIV) type 1

encephalitis (Keblesh et al., 2009). Therefore, better understanding of role of microglia

in lead-induced toxicity may be helpful to develop new pharmaceutical approaches in

treatment of clinical neurological disorders through inhibiting the microglia activation as

target mechanisms. As the main aim of this project, we were interested to investigate

whether low level of lead exposure could activate the human microglia through alteration

in its morphology, phenotype and function as well as neuronal precursor survival in

vitro.

8.4. Microglia-neuron interaction: A possible target of lead toxicity

Similar to previous reported data, in this thesis, we showed the expression of a variety of

chemokine receptors in both microglia and neuronal procures cells under non-

stimulatory conditions. Furthermore, secretion of several chemokines by neurons and

glia during CNS health and pathology has been reported, implicating the important role

of chemokine-receptor interaction in neuronal development and function (Bajetto et al.,

2001, Cartier et al., 2005, De Hass et al., 2007, Krathwohl et al., 2004, Sorensen et al.,

1999). It has been shown that microglia are actively involved in maintaining the brain

homeostasis and controlling the neuronal proliferation, migration as well as

differentiation during developmental period and adulthood (Bessis et al., 2007,

Cunningham et al, 2013, Morgan et al., 2004). As one of the remarkable finding of this

thesis, in the human neuronal precursor cells HCN2, down-regulation in expression level

of several chemokine receptors and surface markers was observed in the presence of

lead. Involvement of chemokine-receptors signaling in neuronal development

(proliferation, migration and differentiation), as well as neuroinflammation has been

previously reported. CCR2 is one of the chemokine receptors that play an important role

in recruiting monocytic cells to the site of injuries and initiate inflammatory responses

(Zhang et al., 2007, Wang et al., 2009). We showed down-regulation in CCR2

expression in both microglia and human neuronal precursor cells in the presence of lead.

Recruitments of the NPCs to the site of inflammation has been shown through CCR2

(Belmadani et al., 2006). Impaired microglia accumulation and early disease progression

has been reported in CCR2 deficient animal model of Alzheimer’s disease (El Khoury et

Page 175: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[154]

al., 2007). These data implicate the importance of CCR2 for cell migration in health and

disease. Therefore, lead-dependent changes in chemokine signaling pathways could be

one of the possible explanations for detrimental effects of lead on neuronal development,

as well as microglia function. At the same time, neurons influence glia functions,

through direct cell-to-cell interactions, as well as the release of soluble mediators such as

neurotransmitters (Carnevale et al., 2007). Lead through its ability to substitute for

calcium can affect the intercellular signaling calcium pathway, release and storage, as

well as expression of receptors for neurotransmitter, secretion of growth factors,

adhesion molecules, etc. (Lidsky and Schneider, 2003).

All these facts together implicate the important role of microglia-neurons interaction in

modulating the neuronal turnover, differentiation and function. Therefore, this

interaction could be an important target in lead-induced neurotoxicity. More studies

about the impact of lead in the context of cell-cell contacts will be helpful toward finding

new approaches in attenuating lead-induced neuronal impairment in the future.

8.5. Role of IL-8 / CXCR1 signaling in microglia-neurons interaction:

One of the signaling pathways in this study suggested to be affected by lead exposure

was IL-8/CXCR1. Our finding show that low level of lead exposure induces secretion of

IL-8 in a concentration-dependentmanner in microglia (Chapter 5), while it enhances the

expression of its receptor, CXCR1 in human NPCs (Chapter 3). This observation has led

us to hypothesize that lead at the low level of exposure may have an effect on microglia-

neuronal precursor communication through the IL-8/CXCR1 signaling pathway, a

mechanism, which may be involved in neurogenesis/neuroregeneration. In this study,

much of interest has focused on the chemokine IL-8. IL-8 is secreted by human cells, but

not present in rodents. IL-8 is known to be synthesized by glia cells in the brain.

Increased in IL-8 secretion by human microglia and astrocytes in response to LPS or pro-

inflammatory stimuli (IL-1β, TNF-α), as well as in neurodegenerative disease models

has been reported. On the other hand, anti-inflammatory cytokines such as IL-4, IL-10

and TGF-β down-regulate the production of this IL-8 (Ehrlich et al., 1998). IL-8 has

traditionally been known to have an important role in neuro-inflammation. However,

increasing evidence suggests that IL-8 also plays an important roles in CNS

development, neuronal survival, modulation of excitability, and neuroimmune responses

(Danik et al., 2003). Neurotrophic effects of IL-8 on survival of cortical neurons in

culture has previously been shown (Araujo and Cotman, 1993). Furthermore, IL-8 has

shown inhibitory effects on amyloid-β induced neuronal cell death, as well as

stimulatory effects on BDNF production in vitro (Ashutosh et al., 2011). Expression of

Page 176: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[155]

both IL-8 and its receptor CXCR1 by CNS cells postulate that the IL-8/CXCR1 axis may

exert neuroprotective effects and may regulate neuronal function by both, a paracrine

and an autocrine loop.

Interestingly, recent studies reported that human neuronal precursor cells express

receptors for IL-8 and CXCL13 (CXCR1 and CXCR5, respectively) and migrate across

brain endothelial cells in vitro, in response to these chemokines. We have observed the

expression of CXCR1 in human neuronal precursor cells (Chapter 3). Danik et al. (2003)

for the first time reported that CXCR1 mRNAs were expressed in different regions of rat

brain (septum, striatum, hippocampus, cerebellum, and cortex) at different levels and

appeared to be regulated independently from CXCR2 during development. They showed

expression of CXCR1 transcripts in different subtypes of neurons and astrocytes in the

rat brain (Danik et al., 2003). To our knowledge, there is no previous evidence showing

the lead toxicity induced IL-8 secretion in CNS (Chapter 5). However, enhanced

expression of IL-8 by human peripheral blood monocytes cultured in the presence of

lead, along with increased secretion of pro-inflammatory cytokines, has been recently

reported (Gillis et al., 2012). This indicates the involvement of IL-8 in lead-driven

inflammatory responses. Taken together, this information suggests the neuromodulatory

effects of the IL-8/CXCR1 axis in the CNS as part of lead neurotoxicity.

8.6. Importance of fractalkine/CX3CR1 in microglia-neuron interaction

Fractalkine (CX3CL1) is a unique chemokine because it is the only known member to

date of the CX3C class of chemokines (Bazan et al., 1997). Fractalkine is expressed as a

trans-membrane anchored protein that can be cleaved to yield the soluble forms. The

soluble form of fractalkine is thought to act more as chemoattractant molecules, while

the membrane-bound form functions as an adhesion molecule and provides cell-cell

interaction (Garton et al., 2001, Hundhausen et al., 2003, Tsou et al., 2001). In CNS,

fractalkine is mainly secreted by neurons. However, microglia and astrocytes have also

been shown fractalkine expression, but at a lower extent. Unlike other chemokines, there

is only one receptor for fractalkine, to date, i.e. CX3CR1 which make this

chemokine/receptor of interest to researchers. CX3CR1 is a G-protein-coupled receptor

that appears most highly expressed on all cells of monocytic lineage i.e.

monocytes/macrophages, dendritic cells and microglia (Imai et al., 1997, Harrison et al.,

1998, Ruitenberg et al., 2008). From extensive studies, it appears that the

fractalkine/CX3CR1 axis plays a direct role in neurodegeneration and/or neuroprotection

depending on the CNS insult. In this study, we o observed increased secretion of

Page 177: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[156]

fractalkine by neuronal precursor cells, as well as increased CX3CR1 in microglia

following by lead exposure (Chapters 3 and 7). Similar to our results, enhanced

expression of fractalkine has been shown in the hippocampus and forebrain of lead

exposed rat brain, suggesting that increase in fractalkine may act as chemoattractant for

microglia/macrophage recruitment and may be the primary responses of neuronal cell to

inflammation and toxicity (Struzynska et al., 2007). It has been surmised that the role of

fractalkine may be either chemotactic or pro-inflammatory in nature, or a protective

reaction against pro-inflammatory cytokines. Numerous studies reported the activation of

CX3CR1 could have neuroprotective effects, mainly through inhibiting the production of

pro-inflammatory cytokines (Cardona et al., 2006, Blomster et al., 2011). It has been

reported that fractalkine could suppress the production of pro-inflammatory cytokine,

such as IL-6 and TNF-, as well as nitric oxide by activated microglia in vitro.

Furthermore, suppression of neuronal cell death induced by microglia activation was

observed in presence of fractalkine (Mizuno et al., 2003, Zujovic et al., 2000).

In addition to neuroprotective/chemotactic role of fractalkine, interactions between

fractalkine and CX3CR1 contribute to microglia ability to maintain a resting phenotype.

However, when neurons are injured, fractalkine levels decrease, which results in

microglia recruitment and activation. Interestingly, polymorphisms in CX3CR1

associated with neuroinflammatory disorders, such as human age-related macular

degeneration (Chan et al., 2005), have been identified. In this study, differential

expression of CX3CR1 was also observed in monocyte -derived microglia generated

from different individuals that may be linked to the different susceptibility of the person

to immune responses and inflammation, and thus to polymorphism.

Recent studies also showed the importance of fractalkine/CX3CR1 signaling in

hippocampal cognitive functions. Disruption in fractalkine signaling has lead to

impairment in cognitive function and synaptic plasticity via increased action of IL-1β

(Rogers et al., 2011). Since cognitive impairment is one of the manifestations of lead

poisoning in children, fractalkine/CX3CR1 may play an important role in lead-induced

cognitive deficits.

To our knowledge, there is not much known about the exact role of fractalkine/CX3CR1

in lead induced inflammation/toxicity. However, increased fractalkine and CX3CR1 in

both microglia and neuron could be related to the neuroprotective properties through

modulate microglia activation as well as enhance neurogenesis. Therefore, future

investigation should focus on the interaction of microglia and neuronal precursor cells

(NPCs) considering the fractalkine/CX3CR1 axis.

Page 178: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[157]

8.7. Immune modulation by lead and IL-6 in the CNS: between

neuroprotection and neuroinflammation

Among the CNS cytokines, IL-6 is a cytokine that has demonstrated both

neurodegenerative and neuroprotective properties (Morales et al., 2010, Godbout and

Johnson, 2004, Peng et al., 2005). Emerging evidence suggest possible function of IL-6

as a physiological regulatory molecule and as a neuroprotective agent in brain pathology.

In this thesis, changes in cytokine and chemokine receptors expression pattern followed

by lead exposure has been shown in both human microglia and neuronal precursor

models in vitro, suggesting the possible immune modulatory effects of lead on CNS cell

development and function. Although IL-6 has been regarded a major inducer of immune

and inflammatory responses, accumulating evidence points to a crucial role of IL-6

within the CNS (Rose-John et al., 2006, Juttler et al., 2002). For instance, IL-6 promotes

the differentiation of cortical precursor cells into oligodendrocytes and astrocytes

(Nakanishi et al., 2007, Rajan and McKay, 1998), and also activates adult astrocytes

(Campbell et al., 1993). In addition, IL-6 may act as a developmental neurotrophic

factor (Marz et al., 1997, Schafer et al., 1999, Thier et al., 1999). It has been shown to

improve survival of neurons in culture (Akaneya et al., 1995, Hama et al., 1991),

protecting neurons from excitotoxic and ischemic insults (Loddick et al., 1998, Yamada

and Hatanaka, 1994, Matsuda et al., 1996), and promoting the growth of axons and

consequently the number of synapses in a region (Ihara et al., 1996, Gadient and Otten,

1997). Moreover, it has been shown that single and local blockade of IL-6 signaling

promotes neuronal differentiation from transplanted embryonic stem cell-derived neural

precursor cells (Gomi et al., 2011). On the other hand, impairment in synaptic plasticity,

changes in length and distribution of dendritic spine, reduced long term potentiation and

memory deficits has been shown following elevation in IL-6 in brain (Wei et al., 2012,

Bellinger et al., 1995). In response to prolonged exposure of lead, inflammation like glia

responses with increased secretion of pro-inflammatory cytokines such as IL-6, IL-1β

and TNF- and enhanced neuronal cell death has been reported in the immature rat brain

(Struzynska et al., 2007). On the other hand, recently Sobin et al. (2013) reported a

concentration-dependentreduction in IL-6 secretion in young mice with early chronic

lead exposure (Sobin et al., 2013). Consequently, rodent studies have to date provided

contradictory data.

In this thesis, using human in vitro models, low level of lead exposure decreased the

neuronal secretion of IL-6 (Chapter 3) with no effects on IL-6 secretion by microglia

Page 179: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[158]

(Chapters 5 and 6). Interestingly, it has been reported that during neurodegeneration,

injured neurons are able to secret the soluble IL-6 receptors while IL-6 is secreted by

microglia and astrocyte. Via trans-signaling, IL-6 bound to the soluble receptor can act

on neuronal precursor cells and induce proliferation and differentiation (Islam et al.,

2009). Overall, these findings from previous studies implicate that constant level of IL-6

play key role in healthy neuronal development and function. However, changes in

regular IL-6 patterns can induce neurodegenerative effects. Taken together, findings

from this thesis suggest that lead may interfere with neuronal turnover, differentiation

and protection through changes in expression pattern of IL-6 in neuronal precursor cells.

Further studies are required. Studies investigating the effect of lead exposure on neurons

in IL-6 knocked-out animal models or the blocking of IL-6 signaling may provide new

information about involvement of this cytokine in lead induced

neuroinflammation/neurotoxicity.

8.8. Research implication and future direction

This research has used human monocyte derived microglia as a human model for

investigating the effects of lead, which will provide valuable information about human

microglia function as immune effectors of the brain. Due to the important role of

microglia as both, neuroprotective and neurotoxic cells, the novel method introduced in

this work, mixture of cytokines in serum free condition, could be used as a cornerstone

for future human cellular microglia models as a reliable and convenient model in

comparison with current established cell lines such as HMC3.

Cognitive deficits, as well as neurobehavioral impairment, especially in children, are the

prominent manifestation of inflammation-mediated neurotoxicity by lead even at low

concentration. Therefore, further studies are needed to elucidate the exact role of human

microglia in lead-induced inflammation as key players in learning and memory. To

better understand the impact of lead on microglia in the brain, as well as on neuronal

development, migration and differentiation, it would be of much interest to investigate

the effect of lead exposure on the neuron- microglia interaction in a human in vitro co-

culture system, which was beyond the scope of this project.

At this point, results from this thesis have shed some light into the role of microglia

biology in lead toxicity in human with the precise concern on involvement of chemokine

and chemokine receptors. As chemokine-receptors biology accounts for a yet not well

understood point in neuroscience, more research about chemokines and lead may

Page 180: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[159]

contribute to the development of therapeutic strategies in mediating neuro-inflammatory

responses induced by lead.

8.9. General conclusion

In summary, this study, conducted as part of this thesis, provides new insights to the role

of microglia in lead neurotoxicity with a major focus on chemokine-receptor expression.

A novel human blood monocyte-derived microglia model has been developed. We

demonstrated that human microglia are able to uptake lead. Furthermore, we reported

that low-level of lead exposure has a significant influence on chemokine-receptor

expression patterns in both, human microglia and neuronal precursor cells in vitro.

Interestingly, our findings showed that lead could induce reciprocal enhancement of IL-

8/CXCR1 and fractalkine/CX3CR1 signaling in human microglia and neuronal precursor

cells, respectively. These results implicate that lead may have impacts on development

and function of CNS cells through altering the microglia-neuron interaction (Figure 8.1).

The data generated from this study present novel findings and indicate the potential

avenue for future research on human microglia and lead toxicity. These findings may

serve as a basis for a new approach to design future pharmacological therapies for the

treatment of lead neurotoxicity in children through the selective targeting of chemokine-

receptor function in the brain and modulate immune response to attenuate

neuroinflammation induced by lead poisoning in human.

Page 181: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

Figure 8.1: Neuromodulatory effects of lead on neuronal precursors and microglia

This figure summarizes the findings of this thesis and showing how lead may possibly have modulatory effects on the microglia neuron interaction

Page 182: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

Chapter 9

Bibliography

Page 183: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[162]

Abbott, N. J., Patabendige, A. A. K., Dolman, D. E. M., Yusof, S. R. & Begley, D. J. (2010) Structure

and function of the blood-brain barrier. Neurobiology of Disease, 37, 13-25. Abrous, D. N., Koehl, M. & Le Moal, M. (2005) Adult neurogenesis: from precursors to network

and physiology. Physiological Reviews, 85, 523-569. Adonaylo, V. & Oteiza, P. (1999) Lead intoxication: antioxidant defenses and oxidative damage

in rat brain. Toxicology, 135, 77 - 85. Adonaylo, V. & Oteiza, P. (1999) Pb2+ promotes lipid oxidation and alterations in membrane

physical properties. Toxicology, 132, 19 - 32. Ajami, B., Bennett, J. L., Krieger, C., Tetzlaff, W. & Rossi, F. M. (2007) Local self-renewal can

sustain CNS microglia maintenance and function throughout adult life. Nature Neuroscience, 10, 1538-43.

Akaneya, Y., Takahashi, M. & Hatanaka, H. (1995) Interleukin-1 beta enhances survival and interleukin-6 protects against MPP+ neurotoxicity in cultures of fetal rat dopaminergic neurons. Experimental Neurology, 136, 44-52.

Aloisi, F. (2001) Immune function of microglia. Glia, 36, 165-179. Aloisi, F., Carè, A., Borsellino, G., Gallo, P., Rosa, S., Bassani, A., Cabibbo, A., Testa, U., Levi, G. &

Peschle, C. (1992) Production of hemolymphopoietic cytokines (IL-6, IL-8, colony-stimulating factors) by normal human astrocytes in response to IL-1 beta and tumor necrosis factor-alpha. The Journal of Immunology, 149, 2358-66.

Aloisi, F., De Simone, R., Columba-Cabezas, S., Penna, G. & Adorini, L. (2000) Functional Maturation of Adult Mouse Resting Microglia into an APC Is Promoted by Granulocyte-Macrophage Colony-Stimulating Factor and Interaction with Th1 Cells. The Journal of Immunology, 164, 1705-1712.

Aloisi, F., Ria, F., Penna, G. & Adorini, L. (1998) Microglia Are More Efficient Than Astrocytes in Antigen Processing and in Th1 But Not Th2 Cell Activation. The Journal of Immunology, 160, 4671-4680.

Aloisi, F. (2001) Immune function of microglia. Glia, 36, 165-179. Al-Saleh, I., Nester, M., Mashhour, A., Moncari, L., Shinwari, N., Mohamed, G. E.-D. & Rabah, A.

(2009) Prenatal and postnatal lead exposure and early cognitive development: longitudinal study in Saudi Arabia. Journal of Environmental Pathology, Toxicology and Oncology, 28, 283-302.

Araujo, D. M. & Cotman, C. W. (1993) Trophic effects of interleukin-4, -7 and -8 on hippocampal neuronal cultures: potential involvement of glial-derived factors. Brain Research, 600, 49-55.

Ashutosh, Kou, W., Cotter, R., Borgmann, K., Wu, L., Persidsky, R., Sakhuja, N. & Ghorpade, A. (2011) CXCL8 protects human neurons from amyloid-beta-induced neurotoxicity: relevance to Alzheimer's disease. Biochemical and Biophysical Research Communications, 412, 565-71.

Bachstetter, A., Xing, B., de Almeida, L., Dimayuga, E., Watterson, D. & Van Eldik, L. (2011) Microglial p38alpha MAPK is a key regulator of proinflammatory cytokine up-regulation induced by toll-like receptor (TLR) ligands or beta-amyloid (Abeta). Journal of Neuroinflammation, 8, 79.

Bacon, K. B. & Harrison, J. K. (2000) Chemokines and their receptors in neurobiology: perspectives in physiology and homeostasis. Journal of Neuroimmunology, 104, 92-7.

Bagaeva, L. V., Rao, P., Powers, J. M. & Segal, B. M. (2006) CXC chemokine ligand 13 plays a role in experimental autoimmune encephalomyelitis. Journal of Immunology, 176, 7676-7685.

Bajetto, A., Bonavia, R., Barbero, S., Florio, T. & Schettini, G. (2001) Chemokines and their receptors in the central nervous system. Frontiers in Neuroendocrinology, 22, 147-184.

Bajetto, A., Bonavia, R., Barbero, S. & Schettini, G. (2002) Characterization of chemokines and their receptors in the central nervous system: physiopathological implications. Journal of Neurochemistry, 82, 1311-1329.

Balasubramaniam, B., Carter, D. A., Mayer, E. J. & Dick, A. D. (2009) Microglia derived IL-6 suppresses neurosphere generation from adult human retinal cell suspensions. Experimental Eye Research, 89, 757-766.

Page 184: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[163]

Banisadr, G., Gosselin, R.-D., Mechighel, P., Kitabgi, P., Rostène, W. & Parsadaniantz, S. M. (2005) Highly regionalized neuronal expression of monocyte chemoattractant protein-1 (MCP-1/CCL2) in rat brain: Evidence for its colocalization with neurotransmitters and neuropeptides. The Journal of Comparative Neurology, 489, 275-292.

Baranowska-Bosiacka, I., Gutowska, I., Rybicka, M., Nowacki, P. & Chlubek, D. (2012) Neurotoxicity of lead. Hypothetical molecular mechanisms of synaptic function disorders. Neurologia i Neurochirurgia Polska, 46, 569-578.

Baron, R., Nemirovsky, A., Harpaz, I., Cohen, H., Owens, T. & Monsonego, A. (2008) IFN-γ enhances neurogenesis in wild-type mice and in a mouse model of Alzheimer’s disease. The FASEB Journal, 22, 2843-2852.

Bassett, T., Bach, P. & Chan, L. H. (2011) Effects of methylmercury on the secretion of pro-inflammatory cytokines from primary microglial cells and astrocytes. Neurotoxicology, 33, 229-234.

Baydas, G., Nedzvetskii, V. S., Nerush, P. A., Kirichenko, S. V. & Yoldas, T. (2003) Altered expression of NCAM in hippocampus and cortex may underlie memory and learning deficits in rats with streptozotocin-induced diabetes mellitus. Life Sciences, 73, 1907-1916.

Bazan, J. F., Bacon, K. B., Hardiman, G., Wang, W., Soo, K., Rossi, D., Greaves, D. R., Zlotnik, A. & Schall, T. J. (1997) A new class of membrane-bound chemokine with a CX3C motif. Nature, 385, 640-4.

Becher, B. & Antel, J. P. (1996) Comparison of phenotypic and functional properties of immediately ex vivo and cultured human adult microglia. Glia, 18, 1-10.

Bellinger, D. C. (2004) Lead. Pediatrics, 113, 1016-1022. Bellinger, D. C. (2008) Very low lead exposures and children's neurodevelopment. Current

Opinion in Pediatrics, 20, 172-1777. Bellinger, F. P., Madamba, S. G., Campbell, I. L. & Siggins, G. R. (1995) Reduced long-term

potentiation in the dentate gyrus of transgenic mice with cerebral overexpression of interleukin-6. Neuroscience Letters, 198, 95-98.

Belmadani, A., Tran, P. B., Ren, D. & Miller, R. J. (2006) Chemokines Regulate the Migration of Neural Progenitors to Sites of Neuroinflammation. The Journal of Neuroscience, 26, 3182-3191.

Blevins, G. & Fedoroff, S. (1995) Microglia in colony-stimulating factor 1-deficient op/op mice. Journal of Neuroscience Research, 40, 535-544.

Ben-Hur, T., Ben-Menachem, O., Furer, V., Einstein, O., Mizrachi-Kol, R. & Grigoriadis, N. (2003) Effects of proinflammatory cytokines on the growth, fate, and motility of multipotential neural precursor cells. Molecular and Cellular Neurosciences, 24, 623-631.

Bennett, L. D., Fox, J. M. & Signoret, N. (2011) Mechanisms regulating chemokine receptor activity. Immunology, 134, 246-56.

Benveniste, E. N. (1997) Role of macrophages/microglia in multiple sclerosis and experimental allergic encephalomyelitis. Journal of Molecular Medicine (Berl), 75, 165-173.

Bessis, A., Béchade, C., Bernard, D. & Roumier, A. (2007) Microglial control of neuronal death and synaptic properties. Glia, 55, 233-238.

Biber, K., Dijkstra, I., Trebst, C., De Groot, C. J. A., Ransohoff, R. M. & Boddeke, H. W. G. M. (2002) Functional expression of CXCR3 in cultured mouse and human astrocytes and microglia. Neuroscience, 112, 487-497.

Bird, C. M. & Burgess, N. (2008) The hippocampus and memory: insights from spatial processing. Nature Reviews. Neuroscience, 9, 182-194.

Bleecker, M. L., Ford, D. P., Vaughan, C. G., Walsh, K. S. & Lindgren, K. N. (2007) The association of lead exposure and motor performance mediated by cerebral white matter change. Neurotoxicology, 28, 318-323.

Blevins, G. & Fedoroff, S. (19F95) Microglia in colony-stimulating factor 1-deficient op/op mice. Journal of Neuroscience Research, 40, 535-544.

Bliss, T. V. & Collingridge, G. L. (1993) A synaptic model of memory: long-term potentiation in the hippocampus. Nature, 361, 31-39.

Block, M. L., Zecca, L. & Hong, J. S. (2007) Microglia-mediated neurotoxicity: uncovering the molecular mechanisms. Nature Reviews. Neuroscience, 8, 57-69.

Page 185: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[164]

Blomster, L. V., Vukovic, J., Hendrickx, D. A., Jung, S., Harvey, A. R., Filgueira, L. & Ruitenberg, M. J. (2011) CX(3)CR1 deficiency exacerbates neuronal loss and impairs early regenerative responses in the target-ablated olfactory epithelium. Molecular and Cellular Neurosciences, 48, 236-245.

Boche, D., Perry, V. H. & Nicoll, J. A. R. (2013) Review: Activation patterns of microglia and their identification in the human brain. Neuropathology and Applied Neurobiology, 39, 3-18.

Boddeke, E. W., Meigel, I., Frentzel, S., Biber, K., Renn, L. Q. & Gebicke-Harter, P. (1999) Functional expression of the fractalkine (CX3C) receptor and its regulation by lipopolysaccharide in rat microglia. European Journal of Pharmacology, 374, 309-313.

Bohatschek, M., Kloss, C. U. A., Kalla, R. & Raivich, G. (2001) In vitro model of microglial

deramification: Ramified microglia transform into amoeboid phagocytes following addition of brain cell membranes to microglia-astrocyte cocultures. Journal of Neuroscience Research, 64, 508-522.

Bond, J. F. & Farmer, S. R. (1983) Regulation of tubulin and actin mRNA production in rat brain: expression of a new beta-tubulin mRNA with development. Molecular and Cellular Biology, 3, 1333-1342.

Boulanger, L. M. (2009) Immune proteins in brain development and synaptic plasticity. Neuron, 64, 93-109.

Boutin, C., Schmitz, B., Cremer, H. & Diestel, S. (2009) NCAM expression induces neurogenesis in vivo. European Journal of Neuroscience, 30, 1209-1218.

Bowen, K. K., Dempsey, R. J. & Vemuganti, R. (2011) Adult interleukin-6 knockout mice show compromised neurogenesis. Neuroreport, 22, 126-130.

Bradbury, M. W. & Deane, R. (1993) Permeability of the blood-brain barrier to lead. Neurotoxicology, 14, 131-136.

Brenneman, D. E., Schultzberg, M., Bartfai, T. & Gozes, I. (1992) Cytokine regulation of neuronal survival. Journal of Neurochemistry, 58, 454-460.

Bressler, J., Kim, K., Chakraborti, T. & Goldstein, G. (1999) Molecular mechanisms of lead neurotoxicity. Neurochemical Research, 24, 595 - 600.

Buchholz (1899) Ueber einen Fall syphilitischer Erkrankung des Centralnervensystems. Archiv fur Psychiatrie und Nervenkrankheiten, 32, 1-56.

Buisson, A., Lesne, S., Docagne, F., Ali, C., Nicole, O., MacKenzie, E. T. & Vivien, D. (2003) Transforming growth factor-beta and ischemic brain injury. Cellular and Molecular Neurobiology, 23, 539-550.

Butovsky, O., Ziv, Y., Schwartz, A., Landa, G., Talpalar, A. E., Pluchino, S., Martino, G. & Schwartz, M. (2006) Microglia activated by IL-4 or IFN-gamma differentially induce neurogenesis and oligodendrogenesis from adult stem/progenitor cells. Molecular and Cellular Neurosciences, 31, 149-160.

Cadosch, D., Sutanto, M., Chan, E., Mhawi, A., Gautschi, O. P., von Katterfeld, B., Simmen, H. P. & Filgueira, L. (2010) Titanium uptake, induction of RANK-L expression, and enhanced proliferation of human T-lymphocytes. Journal of Orthopaedic Research, 28, 341-347.

Cagnin, A., Brooks, D. J., Kennedy, A. M., Gunn, R. N., Myers, R., Turkheimer, F. E., Jones, T. & Banati, R. B. (2001) In-vivo measurement of activated microglia in dementia. The Lancet, 358, 461-467.

Cajal, S. R. (1913) Contribución al conocimiento de la neuroglia del cerebro humano Trabajos del Instituto Cajal de Investigaciones Biologicas, 11, 255-345

Cameron, H. A., Hazel, T. G. & McKay, R. D. (1998) Regulation of neurogenesis by growth factors and neurotransmitters. Journal of Neurobiology, 36, 287-306.

Campbell, I. L., Abraham, C. R., Masliah, E., Kemper, P., Inglis, J. D., Oldstone, M. B. & Mucke, L. (1993) Neurologic disease induced in transgenic mice by cerebral overexpression of interleukin 6. Proceedings of the National Academy of Sciences of the United States of America, 90, 10061-5.

Canfield, R. L., Henderson, C. R., Cory-Slechta, D. A., Cox, C., Jusko, T. A. & Lanphear, B. P. (2003) Intellectual Impairment in Children with Blood Lead Concentrations below 10 μg per Deciliter. New England Journal of Medicine, 348, 1517-1526.

Page 186: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[165]

Cao, X., Ma, J., Wu, G., Zhang, C., Wang, L., Dai, S. & Xu, W. (2012) Thymus-expressed chemokine promotes survival of PC12 cells via PI3K pathway. Neurochemistry International, 60, 163-169.

Cardona, A. E., Pioro, E. P., Sasse, M. E., Kostenko, V., Cardona, S. M., Dijkstra, I. M., Huang, D., Kidd, G., Dombrowski, S., Dutta, R., Lee, J.-C., Cook, D. N., Jung, S., Lira, S. A., Littman, D. R. & Ransohoff, R. M. (2006) Control of microglial neurotoxicity by the fractalkine receptor. Nature Neuroscience, 9, 917-924.

Carnevale, D., De Simone, R. & Minghetti, L. (2007) Microglia-neuron interaction in inflammatory and degenerative diseases: role of cholinergic and noradrenergic systems. CNS and Neurological Disorders Drug Targets, 6, 388-397.

Carpenter, D. O., Matthews, M. R., Parsons, P. J. & Hori, N. (1994) Long-term potentiation in the piriform cortex is blocked by lead. Cellular and Molecular Neurobiology, 14, 723-33.

Cartier, L., Hartley, O., Dubois-Dauphin, M. & Krause, K.-H. (2005) Chemokine receptors in the central nervous system: role in brain inflammation and neurodegenerative diseases. Brain Research Reviews, 48, 16-42.

CDC 1991. Preventing lead poisoning in young children-United States. Morbidity and Mortality Weekly Report. Centers for Disease Control and Prevention.

CDC 2005. Blood lead levels- United States, 1999–2002. Morbality and Mortality Weekly Report. Centers for Disease Control and Prevention.

Chalasani, S., Sabelko, K., Sunshine, M., Littman, D. & Raper, J. (2003) A chemokine, SDF-1, reduces the effectiveness of multiple axonal repellents and is required for normal axon

pathfinding.The Journal of Neuroscience, 23, 1360-1371. Chan, A., Magnus, T. & Gold, R. (2001) Phagocytosis of apoptotic inflammatory cells by

microglia and modulation by different cytokines: mechanism for removal of apoptotic cells in the inflamed nervous system. Glia, 33, 87-95.

Chan, C. C., Tuo, J., Bojanowski, C. M., Csaky, K. G. & Green, W. R. (2005) Detection of CX3CR1 single nucleotide polymorphism and expression on archived eyes with age-related macular degeneration. Histology and Histopathology, 20, 857-863.

Chan, W. Y., Kohsaka, S. & Rezaie, P. (2007) The origin and cell lineage of microglia—New concepts. Brain Research Reviews, 53, 344-354.

Chao, S. L., Moss, J. M. & Harry, G. J. (2007) Lead-induced alterations of apoptosis and neurotrophic factor mRNA in the developing rat cortex, hippocampus, and cerebellum. Journal of Biochemical and Molecular Toxicology, 21, 265-272.

Chapman, G. A., Moores, K., Harrison, D., Campbell, C. A., Stewart, B. R. & Strijbos, P. J. L. M. (2000) Fractalkine Cleavage from Neuronal Membranes Represents an Acute Event in the Inflammatory Response to Excitotoxic Brain Damage. The Journal of Neuroscience, 20, RC87.

Charlet, L., Chapron, Y., Faller, P., Kirsch, R., Stone, A. T. & Baveye, P. C. (2012) Neurodegenerative diseases and exposure to the environmental metals Mn, Pb, and Hg. Coordination Chemistry Reviews, 256, 2147-2163.

Chen, C. J. & Liao, S. L. (2003) Zinc toxicity on neonatal cortical neurons: involvement of glutathione chelation. Journal of Neurochemistry, 85, 443-453.

Chen, C. J., Ou, Y. C., Lin, S. Y., Liao, S. L., Chen, S. Y. & Chen, J. H. (2006) Manganese modulates pro-inflammatory gene expression in activated glia. Neurochemistry International, 49, 62-71.

Cheng, Y.-J., Liu, M.-Y., Wu, T.-P. & Yang, B.-C. (2004) Regulationof tumor necrosis factor-alpha in glioma cells by lead and lipposaccahride:involvement of common signaling pathway. Toxicology Letters, 152, 127-137

Chitu, V. & Stanley, E. R. (2006) Colony-stimulating factor-1 in immunity and inflammation. Current Opinion in Immunology, 18, 39-48.

Cho, C. & Miller, R. J. (2002) Chemokine receptors and neural function. Journal of Neurovirology, 8, 573-584.

Choi, W. S., Kim, S. J. & Kim, J. S. (2011) Inorganic lead (Pb)- and mercury (Hg)-induced neuronal cell death involves cytoskeletal reorganization. Laboratory Animal Research, 27, 219-225.

Columba-Cabezas, S., Serafini, B., Ambrosini, E., Sanchez, M., Penna, G., Adorini, L. & Aloisi, F. (2002) Induction of macrophage-derived chemokine/CCL22 expression in experimental

Page 187: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[166]

autoimmune encephalomyelitis and cultured microglia: implications for disease regulation. Journal of Neuroimmunology, 130, 10-21.

Conductier, G., Blondeau, N., Guyon, A., Nahon, J.-L. & Rovère, C. (2010) The role of monocyte

chemoattractant protein MCP1/CCL2 in neuroinflammatory diseases. Journal of Neuroimmunology, 224, 93-100.

Coon, S., Stark, A., Peterson, E., Gloi, A., Kortsha, G., Pounds, J., Chettle, D. & Gorell, J. (2006) Whole-body lifetime occupational lead exposure and risk of Parkinson's disease. Environmental Health Perspectives, 114, 1872-1876.

Cordova, F. M., Rodrigues, A. L., Giacomelli, M. B., Oliveira, C. S., Posser, T., Dunkley, P. R. & Leal, R. B. (2004) Lead stimulates ERK1/2 and p38MAPK phosphorylation in the hippocampus of immature rats. Brain Research, 998, 65-72.

Coughlan, C. M., McManus, C. M., Sharron, M., Gao, Z. Y., Murphy, D., Jaffer, S., Choe, W., Chen, W., Hesselgesser, J., Gaylord, H., Kalyuzhny, A., Lee, V. M. Y., Wolf, B., Doms, R. W. & Kolson, D. L. (2000) Expression of multiple functional chemokine receptors and monocyte chemoattractant protein-1 in human neurons. Neuroscience, 97, 591-600.

Craig, M. J. & Loberg, R. D. (2006) CCL2 (Monocyte Chemoattractant Protein-1) in cancer bone metastases. Cancer and Metastasis Reviews, 25, 611-619.

Crittenden, P. L. & Filipov, N. M. (2008) Manganese-induced potentiation of in vitro proinflammatory cytokine production by activated microglial cells is associated with persistent activation of p38 MAPK. Toxicology In vitro, 22, 18-27.

Crumpton, T., Atkins, D. S., Zawia, N. H. & Barone Jr, S. (2001) Lead Exposure in Pheochromocytoma (PC12) Cells Alters Neural Differentiation and Sp1 DNA-Binding. Neurotoxicology, 22, 49-62.

Cunningham, C. L., Martinez-Cerdeno, V. & Noctor, S. C. (2013) Microglia regulate the number of neural precursor cells in the developing cerebral cortex. Journal of Neuroscience, 33, 4216-4233.

Danik, M., Puma, C., Quirion, R. & Williams, S. (2003) Widely expressed transcripts for chemokine freceptor CXCR1 in identified glutamatergic, γ-aminobutyric acidergic, and cholinergic neurons and astrocytes of the rat brain: A single-cell reverse transcription-multiplex polymerase chain reaction study. Journal of Neuroscience Research, 74, 286-295.

D'Aversa, T. G., Eugenin, E. A. & Berman, J. W. (2008) CD40-CD40 ligand interactions in human microglia induce CXCL8 (interleukin-8) secretion by a mechanism dependent on activation of ERK1/2 and nuclear translocation of nuclear factor-κB (NFκB) and activator protein-1 (AP-1). Journal of Neuroscience Research, 86, 630-639.

Davey, F. D. & Breen, K. C. (1998) Stimulation of sialyltransferase by subchronic low-level lead exposure in the developing nervous system. A potential mechanism of teratogen action. Toxicology and Applied Pharmacology, 151, 16-21.

Davidovics, Z. & DiCicco-Bloom, E. (2005) Moderate lead exposure elicits neurotrophic effects in cerebral cortical precursor cells in culture. Journal of Neuroscience Research, 80, 817-825.

De Haas, A. H., van Weering, H. R., de Jong, E. K., Boddeke, H. W. & Biber, K. P. (2007) Neuronal chemokines: versatile messengers in central nervous system cell interaction. Molecular Neurobiology, 36, 137-151.

Denes, A., Ferenczi, S., Halasz, J., Kornyei, Z. & Kovacs, K. J. (2008) Role of CX3CR1 (fractalkine receptor) in brain damage and inflammation induced by focal cerebral ischemia in mouse. Journal of Cerebral Blood Flow and Metabolism, 28, 1707-1721.

De Simone, R., Ambrosini, E., Carnevale, D., Ajmone-Cat, M. A. & Minghetti, L. (2007) NGF promotes microglial migration through the activation of its high affinity receptor: Modulation by TGF-[beta]. Journal of Neuroimmunology, 190, 53-60.

Del Rio-Hortega, P. (1932) Microglia. Cytology and Cellular Pathology of the Nervous System, 2, 481-534.

Deverman, B. E. & Patterson, P. H. (2009) Cytokines and CNS Development. Neuron, 64, 61-78. Devi, C. B., Reddy, G. H., Prasanthi, R. P. J., Chetty, C. S. & Reddy, G. R. (2005) Developmental lead

exposure alters mitochondrial monoamine oxidase and synaptosomal catecholamine levels in rat brain. International Journal of Developmental Neuroscience, 23, 375-381.

Page 188: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[167]

Dheen, S. T., Kaur, C. & Ling, E. A. (2007) Microglial activation and its implications in the brain diseases. Current Medicinal Chemistry, 14, 1189-1197.

Dietert, R. R. & Piepenbrink, M. S. (2006) Lead and immune function. Critical Reviews in Toxicology, 36, 359-385.

Donnelly, D. J., Longbrake, E. E., Shawler, T. M., Kigerl, K. A., Lai, W., Tovar, C. A., Ransohoff, R. M. & Popovich, P. G. (2011) Deficient CX3CR1 signaling promotes recovery after mouse spinal cord injury by limiting the recruitment and activation of Ly6Clo/iNOS+ macrophages. Journal of Neuroscience, 31, 9910-9922.

Dribben, W. H., Creeley, C. E. & Farber, N. (2011) Low-level lead exposure triggers neuronal apoptosis in the developing mouse brain. Neurotoxicology and Teratology, 33, 473-480.

Dyatlov, V. A., Platoshin, A. V., Lawrence, D. A. & Carpenter, D. O. (1998) Lead potentiates cytokine- and glutamate-mediated increases in permeability of the blood-brain barrier. Neurotoxicology, 19, 283-291.

Edman, L. C., Mira, H. & Arenas, E. (2008) The beta-chemokines CCL2 and CCL7 are two novel differentiation factors for midbrain dopaminergic precursors and neurons. Experimental Cell Research, 314, 2123-2130.

Edman, L. C., Mira, H., Erices, A., Malmersjo, S., Andersson, E., Uhlen, P. & Arenas, E. (2008) Alpha-chemokines regulate proliferation, neurogenesis, and dopaminergic differentiation of ventral midbrain precursors and neurospheres. Stem Cells, 26, 1891-900.

Ehrlich, L. C., Hu, S., Sheng, W. S., Sutton, R. L., Rockswold, G. L., Peterson, P. K. & Chao, C. C. (1998) Cytokine Regulation of Human Microglial Cell IL-8 Production. The Journal of Immunology, 160, 1944-1948.

Ekdahl, C. T., Kokaia, Z. & Lindvall, O. (2009) Brain inflammation and adult neurogenesis: The dual role of microglia. Neuroscience, 158, 1021-1029.

Eklind, S., Mallard, C., Leverin, A. L., Gilland, E., Blomgren, K., Mattsby-Baltzer, I. & Hagberg, H. (2001) Bacterial endotoxin sensitizes the immature brain to hypoxic--ischaemic injury. European Journal of Neuroscience, 13, 1101-6.

Elkabes, S., DiCiccoBloom, E. M. & Black, I. B. (1996) Brain microglia macrophages express neurotrophins that selectively regulate microglial proliferation and function. Journal of Neuroscience, 16, 2508-2521.

Elkabes, S., Peng, L. & Black, I. B. (1998) Lipopolysaccharide differentially regulates microglial trk receptor and neurotrophin expression. Journal of Neuroscience, 54, 117-122.

El Khoury, J., Toft, M., Hickman, S. E., Means, T. K., Terada, K., Geula, C. & Luster, A. D. (2007) Ccr2 deficiency impairs microglial accumulation and accelerates progression of Alzheimer-like disease. Nature Medicine, 13, 432-438.

Eltayeb, S., Berg, A.-L., Lassmann, H., Wallstrom, E., Nilsson, M., Olsson, T., Ericsson-Dahlstrand, A. & Sunnemark, D. (2007) Temporal expression and cellular origin of CC chemokine receptors CCR1, CCR2 and CCR5 in the central nervous system: insight into mechanisms of MOG-induced EAE. Journal of Neuroinflammation, 4, 1- 14.

Esen, N. & Kielian, T. (2007) Effects of low dose GM-CSF on microglial inflammatory profiles to diverse pathogen-associated molecular patterns (PAMPs). Journal of Neuroinflammation, 4- 10.

Espinosa-Oliva, A. M., de Pablos, R. M., Villaran, R. F., Arguelles, S., Venero, J. L., Machado, A. & Cano, J. (2011) Stress is critical for LPS-induced activation of microglia and damage in the rat hippocampus. Neurobiology of Aging, 32, 85-102.

Etemad, S., Ruitenberg, M. & Filgueira, L. (2012) In vitro generated human microglia derived from blood precursors. The Journal of Immunology, 188, 111-114.

Fanarraga, M. L., Avila, J. & Zabala, J. C. (1999) Expression of unphosphorylated class III beta-tubulin isotype in neuroepithelial cells demonstrates neuroblast commitment and differentiation. European Journal of Neuroscience, 11, 517-527.

Farina, C., Aloisi, F. & Meinl, E. (2007) Astrocytes are active players in cerebral innate immunity. Trends in Immunology, 28, 138-145.

Finkelstein, Y., Markowitz, M. E. & Rosen, J. F. (1998) Low-level lead-induced neurotoxicity in children: an update on central nervous system effects. Brain Research Reviews, 27, 168-176.

Page 189: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[168]

Fischbein, A., Tsang, P., Luo, J. C. & Bekesi, J. G. (1993) The immune system as target for subclinical lead related toxicity. British Journal of Industrial Medicine, 50, 185-186.

Fischer, H.-G. & Reichmann, G. (2001) Brain Dendritic Cells and Macrophages/Microglia in Central Nervous System Inflammation. Journal of Immunology, 166, 2717-2726.

Flynn, G., Maru, S., Loughlin, J., Romero, I. A. & Male, D. (2003) Regulation of chemokine receptor expression in human microglia and astrocytes. Journal of Neuroimmunology, 136, 84-93.

Ford, A., Goodsall, A., Hickey, W. & Sedgwick, J. (1995) Normal adult ramified microglia separated from other central nervous system macrophages by flow cytometric sorting. Phenotypic differences defined and direct ex vivo antigen presentation to myelin basic protein- reactive CD4+ T cells compared. The Journal of Immunology, 154, 4309-4321.

Fricker, M., Oliva-Martin, M. & Brown, G. (2012) Primary phagocytosis of viable neurons by microglia activated with LPS or Abeta is dependent on calreticulin/LRP phagocytic signalling. Journal of Neuroinflammation, 9, 196.

Friedman, W. J. (2000) Neurotrophins induce death of hippocampal neurons via the p75 receptor. Journal of Neuroscience, 20, 6340-6346.

Fuhrmann, M., Bittner, T., Jung, C. K., Burgold, S., Page, R. M., Mitteregger, G., Haass, C., LaFerla, F. M., Kretzschmar, H. & Herms, J. (2010) Microglial Cx3cr1 knockout prevents neuron loss in a mouse model of Alzheimer's disease. Nature Neuroscience, 13, 411-413.

Fujimura, M. & Usuki, F. (2012) Differing effects of toxicants (methylmercury, inorganic mercury, lead, amyloid β and rotenone) on cultured rat cerebrocortical neurons: Differential expression of Rho proteins associated with neurotoxicity. Toxicological Sciences, 126, 506-514.

Fujita, H., Tanaka, J., Toku, K., Tateishi, N., Suzuki, Y., Matsuda, S., Sakanaka, M. & Maeda, N. (1996) Effects of GM-CSF and ordinary supplements on the ramification of microglia in culture: A morphometrical study. Glia, 18, 269-281.

Gadient, R. A. & Otten, U. H. (1997) Interleukin-6 (IL-6)—A molecule with both beneficial and destructive potentials. Progress in Neurobiology, 52, 379-390.

Gao, D., Mondal, T. K. & Lawrence, D. A. (2007) Lead effects on development and function of bone marrow-derived dendritic cells promote Th2 immune responses. Toxicology and Applied Pharmacology, 222, 69-79.

Garber, M. M. & Heiman, A. S. (2002) The in vitro effects of Pb acetate on NO production by C6 glial cells. Toxicology In vitro, 16, 499-508.

Garin, A., Tarantino, N., Faure, S., Daoudi, M., Lécureuil, C., Bourdais, A., Debré, P., Deterre, P. & Combadiere, C. (2003) Two Novel Fully Functional Isoforms of CX3CR1 Are Potent HIV Coreceptors. The Journal of Immunology, 171, 5305-5312.

Garton, K. J., Gough, P. J., Blobel, C. P., Murphy, G., Greaves, D. R., Dempsey, P. J. & Raines, E. W. (2001) Tumor necrosis factor-alpha-converting enzyme (ADAM17) mediates the cleavage and shedding of fractalkine (CX3CL1). Journal of Biological Chemistry, 276, 37993-8001.

Garza, A., Vega, R. & Soto, E. (2006) Cellular mechanisms of lead neurotoxicity. Medical Science Monitor, 12, 57-65.

Gehrmann, J., Banati, R. B. & Kreutzberg, G. W. (1993) Microglia in the immune surveillance of the brain: human microglia constitutively express HLA-DR molecules. Journal of Neuroimmunology, 48, 189-198.

Geissmann, F., Jung, S. & Littman, D. R. (2003) Blood Monocytes Consist of Two Principal Subsets with Distinct Migratory Properties. Immunity, 19, 71-82.

Gemma, C., Bachstetter, A. D. & Bickford, P. C. (2010) Neuron-Microglia Dialogue and Hippocampal Neurogenesis in the Aged Brain. Aging and Diesease, 1, 232-244.

Gidlow, D. A. (2004) Lead toxicity. Occupational Medicine, 54, 76-81. Gilbert, M. E., Kelly, M. E., Samsam, T. E. & Goodman, J. H. (2005) Chronic develometal lead

exposure reduces neurogenesis in adult rat hippocampus but doesn't impair spatial learning. Toxicological Sciences, 86, 365-374.

Gillis, B., Arbieva, Z. & Gavin, I. (2012) Analysis of lead toxicity in human cells. BMC Genomics, 13, 344.

Page 190: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[169]

Ginhoux, F., Greter, M., Leboeuf, M., Nandi, S., See, P., Gokhan, S., Mehler, M. F., Conway, S. J., Ng, L. G., Stanley, E. R., Samokhvalov, I. M. & Merad, M. (2010) Fate Mapping Analysis Reveals That Adult Microglia Derive from Primitive Macrophages. Science, 330, 841-845.

Giulian, D. & Baker, T. J. (1986) Characterization of ameboid microglia isolated from developing mammalian brain. Journal of Neuroscience, 6, 2163-2178.

Glabinski, A. R., Bielecki, B., O'Bryant, S., Selmaj, K. & Ransohoff, R. M. (2002) Experimental autoimmune encephalomyelitis: CC chemokine receptor expression by trafficking cells. Journal of Autoimmunity, 19, 175-181.

Goczalik, I., Ulbricht, E., Hollborn, M., Raap, M., Uhlmann, S., Weick, M., Pannicke, T., Wiedemann, P., Bringmann, A., Reichenbach, A. & Francke, M. (2008) Expression of CXCL8, CXCR1, and CXCR2 in neurons and glial cells of the human and rabbit retina. Investigative Ophthalmology and Visual Science, 49, 4578-4589.

Godbout, J. P. & Johnson, R. W. (2004) Interleukin-6 in the aging brain. Journal of Neuroimmunology, 147, 141-144.

Godefroy, D., Gosselin, R. D., Yasutake, A., Fujimura, M., Combadiere, C., Maury-Brachet, R., Laclau, M., Rakwal, R., Melik-Parsadaniantz, S., Bourdineaud, J. P. & Rostene, W. (2012) The chemokine CCL2 protects against methylmercury neurotoxicity. Toxicological Sciences, 125, 209-218.

Goldstein, G. W. (1990) Lead poisoning and brain cell function. Environmental Health Perspectives, 89, 91-94.

Gomi, M., Aoki, T., Takagi, Y., Nishimura, M., Ohsugi, Y., Mihara, M., Nozaki, K., Hashimoto, N., Miyamoto, S. & Takahashi, J. (2011) Single and local blockade of interleukin-6 signaling promotes neuronal differentiation from transplanted embryonic stem cell-derived neural precursor cells. Journal of Neuroscience Research, 89, 1388-1399.

Gonzalez-Perez, O., Quinones-Hinojosa, A. & Garcia-Verdugo, J. M. (2010) Immunological control of adult neural stem cells. Journal of Stem Cells, 5, 23-31.

Goyer, R. A.& Clarsksom, W. T. (2001). Toxic effects of metals. In: C.D. Klaassen (Ed.), Casarett and Doull’s Toxicology. The Basic Science of Poisons, McGraw-Hill, NY. Guha, D., Nagilla, P., Redinger, C., Srinivasan, A., Schatten, G. & Ayyavoo, V. (2012) Neuronal

apoptosis by HIV-1 Vpr: contribution of proinflammatory molecular networks from infected target cells. Journal of Neuroinflammation, 9, 138.

Guilarte, T. R., Miceli, R. C. & Jett, D. A. (1994) Neurochemical aspects of hippocampal and cortical Pb2+ neurotoxicity. Neurotoxicology, 15, 459-466.

Guillemin, G. J. & Brew, B. J. (2004) Microglia, macrophages, perivascular macrophages, and pericytes: a review of function and identification. Journal of Leukocyte Biology, 75, 388-397.

Guo, J., Walss-Bass, C. & Luduena, R. F. (2010) The beta isotypes of tubulin in neuronal differentiation. Cytoskeleton (Hoboken), 67, 431-441.

Gurer, H. & Ercal, N. (2000) Can antioxidants be beneficial in the treatment of lead poisoning? Free Radical Biology and Medicine, 29, 927 - 945.

Gwaltney-Brant, S. M. & Rumbeiha, W. K. (2002) Newer antidotal therapies. Veterinary Clinics of North America. Small Animal Practice, 32, 323-339.

Halliwell, B. (1992) Reactive Oxygen Species and the Central Nervous System. Journal of Neurochemistry, 59, 1609-1623.

Hama, T., Kushima, Y., Miyamoto, M., Kubota, M., Takei, N. & Hatanaka, H. (1991) Interleukin-6 improves the survival of mesencephalic catecholaminergic and septal cholinergic neurons from postnatal, two-week-old rats in cultures. Neuroscience, 40, 445-452.

Hanas, J. S., Rodgers, J. S., Bantle, J. A. & Cheng, Y. G. (1999) Lead inhibition of DNA-binding mechanism of Cys(2)His(2) zinc finger proteins. Molecular Pharmacology, 56, 982-988.

Hanisch, U. K. & Kettenmann, H. (2007) Microglia: active sensor and versatile effector cells in the normal and pathologic brain. Nature Neuroscience, 10, 1387-1394.

Hanisch, U. K., Prinz, M., Angstwurm, K., Hausler, K. G., Kann, O., Kettenmann, H. & Weber, J. R. (2001) The protein tyrosine kinase inhibitor AG126 prevents the massive microglial cytokine induction by pneumococcal cell walls. European Journal of Immunology, 31, 2104-2115.

Hanisch, U.-K. (2002) Microglia as a source and target of cytokines. Glia, 40, 140-155.

Page 191: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[170]

Hansson, E. & Rönnbäck, L. (2003) Glial neuronal signaling in the central nervous system. The FASEB Journal, 17, 341-348.

Hatori, K., Nagai, A., Heisel, R., Ryu, J. K. & Kim, S. U. (2002) Fractalkine and fractalkine receptors in human neurons and glial cells. Journal of Neuroscience Research, 69, 418-426.

Harrison, J. K., Jiang, Y., Chen, S., Xia, Y., Maciejewski, D., McNamara, R. K., Streit, W. J., Salafranca, M. N., Adhikari, S., Thompson, D. A., Botti, P., Bacon, K. B. & Feng, L. (1998) Role for neuronally derived fractalkine in mediating interactions between neurons and CX3CR1-expressing microglia. Proceedings of the National Academy of Sciences of the United States of America, 95, 10896-10901.

Health, Q. 2008. A report into the results of blood-lead screening program of 1- 4-year-oldchildren in Mt Isa, Queensland.

Heese, K., Hock, C. & Otten, U. (1998) Inflammatory Signals Induce Neurotrophin Expression in Human Microglial Cells. Journal of Neurochemistry, 70, 699-707.

Hendrix, S. & Nitsch, R. (2007) The role of T helper cells in neuroprotection and regeneration. Journal of Neuroimmunology, 184, 100-112.

Heneka, M., O’Banion, M. K., Terwel, D. & Kummer, M. (2010) Neuroinflammatory processes in Alzheimer’s disease. Journal of Neural Transmission, 117, 919-947.

Hernberg, S. (2000) Lead poisoning in a historical perspective. American Journal of Industrial Medicine, 38, 244-254.

Hesselgesser, J. & Horuk, R. (1999) Chemokine and chemokine receptor expression in the central nervous system. Journal of Neurovirology, 5, 13-26.

Hinojosa, A. E., Garcia-Bueno, B., Leza, J. C. & Madrigal, J. L. (2011) CCL2/MCP-1 modulation of microglial activation and proliferation. Journal of Neuroinflammation, 8, 77-78.

Hirt, U. A. & Leist, M. (2003) Rapid, noninflammatory and PS-dependent phagocytic clearance of necrotic cells. Cell Death and Differentiation, 10, 1156-1164.

Holtzman, D., Olson, J. E., DeVries, C. & Bensch, K. (1987) Lead toxicity in primary cultured cerebral astrocytes and cerebellar granular neurons. Toxicology and Applied Pharmacology, 89, 211-225.

Hooper, C., Pinteaux-Jones, F., Fry, V. A. H., Sevastou, I. G., Baker, D., Heales, S. J. & Pocock, J. M.

(2009) Differential effects of albumin on microglia and macrophages; implications for neurodegeneration following blood–brain barrier damage. Journal of Neurochemistry, 109, 694-705.

Hopkins, S. J. & Rothwell, N. J. (1995) Cytokines and the nervous system. I: Expression and recognition. Trends in Neurosciences, 18, 83-88.

Howell, O. W., Rundle, J. L., Garg, A., Komada, M., Brophy, P. J. & Reynolds, R. (2010) Activated Microglia Mediate Axoglial Disruption That Contributes to Axonal Injury in Multiple Sclerosis. Journal of Neuropathology and Experimental Neurology, 69, 1017-1033.

Horuk, R., Martin, A. W., Wang, Z., Schweitzer, L., Gerassimides, A., Guo, H., Lu, Z., Hesselgesser, J., Perez, H. D., Kim, J., Parker, J., Hadley, T. J. & Peiper, S. C. (1997) Expression of chemokine receptors by subsets of neurons in the central nervous system. The Journal of Immunology, 158, 2882-2890.

Hu, Q., Fu, H., Ren, T., Wang, S., Zhou, W., Song, H., Han, Y. & Dong, S. (2008) Maternal low-level lead exposure reduces the expression of PSA-NCAM and the activity of sialyltransferase in the hippocampi of neonatal rat pups. Neurotoxicology, 29, 675-681.

Hu, Q., Fu, H., Song, H., Ren, T., Li, L., Ye, L., Liu, T. & Dong, S. (2011) Low-level lead exposure attenuates the expression of three major isoforms of neural cell adhesion molecule. Neurotoxicology, 32, 255-260.

Huang, F. & Schneider, J. S. (2004) Effects of lead exposure on proliferation and differentiation of neural stem cells derived from different regions of embryonic rat brain. Neurotoxicology, 25, 1001-1012.

Huber, J. D., Campos, C. R., Mark, K. S. & Davis, T. P. (2006) Alterations in blood-brain barrier ICAM-1 expression and brain microglial activation after λ-carrageenan-induced inflammatory pain. American Journal of Physiology - Heart and Circulatory Physiology, 290, H732-H740.

Page 192: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[171]

Hundhausen, C., Misztela, D., Berkhout, T. A., Broadway, N., Saftig, P., Reiss, K., Hartmann, D., Fahrenholz, F., Postina, R., Matthews, V., Kallen, K. J., Rose-John, S. & Ludwig, A. (2003) The disintegrin-like metalloproteinase ADAM10 is involved in constitutive cleavage of CX3CL1 (fractalkine) and regulates CX3CL1-mediated cell-cell adhesion. Blood, 102, 1186-1195.

Ihara, S., Iwamatsu, A., Fujiyoshi, T., Komi, A., Yamori, T. & Fukui, Y. (1996) Identification of interleukin-6 as a factor that induces neurite outgrowth by PC12 cells primed with NGF. Journal of Biochemistry, 120, 865-868.

Imai, T., Hieshima, K., Haskell, C., Baba, M., Nagira, M., Nishimura, M., Kakizaki, M., Takagi, S., Nomiyama, H., Schall, T. J. & Yoshie, O. (1997) Identification and molecular characterization of fractalkine receptor CX3CR1, which mediates both leukocyte migration and adhesion. Cell, 91, 521-30.

Ishida, Y., Hayashi, T., Goto, T., Kimura, A., Akimoto, S., Mukaida, N. & Kondo, T. (2008) Essential involvement of CX3CR1-mediated signals in the bactericidal host defense during septic peritonitis. Journal of Immunology, 181, 4208-4218.

Islam, O., Gong, X., Rose-John, S. & Heese, K. (2009) Interleukin-6 and neural stem cells: more than gliogenesis. Molecular Biology of the Cell, 20, 188-199.

Izikson, L., Klein, R. S., Charo, I. F., Weiner, H. L. & Luster, A. D. (2000) Resistance to Experimental Autoimmune Encephalomyelitis in Mice Lacking the Cc Chemokine Receptor (Ccr2). The Journal of Experimental Medicine, 192, 1075-1080.

Jack, C. S., Arbour, N., Manusow, J., Montgrain, V., Blain, M., McCrea, E., Shapiro, A. & Antel, J. P. (2005) TLR Signaling Tailors Innate Immune Responses in Human Microglia and Astrocytes. The Journal of Immunology, 175, 4320-4330.

Jaerve, A. & Müller, H. (2012) Chemokines in CNS injury and repair. Cell and Tissue Research, 349, 229-248.

Janabi, N., Peudenier, S., Héron, B., Ng, K. H. & Tardieu, M. (1995) Establishment of human microglial cell lines after transfection of primary cultures of embryonic microglial cells with the SV40 large T antigen. Neuroscience Letters, 195, 105-108.

Ji, J. F., He, B. P., Dheen, S. T. & Tay, S. S. W. (2004) Expression of chemokine receptors CXCR4, CCR2, CCR5 and CX3CR1 in neural progenitor cells isolated from the subventricular zone of the adult rat brain. Neuroscience Letters, 355, 236-240.

Jiang, Y. M., Long, L. L., Zhu, X. Y., Zheng, H., Fu, X., Ou, S. Y., Wei, D. L., Zhou, H. L. & Zheng, W. (2008) Evidence for altered hippocampal volume and brain metabolites in workers occupationally exposed to lead: a study by magnetic resonance imaging and (1)H magnetic resonance spectroscopy. Toxicology Letters, 181, 118-125.

Jiang, Y. Q. & Oblinger, M. M. (1992) Differential regulation of beta III and other tubulin genes during peripheral and central neuron development. Journal of Cell Science, 103, 643-651.

Jones, B. A., Beamer, M. & Ahmed, S. (2010) Fractalkine/CX3CL1: a potential new target for inflammatory diseases. Molecular Interventions, 10, 263-270.

Jones, R. S., Minogue, A. M., Connor, T. J. & Lynch, M. A. (2012) Amyloid-beta-Induced Astrocytic Phagocytosis is Mediated by CD36, CD47 and RAGE. Journal Neuroimmune Pharmacology, 8, 301-311.

Jung, D. Y., Lee, H., Jung, B.-Y., Ock, J., Lee, M.-S., Lee, W.-H. & Suk, K. (2005) TLR4, but Not TLR2, Signals Autoregulatory Apoptosis of Cultured Microglia: A Critical Role of IFN-β as a Decision Maker. The Journal of Immunology, 174, 6467-6476.

Juttler, E., Tarabin, V. & Schwaninger, M. (2002) Interleukin-6 (IL-6): A Possible Neuromodulator Induced by Neuronal Activity. The Neuroscientist, 8, 268-275.

Kaindl, A. M., Degos, V., Peineau, S., Gouadon, E., Chhor, V., Loron, G., Le Charpentier, T., Josserand, J., Ali, C., Vivien, D., Collingridge, G. L., Lombet, A., Issa, L., Rene, F., Loeffler, J. P., Kavelaars, A., Verney, C., Mantz, J. & Gressens, P. (2012) Activation of microglial N-methyl-D-aspartate receptors triggers inflammation and neuronal cell death in the developing and mature brain. Annals of Neurology, 72, 536-549.

Kakimura, J., Kitamura, Y., Takata, K., Umeki, M., Suzuki, S., Shibagaki, K., Taniguchi, T., Nomura, Y., Gebicke-Haerter, P. J., Smith, M. A., Perry, G. & Shimohama, S. (2002) Microglial activation and amyloid-beta clearance induced by exogenous heat-shock proteins. FASEB Journal, 16, 601-603.

Page 193: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[172]

Kalla, R., Bohatschek, M., Kloss, C. U. A., Krol, J., Von Maltzan, X. & Raivich, G. (2003) Loss of microglial ramification in microglia-astrocyte cocultures: Involvement of adenylate cyclase, calcium, phosphatase, and Gi-protein systems. Glia, 41, 50-63.

Kalla, R., Liu, Z., Xu, S., Koppius, A., Imai, Y., Kloss, C. U. A., Kohsaka, S., Gschwendtner, A., Möller, J. C., Werner, A. & Raivich, G. (2001) Microglia and the early phase of immune surveillance in the axotomized facial motor nucleus: Impaired microglial activation and lymphocyte recruitment but no effect on neuronal survival or axonal regeneration in macrophage-colony stimulating factor-deficient mice. The Journal of Comparative Neurology, 436, 182-201.

Kamel, F., Umbach, D. M., Munsat, T. L., Shefner, J. M., Hu, H. & Sandler, D. P. (2002) Lead Exposure and Amyotrophic Lateral Sclerosis. Epidemiology, 13, 311-319.

Kanwal, S. K. & Kumar, V. (2011) High prenatal and postnatal lead exposure associated lead encephalopathy in an infant. Indian Journal of Pediatrics, 78, 1420-1423.

Kauppinen, T. M., Higashi, Y., Suh, S. W., Escartin, C., Nagasawa, K. & Swanson, R. A. (2008) Zinc triggers microglial activation. Journal of Neuroscience, 28, 5827-5835.

Karimooy, H. N., Mood, M. B., Hosseini, M. & Shadmanfar, S. (2010) Effects of occupational lead exposure on renal and nervous system of workers of traditional tile factories in Mashhad (northeast of Iran). Toxicology and Industrial Health, 26, 633-638.

Kasten-Jolly, J., Heo, Y. & Lawrence, D. A. (2011) Central nervous system cytokine gene expression: modulation by lead. Journal of Biochemical and Molecular Toxicology, 25, 41-54.

Kasten-Jolly, J., Pabello, N., Bolivar, V. J. & Lawrence, D. A. (2012) Developmental lead effects on behavior and brain gene expression in male and female BALB/cAnNTac mice. Neurotoxicology, 33, 1005-1020.

Kaul, M., Ma, Q., Medders, K. E., Desai, M. K. & Lipton, S. A. (2007) HIV-1 coreceptors CCR5 and CXCR4 both mediate neuronal cell death but CCR5 paradoxically can also contribute to protection. Cell Death and Differentiation, 14, 296-305.

Kawai, T. & Akira, S. (2011) Toll-like receptors and their crosstalk with other innate receptors in infection and immunity. Immunity, 34, 637-650.

Keblesh, J. P., Dou, H., Gendelman, H. E. & Xiong, H. (2009) 4-Aminopyridine improves spatial memory in a murine model of HIV-1 encephalitis. Journal of Neuroimmune Pharmacology, 4, 317-327.

Kelder, W., McArthur, J., Nance-Sproson, T., McClernon, D. & Griffin, D. (1998) Beta-chemokines MCP-1 and RANTES are selectively increased in cerebrospinal fluid of patients with human immunodeficiency virus-associated dementia. Annals of Neurology, 44, 831 - 835.

Keohane, A., Ryan, S., Maloney, E., Sullivan, A. M. & Nolan, Y. M. (2010) Tumour necrosis factor-alpha impairs neuronal differentiation but not proliferation of hippocampal neural precursor cells: Role of Hes1. Molecular and Cellular Neurosciences, 43, 127-135.

Kerkovich, D. M., Sapp, D., Weidenheim, K., Brosnan, C. F., Pfeiffer, S. E., Yeh, H. H. & Busciglio, J. (1999) Fetal human cortical neurons grown in culture: morphological differentiation, biochemical correlates and development of electrical activity. International Journal of Developmental Neuroscience, 17, 347-356.

Kern, M., Audesirk, T. & Audesirk, G. (1993) Effects of inorganic lead on the differentiation and growth of cortical neurons in culture. Neurotoxicology, 14, 319-327.

Kettenmann, H., Hanisch, U., Noda, M. & Verkhratsky, A. (2011) Physiology of microglia. Physiological Reviews, 91, 461 - 553.

Khalil, N., Morrow, L. A., Needleman, H., Talbott, E. O., Wilson, J. W. & Cauley, J. A. (2009) Association of cumulative lead and neurocognitive function in an occupational cohort. Neuropsychology, 23, 10-19.

Kim, S. U. & de Vellis, J. (2005) Microglia in health and disease. Journal of Neuroscience Research, 81, 302-313.

Kim, W.-G., Mohney, R. P., Wilson, B., Jeohn, G.-H., Liu, B. & Hong, J.-S. (2000) Regional Difference in Susceptibility to Lipopolysaccharide-Induced Neurotoxicity in the Rat Brain: Role of Microglia. The Journal of Neuroscience, 20, 6309-6316.

Kizil, C., Dudczig, S., Kyritsis, N., Machate, A., Blaesche, J., Kroehne, V. & Brand, M. (2012) The chemokine receptor cxcr5 regulates the regenerative neurogenesis response in the adult zebrafish brain. Neural Devevlopment, 7, 27-40.

Page 194: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[173]

Kloss, C. U. A., Bohatschek, M., Kreutzberg, G. W. & Raivich, G. (2001) Effect of Lipopolysaccharide on the Morphology and Integrin Immunoreactivity of Ramified Microglia in the Mouse Brain and in Cell Culture. Experimental Neurology, 168, 32-46.

Kofler, J. & Wiley, C. A. (2011) Microglia: key innate immune cells of the brain. Toxicologic Pathology, 39, 103-114.

Koizumi, S., Shigemoto-Mogami, Y., Nasu-Tada, K., Shinozaki, Y., Ohsawa, K., Tsuda, M., Joshi, B. V., Jacobson, K. A., Kohsaka, S. & Inoue, K. (2007) UDP acting at P2Y6 receptors is a mediator of microglial phagocytosis. Nature, 446, 1091-1095.

Koller, K., Brown, T., Spurgeon, A. & Levy, L. (2004) Recent developments in low-level lead exposure and intellectual impairment in children. Environmental Health Perspectives, 112, 987-994.

Koller, k., Levy, L. & Brown, T. (2005) Low-Level Lead Exposure and Intellectual Impairment in Children: Koller et al. Respond. Environmental Health Perspectives, 113, A16-A17.

Krathwohl, M. D. & Kaiser, J. L. (2004) Chemokines Promote Quiescence and Survival of Human Neural Progenitor Cells. Stem Cells, 22, 109-118.

Kruger, C., Laage, R., Pitzer, C., Schabitz, W.-R. & Schneider, A. (2007) The hematopoietic factor GM-CSF (Granulocyte-macrophage colony-stimulating factor) promotes neuronal differentiation of adult neural stem cells in vitro. BMC Neuroscience, 8, 88-95.

Kreutzberg, G. W. (1995) Microglia, the first line of defence in brain pathologies. Arzneimittel-Forschung, 45, 357-360.

Kuhlmann, A. C., McGlothan, J. L. & Guilarte, T. R. (1997) Developmental lead exposure causes spatial learning deficits in adult rats. Neuroscience Letters, 233, 101-114.

Lachyankar, M. B., Condon, P. J., Quesenberry, P. J., Litofsky, N. S., Recht, L. D. & Ross, A. H. (1997) Embryonic precursor cells that express Trk receptors: induction of different cell fates by NGF, BDNF, NT-3, and CNTF. Experimental Neurology, 144, 350-360.

Laflamme, N., Echchannaoui, H., Landmann, R. & Rivest, S. (2003) Cooperation between toll-like receptor 2 and 4 in the brain of mice challenged with cell wall components derived from gram-negative and gram-positive bacteria. European Journal of Immunology, 33, 1127-1138.

Lambert, C., Desbarats, J., Arbour, N., Hall, J. A., Olivier, A., Bar-Or, A. & Antel, J. P. (2008) Dendritic Cell Differentiation Signals Induce Anti-Inflammatory Properties in Human Adult Microglia. The Journal of Immunology, 181, 8288-8297.

Lambertsen, K. L., Clausen, B. H., Babcock, A. A., Gregersen, R., Fenger, C., Nielsen, H. H., Haugaard, L. S., Wirenfeldt, M., Nielsen, M., Dagnaes-Hansen, F., Bluethmann, H., Færgeman, N. J., Meldgaard, M., Deierborg, T. & Finsen, B. (2009) Microglia Protect Neurons against Ischemia by Synthesis of Tumor Necrosis Factor. The Journal of Neuroscience, 29, 1319-1330.

Lan, X., Chen, Q., Wang, Y., Jia, B., Sun, L., Zheng, J. & Peng, H. (2012) TNF-alpha Affects Human Cortical Neural Progenitor Cell Differentiation through the Autocrine Secretion of Leukemia Inhibitory Factor. PLoS ONE, 7(12): e50783. doi:10.1371/journal.pone.0050783

Lanphear, B. P., Dietrich, K., Auinger, P. & Cox, C. (2000) Cognitive deficits associated with blood lead concentrations <10 microg/dL in US children and adolescents. Public Health Reports, 115, 521-9.

Lauro, C., Catalano, M., Trettel, F., Mainiero, F., Ciotti, M. T., Eusebi, F. & Limatola, C. (2006) The Chemokine CX3CL1 Reduces Migration and Increases Adhesion of Neurons with Mechanisms Dependent on the β1 Integrin Subunit. The Journal of Immunology, 177, 7599-7606.

Lavery, T. J., Butterfield, N., Kemper, C. M., Reid, R. J. & Sanderson, K. (2008) Metals and selenium in the liver and bone of three dolphin species from South Australia, 1988-2004. Science of the Total Environment, 390, 77-85.

Lawrence, D. A. (1981) In vivo and in vitro effects of lead on humoral and cell-mediated immunity. Infection and Immunity, 31, 136-143.

Lawrence, D. A. & McCabe Jr, M. J. (2002) Immunomodulation by metals. International Immunopharmacology, 2, 293-302.

Lawson, L. J., Perry, V. H., Dri, P. & Gordon, S. (1990) Heterogeneity in the distribution and morphology of microglia in the normal adult mouse brain. Neuroscience, 39, 151-170.

Page 195: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[174]

Lazarini, F., Gabellec, M.-M., Torquet, N. & Lledo, P.-M. (2012) Early Activation of Microglia Triggers Long-Lasting Impairment of Adult Neurogenesis in the Olfactory Bulb. The Journal of Neuroscience, 32, 3652-3664.

Lee, M., McGeer, E. & McGeer, P. L. (2013) Neurotoxins released from interferon-gamma-stimulated human astrocytes. Neuroscience, 229, 164-175.

Lee, S., Varvel, N. H., Konerth, M. E., Xu, G., Cardona, A. E., Ransohoff, R. M. & Lamb, B. T. (2010) CX3CR1 Deficiency Alters Microglial Activation and Reduces Beta-Amyloid Deposition in Two Alzheimer's Disease Mouse Models. The American journal of pathology, 177, 2549-2562.

Leonard, E. J., Skeel, A. & Yoshimura, T. (1991) Biological aspects of monocyte chemoattractant protein-1 (MCP-1). Advances in Experimental Medicine and Biology, 305, 57-64.

Leone, C., Le Pavec, G., Même, W., Porcheray, F., Samah, B., Dormont, D. & Gras, G. (2006) Characterization of human monocyte-derived microglia-like cells. Glia, 54, 183-192.

Li, B., Bedard, K., Sorce, S., Hinz, B., Dubois-Dauphin, M. & Krause, K. H. (2009) NOX4 Expression in Human Microglia Leads to Constitutive Generation of Reactive Oxygen Species and to Constitutive IL-6 Expression. Journal of Innate Immunity, 1, 570-581.

Lidsky, T. I. & Schneider, J. S. (2003) Lead neurotoxicity in children: basic mechanisms and clinical correlates. Brain, 126, 5-19.

Liu, G., Huang, W., Moir, R. D., Vanderburg, C. R., Lai, B., Peng, Z., Tanzi, R. E., Rogers, J. T. & Huang, X. (2006) Metal exposure and Alzheimer’s pathogenesis. Journal of Structural Biology, 155, 45-51.

Liu, J., Han, D., Li, Y., Zheng, L., Gu, C., Piao, Z., Au, W. W., Xu, X. & Huo, X. (2010) Lead affects apoptosis and related gene XIAP and Smac expression in the hippocampus of developing rats. Neurochemical Research, 35, 473-9.

Liu, M.-C., Liu, X.-Q., Wang, W., Shen, X.-F., Che, H.-L., Guo, Y.-Y., Zhao, M.-G., Chen, J.-Y. & Luo, W.-J. (2012) Involvement of Microglia Activation in the Lead Induced Long-Term Potentiation Impairment. PLoS ONE, 7, e43924.

Liu, X. S., Zhang, Z. G., Zhang, R. L., Gregg, S. R., Wang, L., Yier, T. & Chopp, M. (2007) Chemokine ligand 2 (CCL2) induces migration and differentiation of subventricular zone cells after stroke. Journal of Neuroscience Research, 85, 2120-2125.

Liu, Y., Walter, S., Stagi, M., Cherny, D., Letiembre, M., Schulz-Schaeffer, W., Heine, H., Penke, B., Neumann, H. & Fassbender, K. (2005) LPS receptor (CD14): a receptor for phagocytosis of Alzheimer's amyloid peptide. Brain, 128, 1778-89.

Loddick, S. A., Turnbull, A. V. & Rothwell, N. J. (1998) Cerebral interleukin-6 is neuroprotective during permanent focal cerebral ischemia in the rat. Journal of Cerebral Blood Flow and Metabolism, 18, 176-9.

Losy, J. & Zaremba, J. (2001) Monocyte chemoattractant protein-1 is increased in the cerebrospinal fluid of patients with ischemic stroke. Stroke, 32, 2695-6.

Lucas, S. M., Rothwell, N. J. & Gibson, R. M. (2006) The role of inflammation in CNS injury and disease. British Journal of Pharmacology, 147, S232-240.

Lucchini, R. G., Zoni, S., Guazzetti, S., Bontempi, E., Micheletti, S., Broberg, K., Parrinello, G. & Smith, D. R. (2012) Inverse association of intellectual function with very low blood lead but not with manganese exposure in Italian adolescents. Environmental Research, 118, 65-71.

Luna, A. L., Acosta-Saavedra, L. C., Martínez, M., Torres-Avilés, N., Gómez, R. & Calderón-Aranda, E. S. (2012) TLR4 is a target of environmentally relevant concentration of lead. Toxicology Letters, 214, 301-306.

Luo, W., Ruan, D., Yan, C., Yin, S. & Chen, J. (2012) Effects of chronic lead exposure on functions of nervous system in Chinese children and developmental rats. Neurotoxicology, 33, 862-71.

Lutz, M. B., Suri, R. M., Niimi, M., Ogilvie, A. L., Kukutsch, N. A., Rossner, S., Schuler, G. & Austyn, J. M. (2000) Immature dendritic cells generated with low doses of GM-CSF in the absence of IL-4 are maturation resistant and prolong allograft survival in vivo. European Journal of Immunology, 30, 1813-22.

Lyons, A., Lynch, A. M., Downer, E. J., Hanley, R., O'Sullivan, J. B., Smith, A. & Lynch, M. A. (2009) Fractalkine-induced activation of the phosphatidylinositol-3 kinase pathway attentuates microglial activation in vivo and in vitro. Journal of Neurochemistry, 110, 1547-1556.

Page 196: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[175]

Ma, N & Streilein, J.W. (1999) T cell immunity induced by allogeneic microglia in relation to neuronal retina transplantation. Journal of Immunology, 162, 4482-4489.

Magnus, T., Chan, A., Grauer, O., Toyka, K. V. & Gold, R. (2001) Microglial phagocytosis of apoptotic inflammatory T cells leads to down-regulation of microglial immune activation. Journal of Immunology, 167, 5004-5010.

Maisonpierre, P. C., Belluscio, L., Friedman, B., Alderson, R. F., Wiegand, S. J., Furth, M. E., Lindsay, R. M. & Yancopoulos, G. D. (1990) NT-3, BDNF, and NGF in the developing rat nervous system: Parallel as well as reciprocal patterns of expression. Neuron, 5, 501-509.

Marchetti, C. (2003) Molecular targets of lead in brain neurotoxicity. Neurotoxicity Research, 5, 221 - 236.

Markowitz, M. (2000) Lead poisoning. Pediatrics in Review, 21, 327-335. Marz, P., Herget, T., Lang, E., Otten, U. & Rose-John, S. (1997) Activation of gp130 by IL-

6/soluble IL-6 receptor induces neuronal differentiation. European Journal of Neuroscience, 9, 2765-2773.

Masliah, E., Mallory, M., Hansen, L., Alford, M., Albright, T., Terry, R., Shapiro, P., Sundsmo, M. & Saitoh, T. (1991) Immunoreactivity of CD45, a protein phosphotyrosine phosphatase, in Alzheimer's disease. Acta Neuropathology, 83, 12-20.

Mathias, C., Nathalie, L., Simon, C. & Denis, V. (2004) Cytokines in Neuroinflammation and Alzheimers Disease. Current Drug Targets, 5, 529-534.

Matsuda, S., Wen, T. C., Morita, F., Otsuka, H., Igase, K., Yoshimura, H. & Sakanaka, M. (1996) Interleukin-6 prevents ischemia-induced learning disability and neuronal and synaptic loss in gerbils. Neuroscience Letters, 204, 109-112.

McCabe, M. J., Jr. & Lawrence, D. A. (1990) The heavy metal lead exhibits B cell-stimulatory factor activity by enhancing B cell Ia expression and differentiation. Journal of Immunology, 145, 671-677.

McCabe, M. J., Jr., Singh, K. P. & Reiners, J. J., Jr. (2001) Low level lead exposure in vitro stimulates the proliferation and expansion of alloantigen-reactive CD4(high) T cells. Toxicology and Applied Pharmacology, 177, 219-231.

McGeer, P. L. & McGeer, E. G. (2002) Local Neuroinflammation and the Progression of Alzheimer's Disease. Journal of Neurovirology, 8, 529-538.

Meagher, J., Zellweger, R. & Filgueira, L. (2005) Functional Dissociation of the Basolateral Transcytotic Compartment from the Apical Phago-lysosomal Compartment in Human Osteoclasts. Journal of Histochemistry and Cytochemistry, 53, 665-670.

Mennicken, F., Maki, R., de Souza, E. B. & Quirion, R. (1999) Chemokines and chemokine receptors in the CNS: a possible role in neuroinflammation and patterning. Trends in Pharmacological Sciences, 20, 73-78.

Mestas, J. & Hughes, C. C. W. (2004) Of Mice and Not Men: Differences between Mouse and Human Immunology. The Journal of Immunology, 172, 2731-2738.

Meucci, O., Fatatis, A., Simen, A. A. & Miller, R. J. (2000) Expression of CX3CR1 chemokine receptors on neurons and their role in neuronal survival. Proceedings of the National Academy of Sciences, 97, 8075-8080.

Mildner, A., Mack, M., Schmidt, H., Brück, W., Djukic, M., Zabel, M. D., Hille, A., Priller, J. & Prinz, M. (2009) CCR2+Ly-6Chi monocytes are crucial for the effector phase of autoimmunity in the central nervous system. Brain, 132, 2487-2500.

Miller, R. J., Rostene, W., Apartis, E., Banisadr, G., Biber, K., Milligan, E. D., White, F. A. & Zhang, J. (2008) Chemokine action in the nervous system. Journal of Neuroscience, 28, 11792-11795.

Minami, S. S., Sun, B., Popat, K., Kauppinen, T., Pleiss, M., Zhou, Y., Ward, M. E., Floreancig, P., Mucke, L., Desai, T. & Gan, L. (2012) Selective targeting of microglia by quantum dots. Journal of Neuroinflammation, 9, 22.

Mines, M., Ding, Y. & Fan, G. H. (2007) The many roles of chemokine receptors in neurodegenerative disorders: emerging new therapeutical strategies. Current Medicinal Chemistry, 14, 2456-70.

Mittelbronn, M., Dietz, K., Schluesener, H. J. & Meyermann, R. (2001) Local distribution of microglia in the normal adult human central nervous system differs by up to one order of magnitude. Acta Neuropathology, 101, 249-255.

Page 197: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[176]

Mizuno, T., Kawanokuchi, J., Numata, K. & Suzumura, A. (2003) Production and neuroprotective functions of fractalkine in the central nervous system. Brain Research, 979, 65-70.

Modi, W. S. & Yoshimura, T. (1999) Isolation of novel GRO genes and a phylogenetic analysis of the CXC chemokine subfamily in mammals. Molecular Biology and Evolution, 16, 180-193.

Monnet-Tschudi, F., Zurich, M. G., Boschat, C., Corbaz, A. & Honegger, P. (2006) Involvement of environmental mercury and lead in the etiology of neurodegenerative diseases. Reviews on Environmental Health, 21, 105-117.

Monje, M. L., Toda, H. & Palmer, T. D. (2003) Inflammatory Blockade Restores Adult Hippocampal Neurogenesis. Science, 302, 1760-1765.

Morales, I., Farias, G. & Maccioni, R. B. (2010) Neuroimmunomodulation in the pathogenesis of Alzheimer's disease. Neuroimmunomodulation, 17, 202-204.

Moreira, P. I., Smith, M. A., Zhu, X., Nunomura, A., Castellani, R. J. & Perry, G. (2005) Oxidative Stress and Neurodegeneration. Annals of the New York Academy of Sciences, 1043, 545-552.

Morgan, S. C., Taylor, D. L. & Pocock, J. M. (2004) Microglia release activators of neuronal proliferation mediated by activation of mitogen-activated protein kinase, phosphatidylinositol-3-kinase/Akt and delta-Notch signalling cascades. Journal of Neurochemistry, 90, 89-101.

Morrens, J., Van Den Broeck, W. & Kempermann, G. (2012) Glial cells in adult neurogenesis. Glia, 60, 159-174.

Mou, Y. H., Yang, J. Y., Cui, N., Wang, J. M., Hou, Y., Song, S. & Wu, C. F. (2012) Effects of cobalt chloride on nitric oxide and cytokines/chemokines production in microglia. International Immunopharmacology, 13, 120-125.

Mrak, R. E. & Griffin, W. S. (2005) Glia and their cytokines in progression of neurodegeneration. Neurobiology of Aging, 26, 349-354.

Murphy, K. J. & Regan, C. M. (1999) Low-Level Lead Exposure in the Early Postnatal Period Results in Persisting Neuroplastic Deficits Associated with Memory Consolidation. Journal of Neurochemistry, 72, 2099-2104.

Nadal, A., Fuentes, E., Pastor, J. & McNaughton, P. A. (1995) Plasma albumin is a potent trigger of calcium signals and DNA synthesis in astrocytes. Proceedings of the National Academy of Sciences, 92, 1426-1430.

Nagai, A., Mishima, S., Ishida, Y., Ishikura, H., Harada, T., Kobayashi, S. & Kim, S. U. (2005) Immortalized human microglial cell line: Phenotypic expression. Journal of Neuroscience Research, 81, 342-348.

Nakajima, K., Honda, S., Tohyama, Y., Imai, Y., Kohsaka, S. & Kurihara, T. (2001) Neurotrophin secretion from cultured microglia. Journal of Neuroscience Research, 65, 322-331.

Nakamura, Y., Si, Q. S. & Kataoka, K. (1999) Lipopolysaccharide-induced microglial activation in culture: temporal profiles of morphological change and release of cytokines and nitric oxide. Neuroscience Research, 35, 95-100.

Nakanishi, M., Niidome, T., Matsuda, S., Akaike, A., Kihara, T. & Sugimoto, H. (2007) Microglia-derived interleukin-6 and leukaemia inhibitory factor promote astrocytic differentiation of neural stem/progenitor cells. European Journal of Neuroscience, 25, 649-658.

Nava-Ruiz, C., Méndez-Armenta, M. & Ríos, C. (2012) Lead neurotoxicity: effects on brain nitric oxide synthase. Journal of Molecular Histology, 1-11.

Needleman, H. (2004) Lead poisoning. Annual Review of Medicine, 55, 209 - 222. Nelson, P. T., Soma, L. A. & Lavi, E. (2002) Microglia in diseases of the central nervous system.

Annals of Medicine, 34, 491-500. Nelson, T. E., Olde Engberink, A., Hernandez, R., Puro, A., Huitron-Resendiz, S., Hao, C., De

Graan, P. N. & Gruol, D. L. (2012) Altered synaptic transmission in the hippocampus of transgenic mice with enhanced central nervous systems expression of interleukin-6. Brain, Behavior, and Immunity, 26, 959-971.

Nemsadze, K., Sanikidze, T., Ratiani, L., Gabunia, L. & Sharashenidze, T. (2009) Mechanisms of lead-induced poisoning. Georgian Med News, 92-96.

NHMRC. 2009. Blood Lead level for asutralian [Online]. Available: http://www.nhmrc.gov.au/_files_nhmrc/publications/attachments/gp3-lead-pub-stmnt.pdf.

Page 198: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[177]

Ni, M. & Aschner, M. (2010) Neonatal rat primary microglia: isolation, culturing, and selected applications. Current Protocols in Toxicology, Chapter 12, Unit 12 17.

Nimmerjahn, A., Kirchhoff, F. & Helmchen, F. (2005) Resting Microglial Cells Are Highly Dynamic Surveillants of Brain Parenchyma in vivo. Science, 308, 1314-1318.

Nimmervoll, B., White, R., Yang, J. W., An, S., Henn, C., Sun, J. J. & Luhmann, H. J. (2012) LPS-Induced Microglial Secretion of TNFalpha Increases Activity-Dependent Neuronal Apoptosis in the Neonatal Cerebral Cortex. Cerebral Cortex, 23, 1742-1755.

Nishioku, T., Takai, N., Miyamoto, K., Murao, K., Hara, C., Yamamoto, K. & Nakanishi, H. (2000) Involvement of caspase 3-like protease in methylmercury-induced apoptosis of primary cultured rat cerebral microglia. Brain Research, 871, 160-164.

Noda, M. & Suzumura, A. (2012) Sweepers in the CNS: Microglial Migration and Phagocytosis in the Alzheimer Disease Pathogenesis. International Journal of Alzheimer's Disease, 891087. doi: 10.1155/2012/891087. Epub 2012 May 14

Noto, D., Takahashi, K., Miyake, S. & Yamada, M. (2010) In vitro differentiation of lineage-negative bone marrow cells into microglia-like cells. European Journal of Neuroscience, 31, 1155-1163.

Oberto, A., Marks, N., Evans, H. L. & Guidotti, A. (1996) Lead (Pb+2) promotes apoptosis in newborn rat cerebellar neurons: pathological implications. Journal of Pharmacology and Experimental Therapeutics, 279, 435-442.

Olson, J. K. & Miller, S. D. (2004) Microglia initiate central nervous system innate and adaptive immune responses through multiple TLRs. Journal of Immunology, 173, 3916-3924.

Olympio, K. P., Goncalves, C., Gunther, W. M. & Bechara, E. J. (2009) Neurotoxicity and aggressiveness triggered by low-level lead in children: a review. Revista Panamericana de Salud Publica, 26, 266-275.

Onalaja, A. O. & Claudio, L. (2000) Genetic susceptibility to lead poisoning. Environmental Health Perspectives, 108 Suppl 1, 23-28.

Opanashuk, L. A. & Finkelstein, J. N. (1995) Relationship of lead-induced proteins to stress response proteins in astroglial cells. Journal of Neuroscience Research, 42, 623-632.

Pachot, A., Cazalis, M.-A., Venet, F., Turrel, F., Faudot, C., Voirin, N., Diasparra, J., Bourgoin, N., Poitevin, F., Mougin, B., Lepape, A. & Monneret, G. (2008) Decreased Expression of the Fractalkine Receptor CX3CR1 on Circulating Monocytes as New Feature of Sepsis-Induced Immunosuppression. The Journal of Immunology, 180, 6421-6429.

Papanikolaou, N. C., Hatzidaki, E. G., Belivanis, S., Tzanakakis, G. N. & Tsatsakis, A. M. (2005) Lead toxicity update. A brief review. Medical Science Monitor, 11, RA329-336.

Park, M. H., Lee, Y. K., Lee, Y. H., Kim, Y.-B., Yun, Y. W., Nam, S. Y., Hwang, S. J., Han, S. B., Kim, S. U. & Hong, J. T. (2009) Chemokines released from astrocytes promote chemokine receptor 5-mediated neuronal cell differentiation. Experimental Cell Research, 315, 2715-2726.

Peng, Y. P., Qiu, Y. H., Lu, J. H. & Wang, J. J. (2005) Interleukin-6 protects cultured cerebellar granule neurons against glutamate-induced neurotoxicity. Neuroscience Letters, 374, 192-196.

Penninger, J. M., Irie-Sasaki, J., Sasaki, T. & Oliveira-dos-Santos, A. J. (2001) CD45: new jobs for an old acquaintance. Nature Immunology, 2, 389-396.

Penugonda, S., Mare, S., Lutz, P., Banks, W. A. & Ercal, N. (2006) Potentiation of lead-induced cell death in PC12 cells by glutamate: Protection by N-acetylcysteine amide (NACA), a novel thiol antioxidant. Toxicology and Applied Pharmacology, 216, 197-205.

Perry, V. H. & O'Connor, V. (2010) The role of microglia in synaptic stripping and synaptic degeneration: a revised perspective. ASN Neuro, 2, e00047.

Persidsky, Y., Ghorpade, A., Rasmussen, J., Limoges, J., Liu, X. J., Stins, M., Fiala, M., Way, D., Kim, K. S., Witte, M. H., Weinand, M., Carhart, L. & Gendelman, H. E. (1999) Microglial and astrocyte chemokines regulate monocyte migration through the blood-brain barrier in human immunodeficiency virus-1 encephalitis. American Journal of Pathology, 155, 1599-611.

Petito, C. K., Roberts, B., Cantando, J. D., Rabinstein, A. & Duncan, R. (2001) Hippocampal injury and alterations in neuronal chemokine co-receptor expression in patients with AIDS. Journal of Neuropathology and Experimental Neurology, 60, 377-385.

Page 199: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[178]

Philip, A. & Gerson, B. (1994) Lead poisoning-Part I. Incidence, etiology, and toxicokinetics. Clinics in Laboratory Medicine, 14, 423 - 444.

Piper, D. R., Mujtaba, T., Rao, M. S. & Lucero, M. T. (2000) Immunocytochemical and Physiological Characterization of a Population of Cultured Human Neural Precursors. Journal of Neurophysiology, 84, 534-548.

Politis, M., Pavese, N., Tai, Y. F., Kiferle, L., Mason, S. L., Brooks, D. J., Tabrizi, S. J., Barker, R. A. & Piccini, P. (2011) Microglial activation in regions related to cognitive function predicts disease onset in Huntington's disease: A multimodal imaging study. Human Brain Mapping, 32, 258-270.

Prevention, Advisory Committee on Childhood Lead Poisoning Prevention. 2012. Low Level Lead Exposure Harms Children: A Renewed Call for Primary Prevention. Centers for Disease Control and Prevention.

Prinz, M., Kann, O., Draheim, H. J., Schumann, R. R., Kettenmann, H., Weber, J. R. & Hanisch, U.-K. (1999) Microglial Activation by Components of Gram-Positive and -Negative Bacteria: Distinct and Common Routes to the Induction of Ion Channels and Cytokines. Journal of Neuropathology and Experimental Neurology, 58, 1078-1089.

Proudfoot, A. E. (2002) Chemokine receptors: multifaceted therapeutic targets. Nature Reviews Immunology, 2, 106-115.

Puma, C., Danik, M., Quirion, R., Ramon, F. & Williams, S. (2001) The chemokine interleukin-8 acutely reduces Ca(2+) currents in identified cholinergic septal neurons expressing CXCR1 and CXCR2 receptor mRNAs. Journal of Neurochemistry, 78, 960-971.

Qin, L., Li, G., Qian, X., Liu, Y., Wu, X., Liu, B., Hong, J.-S. & Block, M. L. (2005) Interactive role of the toll-like receptor 4 and reactive oxygen species in LPS-induced microglia activation. Glia, 52, 78-84.

Rai, A., Maurya, S. K., Khare, P., Srivastava, A. & Bandyopadhyay, S. (2010) Characterization of developmental neurotoxicity of As, Cd, and Pb mixture: synergistic action of metal mixture in glial and neuronal functions. Toxicological Sciences, 118, 586-601.

Rajan, P. & McKay, R. D. (1998) Multiple routes to astrocytic differentiation in the CNS. Journal of Neuroscience, 18, 3620-3629.

Ransohoff, R. & Perry, V. (2009) Microglial physiology: unique stimuli, specialized responses. Annual Review of Immunology, 27, 119 - 145.

Ransohoff, R. M., Liu, L. & Cardona, A. E. (2007) Chemokines and chemokine receptors: multipurpose players in neuroinflammation. International Review of Neurobiology, 82, 187-204.

Raoul, W., Auvynet, C., Camelo, S., Guillonneau, X., Feumi, C., Combadiere, C. & Sennlaub, F. (2010) CCL2/CCR2 and CX3CL1/CX3CR1 chemokine axes and their possible involvement in age-related macular degeneration. Journal of Neuroinflammation, 7, 87.

Raunio, S. & Tahti, H. (2001) Glutamate and calcium uptake in astrocytes after acute lead exposure. Chemosphere, 44, 355-359.

Re, F., Belyanskaya, S. L., Riese, R. J., Cipriani, B., Fischer, F. R., Granucci, F., Ricciardi-Castagnoli, P., Brosnan, C., Stern, L. J., Strominger, J. L. & Santambrogio, L. (2002) Granulocyte-macrophage colony-stimulating factor induces an expression program in neonatal microglia that primes them for antigen presentation. Journal of Immunology, 169, 2264-2273.

Reale, M., Iarlori, C., Thomas, A., Gambi, D., Perfetti, B., Di Nicola, M. & Onofrj, M. (2009) Peripheral cytokines profile in Parkinson’s disease. Brain, Behavior, and Immunity, 23, 55-63.

Rebenko-Moll, N. M., Liu, L., Cardona, A. & Ransohoff, R. M. (2006) Chemokines, mononuclear cells and the nervous system: heaven (or hell) is in the details. Current Opinion in Immunology, 18, 683-689.

Reiber, H. (2003) Proteins in cerebrospinal fluid and blood: barriers, CSF flow rate and source-related dynamics. Restorative Neurology and Neuroscience, 21, 79-96.

Rezaie, P., Trillo-Pazos, G., Everall, I. P. & Male, D. K. (2002) Expression of β-chemokines and chemokine receptors in human fetal astrocyte and microglial co-cultures: Potential role of chemokines in the developing CNS. Glia, 37, 64-75.

Robin, A. M., Zhang, Z. G., Wang, L., Zhang, R. L., Katakowski, M., Zhang, L., Wang, Y., Zhang, C. & Chopp, M. (2006) Stromal cell-derived factor 1alpha mediates neural progenitor cell

Page 200: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[179]

motility after focal cerebral ischemia. Journal of Cerebral Blood Flow and Metabolism, 26, 125-134.

Rock, R. B., Gekker, G., Hu, S., Sheng, W. S., Cheeran, M., Lokensgard, J. R. & Peterson, P. K. (2004) Role of Microglia in Central Nervous System Infections. Clinical Microbiology Reviews, 17, 942-964.

Rodero, M., Marie, Y., Coudert, M., Blondet, E., Mokhtari, K., Rousseau, A., Raoul, W., Carpentier, C., Sennlaub, F., Deterre, P., Delattre, J.-Y., Debré, P., Sanson, M. & Combadière, C. (2008) Polymorphism in the Microglial Cell-Mobilizing CX3CR1 Gene Is Associated With Survival in Patients With Glioblastoma. Journal of Clinical Oncology, 26, 5957-5964.

Rodier, P. M. (1995) Developing brain as a target of toxicity. Environmental Health Perspectives, 103 Suppl 6, 73-76.

Rogers, J. & Lue, L.-F. (2001) Microglial chemotaxis, activation, and phagocytosis of amyloid [beta]-peptide as linked phenomena in Alzheimer's disease. Neurochemistry International, 39, 333-340.

Rogers, J. T., Morganti, J. M., Bachstetter, A. D., Hudson, C. E., Peters, M. M., Grimmig, B. A., Weeber, E. J., Bickford, P. C. & Gemma, C. (2011) CX3CR1 deficiency leads to impairment of hippocampal cognitive function and synaptic plasticity. Journal of Neuroscience, 31, 16241-50.

Ronnett, G. V., Hester, L. D., Nye, J. S. & Snyder, S. H. (1994) Human cerebral cortical cell lines from patients with unilateral megalencephaly and Rasmussen's encephalitis. Neuroscience, 63, 1081-1099.

Rose-John, S., Scheller, J., Elson, G. & Jones, S. A. (2006) Interleukin-6 biology is coordinated by membrane-bound and soluble receptors: role in inflammation and cancer. Journal of Leukocyte Biology, 80, 227-236.

Rosner, D., Markowitz, G. & Lanphear, B. (2005) J. Lockhart Gibson and the discovery of the impact of lead pigments on children's health: a review of a century of knowledge. Public Health Reports, 120, 296-300.

Rostene, W., Kitabgi, P. & Parsadaniantz, S. M. (2007) Chemokines: a new class of neuromodulator? Nature Review Neuroscience, 8, 895-903.

Rothwell, N. J. & Strijbos, P. J. (1995) Cytokines in neurodegeneration and repair. International Journal of Developmental Neuroscience, 13, 179-185.

Rubio-Perez, J. M. & Morillas-Ruiz, J. M. (2012) A review: inflammatory process in Alzheimer's disease, role of cytokines. Scientific World Journal, 2012, 756357.

Ruitenberg, M. J., Vukovic, J., Blomster, L., Hall, J. M., Jung, S., Filgueira, L., McMenamin, P. G. & Plant, G. W. (2008) CX3CL1/fractalkine regulates branching and migration of monocyte-derived cells in the mouse olfactory epithelium. Journal of Neuroimmunology, 205, 80-85.

Saez, E. T., Pehar, M., Vargas, M. R., Barbeito, L. & Maccioni, R. B. (2006) Production of nerve growth factor by beta-amyloid-stimulated astrocytes induces p75NTR-dependent tau hyperphosphorylation in cultured hippocampal neurons. Journal of Neuroscience Research, 84, 1098-1106.

Sandhir, R., Julka, D. & Dip Gill, K. (1994) Lipoperoxidative Damage on Lead Exposure in Rat Brain and its Implications on Membrane Bound Enzymes. Pharmacology and Toxicology, 74, 66-71.

Schafer, K. H., Mestres, P., Marz, P. & Rose-John, S. (1999) The IL-6/sIL-6R fusion protein hyper-IL-6 promotes neurite outgrowth and neuron survival in cultured enteric neurons. Journal of Interferon and Cytokine Research, 19, 527-532.

Schilling, M., Strecker, J.-K., Ringelstein, E. B., Schäbitz, W.-R. & Kiefer, R. (2009) The role of CC chemokine receptor 2 on microglia activation and blood-borne cell recruitment after transient focal cerebral ischemia in mice. Brain Research, 1289, 79-84.

Schilling, T. & Eder, C. (2011) Amyloid-beta-induced reactive oxygen species production and priming are differentially regulated by ion channels in microglia. Journal of Cellular Physiology, 226, 3295-3302.

Schilling, T., Nitsch, R., Heinemann, U., Haas, D. & Eder, C. (2001) Astrocyte-released cytokines induce ramification and outward K+ channel expression in microglia via distinct signalling pathways. European Journal of Neuroscience, 14, 463-473.

Page 201: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[180]

Schmidtmayer, J., Jacobsen, C., Miksch, G. & Sievers, J. (1994) Blood monocytes and spleen macrophages differentiate into microglia-like cells on monolayers of astrocytes: Membrane currents. Glia, 12, 259-267.

Schmuck, G., Ahr, H. J. & Schluter, G. (2000) Rat cortical neuron cultures: an in vitro model for differentiating mechanisms of chemically induced neurotoxicity. In vitro and Molecular Toxicology, 13, 37-50.

Schneider, J. S., Anderson, D. W., Sonnenahalli, H. & Vadigepalli, R. (2011) Sex-based differences in gene expression in hippocampus following postnatal lead exposure. Toxicology and Applied Pharmacology, 256, 179-190.

Schneider, J. S., Huang, F. N. & Vemuri, M. C. (2003) Effects of low-level lead exposure on cell survival and neurite length in primary mesencephalic cultures. Neurotoxicology and Teratology, 25, 555-559.

Schwartz, J. & Levin, R. (1991) The risk of lead toxicity in homes with lead paint hazard. Environmental Research, 54, 1-7.

Selvin-Testa, A., Loidl, C. F., Lopez-Costa, J. J., Lopez, E. M. & Pecci-Saavedra, J. (1994) Chronic lead exposure induces astrogliosis in hippocampus and cerebellum. Neurotoxicology, 15, 389-401.

Semple, B. D., Kossmann, T. & Morganti-Kossmann, M. C. (2010) Role of chemokines in CNS health and pathology: a focus on the CCL2/CCR2 and CXCL8/CXCR2 networks. Journal of Cerebral Blood Flow and Metabolism, 30, 459-473.

Sengupta, M. & Bishayi, B. (2002) Effect of lead and arsenic on murine macrophage response. Drug and Chemical Toxicology, 25, 459-472.

Sharma S, Sharma V, Paliwal R & Pracheta (2011) Lead toxicity, oxidative damage and health implications. A review. International Journal for Biotechnology and Molecular Biology Research, 2, 215-221.

Sheng, J. G., Jones, R. A., Zhou, X. Q., McGinness, J. M., Van Eldik, L. J., Mrak, R. E. & Griffin, W. S. (2001) Interleukin-1 promotion of MAPK-p38 overexpression in experimental animals and in Alzheimer's disease: potential significance for tau protein phosphorylation. Neurochemistry International, 39, 341-348.

Sheng, W., Zong, Y., Mohammad, A., Ajit, D., Cui, J., Han, D., Hamilton, J. L., Simonyi, A., Sun, A. Y., Gu, Z., Hong, J. S., Weisman, G. A. & Sun, G. Y. (2011) Pro-inflammatory cytokines and lipopolysaccharide induce changes in cell morphology, and upregulation of ERK1/2, iNOS and sPLA(2)-IIA expression in astrocytes and microglia. Journal of Neuroinflammation, 8, 121.

Sievers, J., Parwaresch, R. & Wottge, H.-U. (1994) Blood monocytes and spleen macrophages differentiate into microglia-like cells on monolayers of astrocytes: Morphology. Glia, 12, 245-258.

Simard, A. R., Soulet, D., Gowing, G., Julien, J.-P. & Rivest, S. (2006) Bone Marrow-Derived Microglia Play a Critical Role in Restricting Senile Plaque Formation in Alzheimer's Disease. Neuron, 49, 489-502.

Simpson, J., Rezaie, P., Newcombe, J., Cuzner, M. L., Male, D. & Woodroofe, M. N. (2000) Expression of the β-chemokine receptors CCR2, CCR3 and CCR5 in multiple sclerosis central nervous system tissue. Journal of Neuroimmunology, 108, 192-200.

Singh, K. K., Hughes, M. D., Chen, J. & Spector, S. A. (2005) Genetic polymorphisms in CX3CR1 predict HIV-1 disease progression in children independently of CD4+ lymphocyte count and HIV-1 RNA load. Journal of Infectious Diseases, 191, 1971-1980.

Škuljec, J., Sun, H., Pul, R., Bénardais, K., Ragancokova, D., Moharregh-Khiabani, D., Kotsiari, A., Trebst, C. & Stangel, M. (2011) CCL5 induces a pro-inflammatory profile in microglia in vitro. Cellular Immunology, 270, 164-171.

Slomianka, L., Rungby, J., West, M. J., Danscher, G. & Andersen, A. H. (1989) Dose-dependent bimodal effect of low-level lead exposure on the developing hippocampal region of the rat: a volumetric study. Neurotoxicology, 10, 177-190.

Smith, J. A., Das, A., Ray, S. K. & Banik, N. L. (2012) Role of pro-inflammatory cytokines released from microglia in neurodegenerative diseases. Brain Research Bulletin, 87, 10-20.

Sobin, C., Gutierrez, M. & Alterio, H. (2009) Polymorphisms of delta-aminolevulinic acid dehydratase (ALAD) and peptide transporter 2 (PEPT2) genes in children with low-level lead exposure. Neurotoxicology, 30, 881-887.

Page 202: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[181]

Soulet, D. & Rivest, S. (2008) Bone-marrow-derived microglia: myth or reality? Current Opinion in Pharmacology, 8, 508-518.

Song, J. H., Wang, C. X., Song, D. K., Wang, P., Shuaib, A. & Hao, C. (2005) Interferon gamma induces neurite outgrowth by up-regulation of p35 neuron-specific cyclin-dependent kinase 5 activator via activation of ERK1/2 pathway. Journal of Biological Chemistry, 280, 12896-12901.

Song, M., Jin, J., Lim, J.-E., Kou, J., Pattanayak, A., Rehman, J., Kim, H.-D., Tahara, K., Lalonde, R. & Fukuchi, K.-i. (2011) TLR4 mutation reduces microglial activation, increases Abeta deposits and exacerbates cognitive deficits in a mouse model of Alzheimer's disease. Journal of Neuroinflammation, 8, 92.

Sorensen, T. L., Tani, M., Jensen, J., Pierce, V., Lucchinetti, C., Folcik, V. A., Qin, S., Rottman, J., Sellebjerg, F., Strieter, R. M., Frederiksen, J. L. & Ransohoff, R. M. (1999) Expression of specific chemokines and chemokine receptors in the central nervous system of multiple sclerosis patients. Journal of Clinical Investigation, 103, 807-815.

Stevens, B. (2008) Neuron-Astrocyte Signaling in the Development and Plasticity of Neural Circuits. Neurosignals, 16, 278-288.

Streit, W. J. (1996) The role of microglia in brain injury. Neurotoxicology, 17, 671-678. Streit, W. J. (2001) Microglia and Macrophages in the Developing CNS. Neurotoxicology, 22,

619-624. Streit, W. J. (2002) Microglia as neuroprotective, immunocompetent cells of the CNS. Glia, 40,

133-139. Struzynska, L., Dabrowska-Bouta, B., Koza, K. & Sulkowski, G. (2007) Inflammation-like glial

response in lead-exposed immature rat brain. Toxicological Sciences, 95, 156 - 162. Sugama, S., Takenouchi, T., Cho, B. P., Joh, T. H., Hashimoto, M. & Kitani, H. (2009) Possible roles

of microglial cells for neurotoxicity in clinical neurodegenerative diseases and experimental animal models. Inflammation and Allergy- Drug Targets, 8, 277-284.

Suzuki, M., El-Hage, N., Zou, S., Hahn, Y. K., Sorrell, M. E., Sturgill, J. L., Conrad, D. H., Knapp, P. E. & Hauser, K. F. (2011) Fractalkine/CX3CL1 protects striatal neurons from synergistic morphine and HIV-1 Tat-induced dendritic losses and death. Molecular Neurodegeneration, 6, 78.

Szczuciński, A. & Losy, J. (2007) Chemokines and chemokine receptors in multiple sclerosis. Potential targets for new therapies. Acta Neurologica Scandinavica, 115, 137-146.

Szelényi, J. (2001) Cytokines and the central nervous system. Brain Research Bulletin, 54, 329-338.

Taga, T. & Fukuda, S. (2005) Role of IL-6 in the neural stem cell differentiation. Clinical Reviews in Allergy and Immunology, 28, 249-256.

Tambuyzer, B. R., Ponsaerts, P. & Nouwen, E. J. (2009) Microglia: gatekeepers of central nervous system immunology. Journal of Leukocyte Biology, 85, 352-370.

Tanaka, S., Kondo, H., Kanda, K., Ashino, T., Nakamachi, T., Sekikawa, K., Iwakura, Y., Shioda, S., Numazawa, S. & Yoshida, T. (2011) Involvement of interleukin-1 in lipopolysaccaride-induced microglial activation and learning and memory deficits. Journal of Neuroscience Research, 89, 506-14.

Tang, H., Wang, Y., Xie, L., Mao, X., Won, S. J., Galvan, V. & Jin, K. (2009) Effect of neural precursor proliferation level on neurogenesis in rat brain during aging and after focal ischemia. Neurobiology of Aging, 30, 299-308.

Tang, Y., Jia, Q., Ma, P., Li, F., Lv, T. & Ha, X. (2010) [The effects of lead exposure on primary cultured hippocampus neurons apoptosis and the expressions of Bcl-2, Bax, p53 and NOS1]. Wei Sheng Yan Jiu, 39, 235-8.

Tanga, F. Y., Nutile-McMenemy, N. & DeLeo, J. A. (2005) The CNS role of Toll-like receptor 4 in innate neuroimmunity and painful neuropathy. Proceedings of the National Academy of Sciences of the United States of America, 102, 5856-5861.

Tanuma, N., Sakuma, H., Sasaki, A. & Matsumoto, Y. (2006) Chemokine expression by astrocytes plays a role in microglia/macrophage activation and subsequent neurodegeneration in secondary progressive multiple sclerosis. Acta Neuropathologica, 112, 195-204.

Thacker, M. A., Clark, A. K., Bishop, T., Grist, J., Yip, P. K., Moon, L. D. F., Thompson, S. W. N., Marchand, F. & McMahon, S. B. (2009) CCL2 is a key mediator of microglia activation in neuropathic pain states. European Journal of Pain (London, England), 13, 263-272.

Page 203: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[182]

Thier, M., Marz, P., Otten, U., Weis, J. & Rose-John, S. (1999) Interleukin-6 (IL-6) and its soluble receptor support survival of sensory neurons. Journal of Neuroscience Research, 55, 411-22.

Thirumangalakudi, L., Yin, L., Rao, H. V. & Grammas, P. (2007) IL-8 Induces Expression of Matrix Metalloproteinases, Cell Cycle and Pro-Apoptotic Proteins, and Cell Death in Cultured Neurons. Journal of Alzheimer's Disease, 11, 305-311.

Thomas, M. P., Chartrand, K., Reynolds, A., Vitvitsky, V., Banerjee, R. & Gendelman, H. E. (2007) Ion channel blockade attenuates aggregated alpha synuclein induction of microglial reactive oxygen species: relevance for the pathogenesis of Parkinson's disease. Journal of Neurochemistry, 100, 503-519.

Tiffany-Castiglion, E. & Qian, Y. (2001) Astroglia as metal depots: molecular mechanisms for metal accumulation, storage and release. Neurotoxicology, 22, 577-592.

Tiffany-Castiglioni, E. (1993) Cell culture models for lead toxicity in neuronal and glial cells. Neurotoxicology, 14, 513-536.

Tiffany-Castiglioni, E. & Qian, Y. (2001) Astroglia as Metal Depots: Molecular Mechanisms for Metal Accumulation, Storage and Release. Neurotoxicology, 22, 577-592.

Tissir, F., Wang, C.-E. & Goffinet, A. M. (2004) Expression of the chemokine receptor Cxcr4 mRNA during mouse brain development. Developmental Brain Research, 149, 63-71.

Todd, A. C., Wetmur, J. G., Moline, J. M., Godbold, J. H., Levin, S. M. & Landrigan, P. J. (1996) Unraveling the chronic toxicity of lead: an essential priority for environmental health. Environmental Health Perspectives, 104 Suppl 1, 141-146.

Tonchev, A. B. (2011) The nerve growth factor in health and unhealth neurons. Archives Italiennes de Biologie, 149, 225-231.

Toscano, C. & Guilarte, T. (2005) Lead neurotoxicity: from exposure to molecular effects. Brain Research. Brain Research Reviews, 49, 529 - 554.

Tran, P. B., Banisadr, G., Ren, D., Chenn, A. & Miller, R. J. (2007) Chemokine receptor expression by neural progenitor cells in neurogenic regions of mouse brain. Journal of Comparative Neurology, 500, 1007-1033.

Tran, P. B. & Miller, R. J. (2003) Chemokine receptors: signposts to brain development and disease. Nature Review Neuroscience, 4, 444-455.

Tremblay, M. E. (2011) The role of microglia at synapses in the healthy CNS: novel insights from recent imaging studies. Neuron Glia Biology, 7, 67-76.

Tremblay, M. E. & Majewska, A. K. (2011) A role for microglia in synaptic plasticity? Communicative and Integrative Biology, 4, 220-222.

Tremblay, M. E., Stevens, B., Sierra, A., Wake, H., Bessis, A. & Nimmerjahn, A. (2011) The Role of Microglia in the Healthy Brain. Journal of Neuroscience, 31, 16064-16069.

Triantafilou, M. & Triantafilou, K. (2002) Lipopolysaccharide recognition: CD14, TLRs and the LPS-activation cluster. Trends in Immunology, 23, 301-304.

Trudler, D., Farfara, D. & Frenkel, D. (2010) Toll-Like Receptors Expression and Signaling in Glia Cells in Neuro-Amyloidogenic Diseases: Towards Future Therapeutic Application. Mediators of Inflammation, 2010.

Tsou, C. L., Haskell, C. A. & Charo, I. F. (2001) Tumor necrosis factor-alpha-converting enzyme mediates the inducible cleavage of fractalkine. Journal of Biological Chemistry, 276, 44622-6.

Turbic, A., Leong, S. Y. & Turnley, A. M. (2011) Chemokines and inflammatory mediators interact to regulate adult murine neural precursor cell proliferation, survival and differentiation. PLoS ONE, 6, e25406.

Vairano, M., Graziani, G., Tentori, L., Tringali, G., Navarra, P. & Russo, C. D. (2004) Primary cultures of microglial cells for testing toxicity of anticancer drugs. Toxicology Letters, 148, 91-94.

Valdo, P., Stegagno, C., Mazzucco, S., Zuliani, E., Zanusso, G., Moretto, G., Raine, C. S. & Bonetti, B. (2002) Enhanced expression of NGF receptors in multiple sclerosis lesions. Journal of Neuropathology and Experimental Neurology, 61, 91-98.

Vallieres, L., Campbell, I. L., Gage, F. H. & Sawchenko, P. E. (2002) Reduced hippocampal neurogenesis in adult transgenic mice with chronic astrocytic production of interleukin-6. Journal of Neuroscience, 22, 486-492.

Page 204: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[183]

Vallieres, L. & Rivest, S. (1997) Regulation of the genes encoding interleukin-6, its receptor, and gp130 in the rat brain in response to the immune activator lipopolysaccharide and the proinflammatory cytokine interleukin-1 beta. Journal of Neurochemistry, 69, 1668-1683.

Van Der Meer, P., Goldberg, S. H., Fung, K. M., Sharer, L. R., González-Scarano, F. & Lavi, E. (2001) Expression Pattern of CXCR3, CXCR4, and CCR3 Chemokine Receptors in the Developing Human Brain. Journal of Neuropathology and Experimental Neurology, 60, 25-32.

Verri, M., Pastoris, O., Dossena, M., Aquilani, R., Guerriero, F., Cuzzoni, G., Venturini, L., Ricevuti, G. & Bongiorno, A. I. (2012) Mitochondrial alterations, oxidative stress and neuroinflammation in Alzheimer’s disease. International Journal of Immunopathology and Pharmacology, 25, 345-353.

Verstraeten, S., Aimo, L. & Oteiza, P. (2008) Aluminium and lead: molecular mechanisms of brain toxicity. Archives of Toxicology, 82, 789 - 802.

Vila, M., Jackson-Lewis, V., Guegan, C., Wu, D. C., Teismann, P., Choi, D. K., Tieu, K. & Przedborski, S. (2001) The role of glial cells in Parkinson's disease. Current Opinion in Neurology, 14, 483-489.

illeda-Herna ndez, J., Barroso-Moguel, R., Me ndez-Armenta, M., Nava-Ru z, C., Huerta-Romero, R. & R os, C. (2001) Enhanced brain regional lipid peroxidation in developing rats exposed to low level lead acetate. Brain Research Bulletin, 55, 247-251.

Viviani, B., Corsini, E., Galli, C. L. & Marinovich, M. (1998) Glia Increase Degeneration of Hippocampal Neurons through Release of Tumor Necrosis Factor-α. Toxicology and Applied Pharmacology, 150, 271-276.

Voloboueva, L. A. & Giffard, R. G. (2011) Inflammation, mitochondria, and the inhibition of adult neurogenesis. Journal of Neuroscience Research, 89, 1989-1996.

Vukovic, J., Colditz, M. J., Blackmore, D. G., Ruitenberg, M. J. & Bartlett, P. F. (2012) Microglia modulate hippocampal neural precursor activity in response to exercise and aging. Journal of Neuroscience, 32, 6435-6443.

Walker, D. G., Dalsing-Hernandez, J. E., Campbell, N. A. & Lue, L. F. (2009) Decreased expression of CD200 and CD200 receptor in Alzheimer's disease: a potential mechanism leading to chronic inflammation. Experimental Neurology, 215, 5-19.

Wang, L., Li, Y., Chen, X., Chen, J., Gautam, S., Xu, Y. & Chopp, M. (2002) MCP-1, MIP-1, IL-8 and Ischemic Cerebral Tissue Enhance Human Bone Marrow Stromal Cell Migration in Interface Culture. Hematology, 7, 113-117.

Wang, X., Fu, S., Wang, Y., Yu, P., Hu, J., Gu, W., Xu, X. M. & Lu, P. (2007) Interleukin-1beta mediates proliferation and differentiation of multipotent neural precursor cells through the activation of SAPK/JNK pathway. Molecular and Cellular Neurosciences, 36, 343-354.

Wang, X. M., Liu, W. J., Zhang, R. & Zhou, Y. K. (2012) Effects of exposure to low-level lead on spatial learning and memory and the expression of mGluR1, NMDA receptor in different developmental stages of rats. Toxicology and Industrial Health, published on February 23, 2012, DOI: 10.1177/0748233712436641.Waterman, S. J., el-Fawal, H. A. & Snyder, C. A. (1994) Lead alters the immunogenicity of two neural proteins: a potential mechanism for the progression of lead-induced neurotoxicity. Environmental Health Perspectives, 102, 1052-1056.

Węgiel, J., Wiśniewski, H. M., Dziewiątkowski, J., Tarnawski, M., Kozielski, R., Trenkner, E. & Wiktor-Jędrzejczak, W. (1998) Reduced number and altered morphology of microglial cells in colony stimulating factor-1-deficient osteopetrotic op/op mice. Brain Research, 804, 135-139.

Wei, H., Chadman, K. K., McCloskey, D. P., Sheikh, A. M., Malik, M., Brown, W. T. & Li, X. (2012) Brain IL-6 elevation causes neuronal circuitry imbalances and mediates autism-like behaviors. Biochimica et Biophysica Acta, 1822, 831-42.

Welzl, H. & Stork, O. (2003) Cell Adhesion Molecules: Key Players in Memory Consolidation? Physiology, 18, 147-150.

White, L., Cory-Slechta, D., Gilbert, M., Tiffany-Castiglioni, E., Zawia, N., Virgolini, M., Rossi-George, A., Lasley, S., Qian, Y. & Basha, M. (2007) New and evolving concepts in the neurotoxicology of lead. Toxicology and Applied Pharmacology, 225, 1 - 27.

Page 205: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[184]

Whitney, N. P., Eidem, T. M., Peng, H., Huang, Y. & Zheng, J. C. (2009) Inflammation mediates varying effects in neurogenesis: relevance to the pathogenesis of brain injury and neurodegenerative disorders. Journal of Neurochemistry, 108, 1343-1359.

WHO. 2010. Chilhood Lead Poisioning [Online]. World Health Organization. Available: http://www.who.int/ceh/publications/leadguidance.pdf.

Williams, K., Jr., Ulvestad, E., Cragg, L., Blain, M. & Antel, J. P. (1993) Induction of primary T cell responses by human glial cells. Journal of Neuroscience Research, 36, 382-390.

Wirenfeldt, M., Ladeby, R., Dalmau, I., Banati, R. B. & Finsen, B. (2005) Microglia - Biology and relevance to disease. Mikroglia - Biologi og sygdomsmæssig relevans, 167, 3025-3030.

Woolley, D. E. (1984) A perspective of lead poisoning in antiquity and the present. Neurotoxicology, 5, 353-61.

Wu, Y., Chen, Q., Peng, H., Dou, H., Zhou, Y., Huang, Y. & Zheng, J. (2012) Directed migration of human neural progenitor cells to interleukin-1beta is promoted by chemokines stromal cell-derived factor-1 and monocyte chemotactic factor-1 in mouse brains. Translational Neurodegeneration, 1, 15.

Wyss-Coray, T. & Mucke, L. (2002) Inflammation in Neurodegenerative Disease—A Double-Edged Sword. Neuron, 35, 419-432.

Xia, M. & Hyman, B. T. (1999) Chemokines/chemokine receptors in the central nervous system and Alzheimer's disease. Journal of Neurovirology, 5, 32-41.

Xiang, Z., Chen, C. J., Ping, J., Dunn, P., Lv, J., Jiao, B. & Burnstock, G. (2006) Microglial Morphology and Its Transformation After Challenge by Extracellular ATP In vitro. Journal of Neuroscince Research, 83, 91-101.

Xiong, H., Boyle, J., Winkelbauer, M., Gorantla, S., Zheng, J., Ghorpade, A., Persidsky, Y., Carlson, K. A. & Gendelman, H. E. (2003) Inhibition of long-term potentiation by interleukin-8: Implications for human immunodeficiency virus-1-associated dementia. Journal of Neuroscience Research, 71, 600-607.

Xu, F., Farkas, S., Kortbeek, S., Zhang, F.-X., Chen, L., Zamponi, G. & Syed, N. (2012) Mercury-induced toxicity of rat cortical neurons is mediated through N-methyl-D-Aspartate receptors. Molecular Brain, 5, 30.

Xu, J., Ji, L.-D. & Xu, L.-H. (2006) Lead-induced apoptosis in PC 12 cells: Involvement of p53, Bcl-2 family and caspase-3. Toxicology Letters, 166, 160-167.

Xu, J., Yan, H. C., Yang, B., Tong, L. S., Zou, Y. X. & Tian, Y. (2009) Effects of lead exposure on hippocampal metabotropic glutamate receptor subtype 3 and 7 in developmental rats. Journal of Negative Results in Biomedicine, 8, 5.

Xu, S. Z., Shan, C. J., Bullock, L., Baker, L. & Rajanna, B. (2006) Pb2+ reduces PKCs and NF-kappaB in vitro. Cell Biology and Toxicology, 22, 189-198.

Xu, Y., Zeng, K., Han, Y., Wang, L., Chen, D., Xi, Z., Wang, H., Wang, X. & Chen, G. (2012) Altered expression of CX3CL1 in patients with epilepsy and in a rat model. American Journal of Pathology, 180, 1950-1962.

Yamada, M. & Hatanaka, H. (1994) Interleukin-6 protects cultured rat hippocampal neurons against glutamate-induced cell death. Brain Research, 643, 173-180.

Yang, C. S., Lee, H. M., Lee, J. Y., Kim, J. A., Lee, S. J., Shin, D. M., Lee, Y. H., Lee, D. S., El-Benna, J. & Jo, E. K. (2007) Reactive oxygen species and p47phox activation are essential for the Mycobacterium tuberculosis-induced pro-inflammatory response in murine microglia. Journal of Neuroinflammation, 4, 27.

Yang, Z., Yang, S., Qian, S. Y., Hong, J. S., Kadiiska, M. B., Tennant, R. W., Waalkes, M. P. & Liu, J. (2007) Cadmium-induced toxicity in rat primary mid-brain neuroglia cultures: role of oxidative stress from microglia. Toxicological Sciences, 98, 488-494.

Yeh, S.-H., Hung, J.-J., Gean, P.-W. & Chang, W.-C. (2008) Hypoxia-Inducible Factor-1α Protects Cultured Cortical Neurons from Lipopolysaccharide-Induced Cell Death via Regulation of NR1 Expression. The Journal of Neuroscience, 28, 14259-14270.

Yoneyama, M., Shiba, T., Hasebe, S. & Ogita, K. (2011) Adult neurogenesis is regulated by endogenous factors produced during neurodegeneration. Journal of Pharmacological Sciences, 115, 425-432.

Zeller, I., Knoflach, M., Seubert, A., Kreutmayer, S. B., Stelzmuller, M. E., Wallnoefer, E., Blunder, S., Frotschnig, S., Messner, B., Willeit, J., Debbage, P., Wick, G., Kiechl, S., Laufer, G. & Bernhard, D. (2010) Lead contributes to arterial intimal hyperplasia through nuclear

Page 206: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[185]

factor erythroid 2-related factor-mediated endothelial interleukin 8 synthesis and subsequent invasion of smooth muscle cells. Arteriosclerosis, Thrombosis, and Vascular Biology, 30, 1733-1740.

Zhang, J., Geula, C., Lu, C., Koziel, H., Hatcher, L. M. & Roisen, F. J. (2003) Neurotrophins regulate proliferation and survival of two microglial cell lines in vitro. Experimental Neurology, 183, 469-481.

Zhang, J., Shi, X. Q., Echeverry, S., Mogil, J. S., De Koninck, Y. & Rivest, S. (2007) Expression of CCR2 in Both Resident and Bone Marrow-Derived Microglia Plays a Critical Role in Neuropathic Pain. The Journal of Neuroscience, 27, 12396-12406.

Zhang, T., Somasundaram, R., Berencsi, K., Caputo, L., Gimotty, P., Rani, P., Guerry, D., Swoboda, R. & Herlyn, D. (2006) Migration of cytotoxic T lymphocytes toward melanoma cells in three-dimensional organotypic culture is dependent on CCL2 and CCR4. European Journal of Immunology, 36, 457-467.

Zhang, Z., Drzewiecki, G. J., Hom, J. T., May, P. C. & Hyslop, P. A. (1994) Human cortical neuronal (HCN) cell lines: a model for amyloid β neurotoxicity. Neuroscience Letters, 177, 162-164.

Zhao, C., Deng, W. & Gage, F. H. (2008) Mechanisms and functional implications of adult neurogenesis. Cell, 132, 645-660.

Zheng, G., Tian, L., Liang, Y., Broberg, K., Lei, L., Guo, W., Nilsson, J., Bergdahl, I. A., Skerfving, S. & Jin, T. (2011) delta-Aminolevulinic acid dehydratase genotype predicts toxic effects of lead on workers' peripheral nervous system. Neurotoxicology, 32, 374-382.

Zheng, W., Aschner, M. & Ghersi-Egea, J. F. (2003) Brain barrier systems: a new frontier in metal neurotoxicological research. Toxicology and Applied Pharmacology, 192, 1-11.

Zimmer, B., Lee, G., Stiegler, N. V., Meganathan, K., Sachinidis, A., Studer, L. & Leist, M. (2012) Evaluation of Developmental Toxicants and Signaling Pathways in a Functional Test Based on the Migration of Human Neural Crest Cells. Environmental Health Perspectives, 120, 1116-1122.

Ziv, Y. & Schwartz, M. (2008) Immune-based regulation of adult neurogenesis: Implications for learning and memory. Brain, Behavior, and Immunity, 22, 167-176.

Zuckerman, S. H., Gustin, J. & Evans, G. F. (1998) Expression of CD54 (intercellular adhesion molecule-1) and the beta 1 integrin CD29 is modulated by a cyclic AMP dependent pathway in activated primary rat microglial cell cultures. Inflammation, 22, 95-106.

Zuiderwijk-Sick, E. A., van der Putten, C., Bsibsi, M., Deuzing, I. P., de Boer, W., Persoon-Deen, C., Kondova, I., Boven, L. A., van Noort, J. M., t Hart, B. A., Amor, S. & Bajramovic, J. J. (2007) Differentiation of primary adult microglia alters their response to TLR8-mediated activation but not their capacity as APC. Glia, 55, 1589-1600.

Zujovic, V., Benavides, J., Vigé, X., Carter, C. & Taupin, V. (2000) Fractalkine modulates TNF-α secretion and neurotoxicity induced by microglial activation. Glia, 29, 305-315.

Zurich, M. G., Monnet-Tschudi, F., Bérode, M. & Honegger, P. (1998) Lead acetate toxicity in vitro: Dependence on the cell composition of the cultures. Toxicology in vitro, 12, 191-196.

Zurich, M.-G., Eskes, C., Honegger, P., Bérode, M. & Monnet-Tschudi, F. (2002) Maturation-dependent neurotoxicity of lead acetate in vitro: Implication of glial reactions. Journal of Neuroscience Research, 70, 108-116.

Page 207: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

Chapter 10

Appendix

Page 208: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[187]

A

Page 209: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[188]

B

B

Page 210: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[189]

C

Page 211: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

D

Page 212: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

[191]

Page 213: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

B

Am

Sa

b

c

h

a

ARRA

KMHSFCMGCNP

mCD

0h

Journal of Neuroscience Methods 209 (2012) 79– 89

Contents lists available at SciVerse ScienceDirect

Journal of Neuroscience Methods

jou rna l h om epa ge: www.elsev ier .com/ locate / jneumeth

asic Neuroscience

novel in vitro human microglia model: Characterization of humanonocyte-derived microglia

amar Etemada, Rasheeda Mohd Zamina,b, Marc J. Ruitenbergc, Luis Filgueiraa,∗

School of Anatomy, Physiology and Human Biology, University of Western Australia, WA 6009, AustraliaDepartment of Anatomy, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, MalaysiaSchool of Biomedical Sciences, University of Queensland, Brisbane, QLD 4072, Australia

i g h l i g h t s

New culture protocol for the gen-eration of human monocyte-derivedmicroglia in vitro.Morphological, phenotypic and func-tional characterization of humanmonocyte-derived microglia.Expression of surface markers byhuman microglia.Chemokine receptors expressionpattern in human microglia.Comparison of human microglia withmonocytes and dendritic cells.

g r a p h i c a l a b s t r a c t

r t i c l e i n f o

rticle history:eceived 21 December 2011eceived in revised form 20 April 2012ccepted 22 May 2012

eywords:icrogliauman peripheral blood monocyteserum-free mediumlow cytometryhemokine receptors-CSF

a b s t r a c t

Microglia are the innate immune cells of the central nervous system. They help maintaining physiologicalhomeostasis and contribute significantly to inflammatory responses in the course of infection, injuryand degenerative processes. To date, there is no standardized simple model available to investigate thebiology of human microglia. The aim of this study was to establish a new human microglia model. Forthat purpose, human peripheral blood monocytes were cultured in serum free medium in the presenceof M-CSF, GM-CSF, NGF and CCL2 to generate monocyte-derived microglia (M-MG). M-MG were clearlydifferent in morphology, phenotype and function from freshly isolated monocytes, cultured monocytesin the absence of the cytokines and monocyte-derived dendritic cells (M-DC) cultured in the presenceof GM-CSF and IL-4. M-MG acquired a ramified morphology with primary and secondary processes. M-MG displayed a comparable phenotype to the human microglia cell line HMC3, expressing very lowlevels of CD45, CD14 and HLA-DR, CD11b and CD11c; and undetectable levels of CD40, CD80 and CD83,

M-CSFCL2GFhagocytosis

and a distinct pattern of chemokine receptors (positive for CCR1, CCR2, CCR4, CCR5, CXCR1, CXCR3,CX3CR1; negative for CCR6 and CCR7). In comparison with M-DC, M-MG displayed lower T-lymphocytestimulatory capacity, as well as lower phagocytosis activity. The described protocol for the generationof human monocyte-derived microglia is feasible, well standardized and reliable, as it uses well definedculture medium and recombincertainly be very helpful for fu

Abbreviations: CNS, central nervous system; PBMC, peripheral mononuclear bloacrophage colony stimulating factor; NGF, nerve growth factor; CCL2, c-c motif ligand

CR1, c-c motif receptor 1; CCR2, c-c motif receptor 2; CXCR3, c-x-c motif receptor 3;

ulbecco’s Modification of Eagle’s Medium; FBS, fetal bovine serum.∗ Corresponding author at: School of Anatomy and Human Biology, University of Weste

Tel.: +61 8 64883907; fax: +61 8 64881051.E-mail address: [email protected] (L. Filgueira).

165-0270/$ – see front matter © 2012 Elsevier B.V. All rights reserved.ttp://dx.doi.org/10.1016/j.jneumeth.2012.05.025

ant cytokines, but no serum or conditioned medium. This protocol willture studies investigating the biology and pathology of human microglia.

© 2012 Elsevier B.V. All rights reserved.

od cells; M-CSF, macrophage-colony stimulating factor; GM-CSF, granulocyte- 2; M-MG, monocyte-derived microglia; M-DC, monocyte-derived dendritic cell;CX3CR1, cxc3 chemokine receptor 1; CSF-1, colony stimulating factor 1; DMEM,

rn Australia, 35 Stirling Highway, M309, Crawley, WA 6009, Australia.

cs_stud1
Stamp
cs_stud1
Sticky Note
Accepted set by cs_stud1
cs_stud1
Sticky Note
MigrationConfirmed set by cs_stud1
Page 214: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

8 roscie

1

vhsaLqccptSglermmeaaEiloatT

i(kmemts(oc

mft2aiduaBswKfiobiWs

b(w

0 S. Etemad et al. / Journal of Neu

. Introduction

Microglia are resident innate immune cells of the central ner-ous system (CNS) and play a crucial role in maintaining theealthy physiological homeostasis. Microglia also contribute sub-tantially to inflammation in response to injury, toxins, pathogensnd degenerative processes (Streit, 2001, 1996; Howell et al., 2010;ee et al., 2010; Politis et al., 2011). Microglia are characterized by auick response to various stimuli, resulting in activation with rapidhanges in morphology, phenotype and function. Morphologicalhanges include shortening of cell processes and cellular hypertro-hy. To date, there is no specific microglia marker clearly separatinghis cell type from other cells of the monocyte/macrophage lineage.ome studies suggested that primary microglia could be distin-uished from other tissue macrophages according to expressionevels of markers such as CD11b and CD45 (Aloisi et al., 2000; Fordt al., 1995). Under healthy condition, resting microglia with typicalamified morphology show low expression levels of these com-on myeloid lineage markers (Nimmerjahn et al., 2005). However,icroglia have been shown to share expression of surface mark-

rs common to other immune cells of the macrophage family suchs CD45, CD14, MHC-class II, CD68, immunoglobulin Fc receptorsnd �2 integrins (Lambertsen et al., 2009; Wirenfeldt et al., 2005).xpression levels of these surface markers may depend on thenflammatory status of the CNS and on microglia activation. Simi-arly, functional inflammatory changes are dominated by secretionf pro-inflammatory cytokines, chemokines, neurotrophic factorsnd up-regulation of corresponding receptors, as well as produc-ion of nitric oxide and reactive oxygen intermediates (Aloisi, 2001;ambuyzer et al., 2009).

Microglia have recently been shown in a mouse model to orig-nate from a myeloid yolk sac population during embryogenesisGinhoux et al., 2010). In humans, the microglia origin is still notnown. Furthermore, little is known about the turnover rate oficroglia in the healthy CNS. It has been postulated that prolif-

ration of the resident microglia population is rather low and thereay be immigration of bone marrow derived precursor cells into

he CNS. Invasion of bone-marrow derived microglia have beenhown to migrate into the CNS using chimeric irradiated miceSimard et al., 2006; Zhang et al., 2007). However, the proportionf bone-marrow progenitor cells in replenishment of microglia isontroversial.

A variety of cytokines have been shown to contribute toicroglia development and differentiation. Colony stimulating

actor-1/M-CSF and its receptor CSF-1R play an important role inhe development of macrophage populations (Chitu and Stanley,006). Recently, Ginhoux et al. (2010) reported the constantbsence of microglia throughout life in CSF-1R deficient mice,ndicating that M-CSF is essential for microglia development andifferentiation. In addition, granulocyte-macrophage colony stim-lating factor (GM-CSF) also contributes to microglia developmentnd differentiation (Aloisi et al., 2000; Esen and Kielian, 2007).oth, M-CSF and GM-CSF have crucial effects on proliferation andurvival of primary human fetal and adult microglia in cultureith GM-CSF having a greater impact on proliferation (Esen andielian, 2007; Lee et al., 1994). M-CSF plays a crucial role in thenal maturation stage of microglia. In op/op mice, a model of humansteopetrosis with a functional mutation the M-CSF gene, the num-er of microglia are decreased and the cells are smaller and have

mpaired activation ability in response to injury (Kalla et al., 2001;egiel et al., 1998). Importantly, GM-CSF or interleukin-3 does not

ubstitute for M-CSF (Blevins and Fedoroff, 1995).

Nerve growth factor (NGF), a member of neurotrophins, has also

een shown to act on the proliferation and survival of microgliaZhang et al., 2003). NGF binds and acts through the P75 receptor,hich has been shown to be expressed in microglia in multiple

nce Methods 209 (2012) 79– 89

sclerosis lesions (Valdo et al., 2002). It has also been shown thatmicroglia express the nerve growth factor receptor TrkA (Tonchev,2011). NGF induces migration of the microglia cell through activa-tion of the TrkA (TrkA activation) (De Simone et al., 2007). NGF isexpressed by activated microglia, astrocytes and hippocampal neu-rons (Friedman, 2000; Saez et al., 2006; Tonchev, 2011). However,inflammation certainly induces neurotrophin secretion by humanmicroglia (Heese et al., 1998; Nakajima et al., 2001).

Chemokines have also been shown to play an important role inmicroglia biology. Monocyte chemoattractant protein-1 (MCP-1),or CCL2, is one of the prominent chemokines in the regulation ofthe microglia migration to the site of inflammation in experimentalmodels (Leonard et al., 1991; Zhang et al., 2007). CCL2 acts throughits specific receptor CCR2 which is expressed by microglia and othercells of monocytic lineage, such as macrophages and dendritic cells(Rebenko-Moll et al., 2006). CCL2 acts also through CCR4, but littleinformation is available about CCR4 expression in microglia (Craigand Loberg, 2006; Zhang et al., 2006). Studies on normal rat CNSshowed that CCL2 is constitutively expressed by astrocytes andneurons and it can be found in various brain regions, including thecerebral cortex, the hippocampus and the hypothalamus (Banisadret al., 2005). CCL2 is also secreted by astrocytes and neurons underinflammatory condition (Banisadr et al., 2005; Farina et al., 2007;Tanuma et al., 2006). It has been shown that CCL2 is secreted by bothundamaged and damaged spinal sensory neurons in rat, result-ing in activation of spinal microglia and initiating neuropathic-likepain (Thacker et al., 2009). Up-regulation of CCL2 expression dur-ing Alzheimer’s disease and multiple sclerosis correlates withactivation of microglia and may contribute to pathogenesis of neu-rodegenerative diseases (Conductier et al., 2010; Simpson et al.,2000a). In that respect, CCL2 contributes to recruitment of mononu-clear cells into the inflamed CNS, followed by activation of themicroglia. Zhang et al. (2007) showed that CCL2 can induce spinalmicroglia activation in mice. They also reported in chimeric micethat CCL2 recruits bone marrow-derived macrophages, which pro-liferate and differentiate into microglia in the spinal cord andinduce inflammation and microgliosis after nerve injury. In exper-imental autoimmune encephalomyelitis (EAE), a murine model ofmultiple sclerosis, CCR2 deficient mice do not develop the mononu-clear cell infiltration and inflammation. Indeed, no chemokinesincrease was detected in the CNS in that model (Izikson et al.,2000). On the other hand, CCR2 deficiency in a mouse model ofAlzheimer’s disease impairs significantly microglia accumulation atsites of plaque formation, resulting in a decrease of �-amyloid (A�)clearance and in accelerating early disease progression (El Khouryet al., 2007).

In vitro culture of mouse and human microglia has oftenbeen used as a model by various researchers. Thereby, astrocyte-conditioned medium (ACM) has been used to keep microglia inculture with a ramified resting morphology. ACM contains M-CSF,GM-CSF and transforming growth factor � (TGF �), all known to besecreted by astrocytes. Microglia lose their ramified morphology inthe presence of antibodies against the mentioned cytokines, indi-cating that M-CSF and GM-CSF are essential for microglia culturing(Schilling et al., 2001). GM-CSF, even at low concentration in serumfree medium, keeps microglia at a resting state with morphologi-cal ramifications (Fujita et al., 1996). A recent study reported theproliferative effect of CCL2 on neonatal and embryonic primary ratmicroglia in culture without inducing additional changes in mor-phology, phenotype and cytokine expression (Hinze and Stolzing,2011).

Most interestingly, murine bone marrow-derived precursor

cells can be differentiated towards microglia-like cells when cul-tured in the presence of astrocytes or in mixed glial cultures.Thereby, M-CSF alone is not sufficient for microglia differentiation,which indicates the importance of additional astrocyte-derived
Page 215: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

roscie

frsSpaet(

tHcofhacimai

bmaMrteecpoutaf

2

2

bdAup(t5g(stoctecDpdw

S. Etemad et al. / Journal of Neu

actors (Noto et al., 2010). Differentiation of murine bone mar-ow stem cells toward microglia-like cells has recently also beenuccessful, using ACM supplemented with GM-CSF (Hinze andtolzing, 2011). Culturing fetal and adult human microglia in theresence of GM-CSF induces functional maturation towards anntigen presenting cell type, especially in adult microglia. How-ver, GM-CSF does not induce maturation of microglia to acquirehe complete phenotype and function of mature dendritic cellsLambert et al., 2008; Re et al., 2002).

In vitro studies on microglia used mainly primary microglia cul-ure from embryonic or neonatal murine brains (Bassett et al., 2012;inojosa et al., 2011). In human, brain-derived microglia is diffi-ult to obtain for ethical reasons. In addition, only low numbersf cells are collected. Also, fetal microglia seems to be quite dif-erent from adult microglia. Only few human microglia cell linesave been generated, including HMO6 cells (Nagai et al., 2005)nd HMC3 cells (Janabi et al., 1995). These cell lines cannot beonsidered as an optimal model for microglia cells due the signif-cant modification in morphology and function as result of genetic

anipulation and long-term culture. Therefore, a more convenientnd more appropriate model for in vitro microglia human studiess still missing.

Most importantly, Leone et al. (2006) showed that humanlood-derived monocyte, cultured with astrocytes-conditionededium (ACM), acquired the ramified morphology of microglia

nd expressed substance P, calcium binding protein Iba1 and dimlyHCII, three typical characteristics of microglia. There are also

eports showing successful differentiation of rat blood monocytesowards microglia using ACM (Schmidtmayer et al., 1994; Sieverst al., 1994). Recently, Hinze and Stolzing (2011) showed the differ-ntiation of murine bone marrow stem cells towards microglia-likeells in the presence of ACM with similar phenotypic and functionalroperties to primary brain-derived microglia cultures. In the lightf these data, we developed a new in vitro human microglia modelsing human blood peripheral mononuclear cells, a serum-free cul-ure condition and a panel of factors, including M-CSF, GM-CSF, NGFnd CCL2. This new human microglia model has then been assessedor morphology, phenotype, and function.

. Materials and methods

.1. Cell Isolation

Human blood mononuclear cells (PBMC) were isolated fromuffy coats (50 buffy coats were used for this study) of healthyonors (Australian Red Cross Blood Service (ARCBS), Perth, WA,ustralia; ethics approval was granted by UWA and ARCBS)sing Ficoll-Paque (Amersham Biosciences, Uppsala, Sweden) asublished before (Meagher et al., 2005). To obtain monocytesadherent PBMC), the isolated blood cells were cultured in T25issue culture flasks (Sarstedt, Numbrecht, Germany) (2 × 106 to

× 106 cells/ml) using RPMI-1640 Glutamax medium (Invitro-en, Auckland, NZ) supplemented with 1% antibiotics/antimycotic10,000 units/ml penicillin G sodium, 10,000 �g/ml streptomycinulfate and 25 �g/ml Amphotericin B, Invitrogen). It is importanto note that no serum was used in all microglia cultures. Aftervernight incubation (humidified air, 37 ◦C, 5% CO2), non-adherentells (consisting mainly of T-lymphocytes) were separated byhorough washing with PBS (Invitrogen) and used for furtherxperiments. The fresh adherent cells, which were mainly mono-ytes (>90%), were used for the generation of microglia (M-MG).

endritic cells (M-DC) and cultured monocytes, as well as for com-arison with the generated cell populations. All experiments wereone with cells from at least 6 different blood donors, if not other-ise stated.

nce Methods 209 (2012) 79– 89 81

2.2. Cell culture

To induce the differentiation of M-MG, adherent PBMC(about 105/ml) were cultured in 6-well tissue culture plates(Sarstedt) using RPMI-1640 Glutamax supplemented with 1%antibiotic/antimycotic (serum-free condition) and a mixture ofhuman recombinant cytokines, including M-CSF (10 ng/ml; Pepro-Tech, Rocky Hill, NJ), GM-CSF (10 ng/ml; PeproTech), beta-nervegrowth factor (NGF-�; 10 ng/ml; PeproTech) and CCL2 (100 ng/ml;PeproTech) at standard humidified culture conditions (37 ◦C, 5%CO2) for up to 14 days. The cells were used for experiments andcharacterization at different time points as indicated.

Monocytes were also cultured in RPMI-1640 Glutamax supple-mented with 1% antibiotic/antimycotic, but no serum, for up to 14days as control cell population.

Furthermore, to obtain monocyte-derived dendritic cells (M-DC), adherent PBMC were cultured in 6-well tissue culture plates(Sarstedt) using RPMI-1640 Glutamax, 1% antibiotic/antimycoticsupplemented with 10% fetal calf serum (FCS; Serana, Bunbury,WA, Australia) in the presence of human recombinant GM-CSF(10 ng/ml; PeproTech) and interleukin-4 (IL-4; 10 ng/ml; Pepro-Tech) for up to 14 days (Meagher et al., 2005). The cells were usedfor experiments at different time points, as indicated.

Human microglia clone 3 (HMC3) cells, kindly provided byProf. Karl-Heinz Krause (University of Geneva, Switzerland) werecultured in T25 tissue culture flask (Sarstedt) using DMEM/F12Glutamax medium (Invitrogen) supplemented with 15% FCS and1% antibacterial/antimycotic. The cells were passaged and used forexperiments when they reached 70–80% confluence.

2.3. Morphological studies

Morphology and viability (using trypan blue dye exclusion: cellviability was >90% for all experiments) of M-MG, M-DC and HMC3during the 2 weeks in culture were studied using an invertedLeica DM IL phase contrast microscope. Images were taken withPhotometrix CoolSNAP Fx digital camera and processed with NIS-elements F3.0 software. HMC3 and M-MG cells were also stainedfor expression of Iba1, a well-known characteristic marker formicroglia cells. For that purpose cells were grown on glass cover-slips, fixed in 1% paraformaldehyde in PBS/sucrose and incubatedwith a polyclonal rabbit anti-Iba1 (Santa Cruz biotechnology, SantaCruz, CA) and a donkey anti-rabbit Alexa Fluor 488 (Invitrogen).Negative control stains were used by omitting the primary anti-body against Iba1. Cell nuclei were stained with DAPI (1ug/ml inPBS, 4′,6-diamidine-2′-phenylindole dihydrochloride; Roche Diag-nostics, Mannheim, Germany). Imaging was done using confocalmicroscopy (Leica TCS SP2 multiphoton confocal microscope).

2.4. Flow cytometry

We studied the expression of specific surface molecules onfreshly isolated and cultured monocytes, M-MG, M-DC and HMC3using flow cytometry. Cells (105/ml per staining condition) weredetached using a scraper (Sarstedt) and washed twice with PBS.Staining was performed according to protocol recommended byBD.

A panel of directly labeled mouse anti-human antibodies,CD45-FITC, CD14-PE, CD40-PE, CD68-APC, CD80-PE,CD11c-PE,HLA-DR–PERCP, CD11b-AF488, CD11c-PE, CCR1-AF647, CCR2-AF647, CCR4-PE-Cy7, CCR5-FITC, CXCR1-PE, CXCR3-AF488 (BDPharmingen, San Jose, CA), CCR6-FITC, CCR7-APC (R&A systems,

Minneapolis, MN), NGFR-FITC (Santa Cruz Biotechnology, SantaCruz, CA) were added at the supplier’s recommended concen-tration. Corresponding labeled isotype control antibodies wereincluded in all experiments. A rabbit anti-CX3CR1 polyclonal
Page 216: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

82 S. Etemad et al. / Journal of Neuroscience Methods 209 (2012) 79– 89

d hum

aSAcwPFfl

2

eaauhTbcl(au

2

awaPsM(lopac(t

2

s

Fig. 1. Phase contrast microscopy of monocyte derived Microglia (M-MG) (A) an

ntibody was used at the final concentration of 1 �g/ml (AbDerotec, Oxford, UK), followed by a secondary donkey anti-rabbit-F 647 used at the final concentration of 1:200 (Invitrogen). Theells were incubated with antibodies for 30 min to 1 h at 4 ◦C,ashed with PBS, resuspended and fixed with 1% formaldehyde in

BS containing 2% sucrose. The fixed samples were measured with aacsCallibure (BD Biosciences). Data analysis was performed usingowjo software version 7.6.3 (Tree Star, Ashland, OR).

.5. Phagocytosis assay

Phagocytosis was assessed by measuring the uptake of tetram-thylrhodamine (TMR)-labeled Staphylococcus aureus (Invitrogen)s published before (Meagher et al., 2005). Bacterial particles weredded to the cells (107/ml) at a ratio of 1:1000 (according to man-facture’s protocol) and incubated for 4 h at 37 ◦C. Cells were thenarvested and fixed with 1% paraformaldehyde in PBS/2% sucrose.otal ingested fluorescent-labeled bacteria per cell was measuredy flow cytometry. To verify that the bacteria were located intra-ellularly, cells were cultured on glass coverslips, treated withabeled bacteria, fixed and visualized using confocal microscopyLeica TCS SP2 multiphoton confocal microscope). Dendritic cellsnd freshly isolated monocytes were used as positive control pop-lations. Untreated cells were used as negative controls.

.6. Mixed leukocyte reaction (MLR)

The T-lymphocyte stimulatory capacity and correspondingntigen-presenting cell (APC) properties of M-MG, in comparisonith M-DC and HMC3, was assessed in MLRs using a BrdU prolifer-

tion assay (Cadosch et al., 2010). Freshly processed non-adherentBMC containing mainly (>90%) T-lymphocytes (2 × 105/well) wereeeded in a round bottom 96-well plate (Sarstedt). Allogeneic-MG, M-DC and HMC3 were used as antigen-presenting cells

APC) and added at various ratios (1/20, 1/40 and 1/80) to the T-ymphocyte (4 wells for same condition). T-lymphocytes or APCnly were used as control conditions. After 5 days in culture, BrdUroliferation assays (Roche, Mannheim, Germany) were performedccording to the supplier’s protocol and the absorbance of theolorimetric reaction was measured by ELISA reader at 405 nmLabsystems Multiskan RC, Helsinki, Finland). Mean proliferation ofhe different conditions was statistically analyzed and compared.

.7. Statistics

Data were statistically analyzed through the SPSS statistics 17.0oftware. A one-way ANOVA and uni-variant linear model were

an microglia cell line HMC3 (B) after 10 days in culture. Bar indicates 100 �M.

used to compare means. The LSD post hoc test was also used todetermine the significant difference between groups. In all theanalysis p value < 0.05 were considered as statistically significant.Graphs with error bars represent the standard deviation (SD).

3. Results

3.1. Morphological and phenotypic changes of monocyte-derivedmicroglia in culture

Adherent PBMC, representing mainly the monocytic populationof blood leukocytes, were cultured for up to 2 weeks in the presenceM-CSF, GM-CSF, NGF-� and CCL2, in RPMI 1640 without addition ofserum to create optimally standardized conditions. The cells werecharacterized for morphological changes over the period of 2 weeksin culture (Fig. 1). The well established HMC3 human microgliacells line was used as comparison. After 5 days in culture, the small(10–15 �m diameter) round-shaped and semi-adhered monocyticcells expanded their cell body and developed few unbranched pro-cesses. During the second week, the cells reduced the portion of thecell body and they increased the processes in length and numberacquiring ramified forms. M-MG obtained a ramified morphologyafter 10–14 days in culture, including a polygonal or oval cell bodyand up to 7 primary processes and numerous fine secondary pro-cess or fine spines. Over the 2 weeks in culture, the cells doubledin size and increased in cell number indicating proliferation. After2 weeks, M-MG displayed a very similar morphology to the humanmicroglia cell line HMC3 and to previously described human brain-derived microglia in published studies (Leone et al., 2006).

Basic known microglia markers, including expression of Iba1and CD68, were also tested positive, supporting that the describedprotocol differentiates monocytes towards M-MG (Figs. 2 and 3).

3.2. Expression pattern of specific surface markers

After culture in the presence or absence of the correspond-ing cytokines cocktails, monocytes, M-MG, M-DC and the humanmicroglia cell line (HMC3) were tested for expression of surfacemarkers relevant to human microglia. Fig. 4 summarizes expres-sion of all relevant markers for all tested cell types, including CD45,CD14, MHC class II, CCR2 and CCR4.

Firstly, the common leukocyte antigen CD45 was tested. CD45is a characteristic marker expressed by all leukocytes. As expected,

CD45 was highly expressed in monocytes, which did not changeCD45 expression levels over the 2 weeks in culture in serum-freemedium without addition of cytokines. M-DC reduced only slightlyCD45 expression over the 2 weeks in culture. However, M-MG
Page 217: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

S. Etemad et al. / Journal of Neuroscience Methods 209 (2012) 79– 89 83

F (D). Co3

rtt

er

cIlhMrm

o2eeCoud

rlwerc

o

ig. 2. Expression of Iba1, a characteristic microglia marker in HMC3(B) and M-MG

0 �M.

educed gradually CD45 expression levels over the 2 weeks in cul-ure reaching very low levels, as expected for microglia and similaro HMC3 (Janabi et al., 1995; Li et al., 2009).

CD14 is known to be characteristic for monocytes, beingxpressed at highest level on these cells. As predicted, M-MGeduced expression of CD14 to very low levels, similar to HMC3.

Activated microglia may process and present antigens on MHClass II. For that purpose, activated microglia increase MCH classI expression, such as HLA-DR. Usually, resting microglia expressow levels of HLA DR, whereas freshly isolated monocytes expressigh levels of HLA-DR, which is also highly expressed in immature-DC. In our study, HLA-DR was clearly down-regulated in M-MG,

eaching similar levels detected in HMC3, indicating that M-MGay represent resting microglia cells.Chemokines play a crucial role in recruitment and maturation

f migratory, bone marrow-derived microglia cells (Flynn et al.,003). CCR1, CCR2, CCR5 expression has been thus reported to bexpressed at low levels in microglia (Bajetto et al., 2002; Skulject al., 2011). In our study, monocytes expressed low levels ofCR2, but up-regulation was seen when cultured in medium with-ut cytokines. Interestingly, M-DC decreased CCR2 expression tondetectable levels, whereas M-MG and HMC3 expressed clearlyetectable CCR2 at rather low levels, as expected for microglia.

Expression of CCR4 and their ligand (CCL22 and CCL17) has beeneported in the CNS (Columba-Cabezas et al., 2002). Freshly iso-ated monocyte expressed CCR4 at very low levels, while after one

eek in culture without cytokines, a sub-population (about 25%)xpressed CCR4 at higher levels. However, M-DC and M-MG up

egulated CCR4 expression to clearly detectable levels over time inulture to similar levels as HMC3.

Fig. 5 summarizes expression of various chemokine receptorsn the different cell populations that were investigated. Freshly

rresponding negative staining controls have been included (A and B). Bar indicates

isolated monocytes expressed intermediate levels of CCR1. Dif-ferentiation towards M-MG decreased CCR1 expression to similarlevels known for non-activated microglia and also seen in HMC3.CCR5 expression level was detected at low levels in monocytes, butexpressed at higher levels in both M-MG and HMC3, as expectedfor microglia. CXCR1 was expressed at very low levels in freshlyisolated monocyte, but was clearly expressed at higher levels inM-MG and HMC3.

Freshly isolated monocyte did not express neither CXCR3 norCX3CR1. However, both these markers were clearly up-regulatedin M-MG similar to HMC3. CXCR3 expression in M-MG was atintermediate levels, while HMC3 showed higher expression levels.Interestingly, almost similar expression of CX3CR1 was seen in bothM-MG and HMC3. In summary, M-MG and HMC3 expressed verysimilar chemokine receptor patterns, supporting the microglia-like properties of M-MG.Additional markers were also investigated(data not shown), including CD 11c, CD11b, CD40, CD80, CD83,CCR6 and CCR7. M-MG were negative for CCR6 and CCR7. CD11cand CD11b were both expressed on M-MG and HMC3, but at muchlower levels than on M-DC. Immature M-DC expressed the den-dritic cell markers CD40 and CD83 at an intermediate intensitylevel. However, CD80 expression was undetectable on M-DC. Mostimportant, neither M-MG nor HMC3 expressed any of the den-dritic cell specific surface markers, such as CD83, CD80 or CD40,supporting that M-MG are clearly different from dendritic cells.

3.3. Phagocytic capacity

Microglia play a major role in phagocytosis and clearance ofdead cells and debris, especially during inflammatory processescaused by injuries or neurodegenerative diseases. To determinethe phagocytic ability, we conducted phagocytosis experiments

Page 218: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

84 S. Etemad et al. / Journal of Neuroscie

Fig. 3. Expression of CD68 in freshly isolated blood derived monocytes (A), HMC3(B) and M-MG (C). The grey line indicates the negative staining control using a corre-sponding isotype control antibody. The black line indicates the specific test stain forCD68. Note that there is no CD68 expression in monocytes, whereas the 2 microgliapopulations clearly express CD68.

nce Methods 209 (2012) 79– 89

(Meagher et al., 2005). M-MG were compared to immature M-DC,potent phagocytic immune cells (Filgueira et al., 1996). HMC3 wereused as microglia control cells. Fluorescent-labeled S. aureus wasused for measuring the phagocytic capacity of the various cell pop-ulations. Flow cytometry (Fig. 6) and confocal microscopy (Fig. 7)was used for the detection and quantification of bacterial up-take.As expected, M-DC displayed the highest phagocytosis capacity,whereas M-MG showed clearly decreased phagocytosis, similar toHMC3, which displayed even lower bacterial up-take. However,both M-MG and HMC3 showed up-take of bacteria, with clear intra-cellular location, as documented through confocal imaging.

3.4. T-lymphocyte stimulatory capacity of M-GM

As innate immune cells of the CNS, microglia are thought tobe antigen presenting cells (APC), able to process and presentantigen to T-lymphocyte, activate them and induce their prolifer-ation. In this study, mixed leukocyte reactions (MLR) were usedto determine the T-lymphocyte stimulatory capacity of M-MGusing proliferation assays. Immature dendritic cells (M-DC) andthe microglia cell line HMC3 were used as comparison. A repre-sentative example is shown in Fig. 8. As expected, M-DC inducedthe most pronounced and significant proliferation of allogeneicT-lymphocyte. M-MG were also able to induce significant prolif-eration of allogeneic T-lymphocytes, but to a much lower degreein comparison to M-DC. No significant T-lymphocyte stimulatoryeffect was seen with HMC3, which is in accordance with previousstudies (Fischer and Reichmann, 2001; Janabi et al., 1995; Nagaiet al., 2005; Zuiderwijk-Sick et al., 2007).

4. Discussion

Microglia play crucial roles in maintaining homeostasis and con-tributing to neuroinflammation in response to any disturbances,such as injuries, toxins and pathogen (Streit, 2001, 1996). To date,in vitro studies on microglia have used mainly primary cells derivedfrom embryonic or neonatal murine and human brains (Bassettet al., 2012; Hinojosa et al., 2011). This study provides the firstsimple, standardized and reliable protocol to generate humanmicroglia-like cells from blood monocytes. The protocol takes theadvantage of a serum-free culture condition, as well as a cocktailof 4 well defined recombinant human cytokines, i.e. M-CSF, GM-CSF, NGF and CCL-2. The cells generated according to the describedprotocol display most known characteristics of human microglia,including specific morphology, phenotype and functions.

In our model, the serum-free culture system is essential as itrepresents the physiological condition of the extracellular fluid ofthe CNS, which is equal to the cerebrospinal fluid. Normal cere-brospinal fluid (CSF) contains extremely low levels of proteins andbioactive factors and is usually deficient in blood serum proteins(Reiber, 2003). The blood–brain barrier prevents trafficking of bloodserum proteins and other factors into the extracellular space of theCNS, as they may damage glial and neuronal cells through activa-tion, resulting in apoptosis (Abbott et al., 2010; Hooper et al., 2009;Nadal et al., 1995). For example, in vitro culture of rat microgliain presence of bovine serum albumin, human or rat serum hasbeen shown to enhance the production of superoxide (O2

−) asone of the mechanisms involved in pathogenesis of neuronal dam-ages (Elkabes et al., 1998). Knowing about the potential bio-activeand even toxic effects of blood serum, we established thereforethis serum-free protocol for the generation of microglia-like cells

derived from blood monocytes.The cocktail of cytokines has beenchosen according to the known effects of the cytokines on microgliain published reports, as stated in the introduction, and exten-sive preliminary experiments. Thereby, the combination of M-CSF,
Page 219: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

S. Etemad et al. / Journal of Neuroscience Methods 209 (2012) 79– 89 85

F CCR4c lls (C)c

Gm

rsoin

ig. 4. Expression of surface markers CD45 (a), CD14 (b), HLA-DR (c), CCR2 (d) andultured monocytes in the absence of cytokines (B), monocyte derived dendritic ceultured for 10 days.

M-CSF, NGF and CCL-2 has shown to differentiate M-MG withost similar characteristics to microglia.Under healthy condition, resting “surveying microglia” with

amified morphology is responsible for maintaining the homeo-

tatic condition of the CNS. Using the described protocol, M-MGbtained a ramified morphology after 10–14 days in culture, includ-ng a polygonal or oval cell body and up to 7 primary processes andumerous fine secondary process or fine spines. Our morphological

(e) measured with flow cytometry: comparison of freshly isolated monocytes (A),, monocyte derived microglia (D) and microglia cell line HMC3 (E). The cells were

findings for M-MG correspond to the report by Leone et al. (2006),who used a less well defined protocol. In addition, M-MG displayedsimilar morphology to the human microglia cell line HMC3. Inaddition, M-MG expressed Iba1 and CD68, well known microglia

markers.

Most important was the phenotypic characterization of thecells using a broad panel of antibodies and flow cytometry. Mono-cytes cultured over 2 weeks in the presence of the 4 cytokines

Page 220: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

86 S. Etemad et al. / Journal of Neuroscience Methods 209 (2012) 79– 89

Fig. 5. Expression of chemokine receptors CCR1 (a), CCR5 (b), CXCR1(c), CXCR3 (d) andmonocytes (A), monocyte derived microglia (B) and the microglia cell line HMC3 (C). The

Fig. 6. Phagocytic capacity of M-DC (black line), M-MG (dotted line) and HMC3(dashed line) towards TM Rhodamine-labeled S. aureus measured with flow cytom-etry. Bacterial particles were added to culture medium at a ratio of 1:1000 (107

bacteria per ml) and incubated for 4 h at 37 ◦C. Flow cytometry data represented thedifferent levels of engulfed fluorescent-labeled bacteria.

CX3CR1 (e) measured with flow cytometry: expression levels in freshly isolated cells were cultured for 10 days.

changed significantly expression of surface markers towards theknown pattern for microglia. CD45, CD14 and HLA-Dr expressionlevels decreased substantially in M-MG reaching levels expectedfor microglia. CD45 expression levels have been often used fordiscriminating between tissue macrophages and microglia in CNSstudies (Lambertsen et al., 2009; Wirenfeldt et al., 2005). Penningeret al. (2001) states that in the CNS, microglia express differ-ent levels of CD45 depending on their differentiation state, withdecreased expression levels in mature resting microglia. How-ever, CD45 expression increases in activated microglia, e.g. inAlzheimer patients, as this surface marker seems to play a rolein degenerative and inflammatory CNS diseases (Masliah et al.,1991). CD 14 is regarded as an important lipopolysaccharide (LPS)receptor (Triantafilou and Triantafilou, 2002). Supporting our pro-tocol, Lambert et al. (2008) reported down-regulation of CD14 inmonocyte-derived DC and microglia in presence of GM-CSF, M-CSFand LPS.

Activated microglia is considered as the antigen presenting cellsof the CNS, as they express MHC class II molecules. However, rest-ing microglia express low levels of MHC class II. M-MG decreasedHLA-DR expression to low levels, similar to HMC3, but to muchlower levels than freshly isolated monocytes or immature M-DC(Lutz et al., 2000). Furthermore, it was important to discriminate

between M-MG and M-DC, as both may display similar morpholog-ical properties. In addition to the differences in expression levels ofMHC class II, CD11b and CD11c, M-MG did not express characteris-tic M-DC markers, including CD40, CD80 and CD83. The differences
Page 221: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

S. Etemad et al. / Journal of Neuroscience Methods 209 (2012) 79– 89 87

F -stacb eria in

btdtpM

c

Foccac

ig. 7. Up-take of TM Rhodamine-labeled S. aureus by M-MG using on a selected Zacteria at a ration 1:1000. Note the intra-cellular accumulation of red labeled bact

etween M-MG and M-DC were further supported by the functionalests, such as MLR and phagocytosis assays. As expected, M-MGisplayed much lower T-lymphocyte stimulatory capacity and bac-erial up-take, in comparison with M-DC. In summary, our datarovide clear evidence that our M-MG protocol does not generate

-DC, but a distinct microglia-like population.Our study also revealed the expression level of various

hemokine receptors, including CCR1, CCR2, CCR4, CCR5, CXCR1,

ig. 8. Mixed leukocyte reactions (MLR) testing T-lymphocyte stimulatory capacityf HMC3 (black bars), M-MG (dotted bars) and M-DC (hatched bars) at differentell ratios (antigen presenting cells: allogeneic T-lymphocytes). The cells were co-ultured for 5 days before proliferation was measured using a BrdU incorporationssay. Error bars represent the ±2 SD. Statistically significant differences betweenontrol and test condition are indicated by *p < 0.05.

k image taken with confocal microscopy. M-MG cells were incubated for 4 h with the M-MG cells on the optical confocal section.

CXCR3 and CX3CR1 in M-MG and HMC3. Freshly isolatedmonocytes showed intermediate expression level of CCR1, lowexpression levels for CCR2, CXCR1 and CCR5, and undetectableexpression levels of CCR4, CXCR3 and CX3CR1, as reported previ-ously by others (Geissmann et al., 2003). M-MG changed expressionlevels of the various chemokine receptors substantially, in compari-son with freshly isolated monocytes. However, expression patternsof M-MG were very similar to that of HMC3. M-MG showeddown-regulation of CCR1 to still well detectable levels. On theother hand, M-MG displayed significant up-regulation of CCR4 andCCR5. Interestingly, M-MG expressed CXCR3 and CX3CR1, whichwere not detected in monocytes. Up-regulation of CXCR1 was alsoseen in M-MG. Our findings are in accordance with the report byBajetto et al. (2002), who published that non-activated microgliaexpress CCR1, CCR2 and CCR5 constitutively at low levels. Sev-eral other researchers reported that activated microglia expresshigher level of CCR2, CCR3, CCR4, CCR5, CX3CR1 (Eltayeb et al.,2007; Simpson et al., 2000b; Szczucinski and Losy, 2007). However,little is known about chemokine receptor expression in humanmicroglia. Our protocol may help in future research to elucidateregulation of these receptors and chemokine responses in humanmicroglia.

5. Conclusion

We developed a new in vitro protocol for the generation ofhuman microglia from blood monocytes using a serum-free culturecondition and a novel mixture of 4 recombinant human cytokines,i.e. M-CSF, GM-CSF, NGF and CCL2. Detailed characterization of M-MG revealed a cell population representing resting microglia withtheir specific morphological, phenotypic and functional properties.The described protocol is easy to handle, well standardized and

very reproducible, as it uses only well defined culture medium andrecombinant cytokines, but no serum or conditioned medium. Thisprotocol will certainly be very helpful for future studies investigat-ing the biology and pathology of human microglia.
Page 222: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

8 roscie

C

A

PAat&CaeHb

R

A

AA

B

B

B

B

C

C

C

C

C

D

E

E

E

E

F

F

F

F

F

8 S. Etemad et al. / Journal of Neu

onflict of interest statement

None of the authors have any conflict of interest to disclose.

cknowledgements

Microscopes were provided by Cell Central, School of Anatomy,hysiology and Human Biology. In that respect, we thank Guy Ben-ry and Steve Parkinson for their excellent support. The authorscknowledge the facilities (Confocal microscopy and flow cytome-ry), scientific and technical assistance of the Australian Microscopy

Microanalysis Research Facility at the Centre for Microscopy,haracterisation & Analysis, The University of Western Australia,

facility funded by the University, State and Commonwealth Gov-rnments. In that respect, we acknowledge the support by Kathyeel, Paul Rigby and Tracey Lee-Pullen. The project was supportedy a UWA Collaboration Award.

eferences

bbott NJ, Patabendige AAK, Dolman DEM, Yusof SR, Begley DJ. Structure and func-tion of the blood–brain barrier. Neurobiol Dis 2010;37:13–25.

loisi F. Immune function of microglia. Glia 2001;36:165–79.loisi F, De Simone R, Columba-Cabezas S, Penna G, Adorini L. Functional maturation

of adult mouse resting microglia into an APC is promoted by granulocyte-macrophage colony-stimulating factor and interaction with Th1 cells. J Immunol2000;164:1705–12.

ajetto A, Bonavia R, Barbero S, Schettini G. Characterization of chemokines andtheir receptors in the central nervous system: physiopathological implications.J Neurochem 2002;82:1311–29.

anisadr G, Gosselin R-D, Mechighel P, Kitabgi P, Rostène W, ParsadaniantzSM. Highly regionalized neuronal expression of monocyte chemoattractantprotein-1 (MCP-1/CCL2) in rat brain: evidence for its colocalization with neuro-transmitters and neuropeptides. J Comp Neurol 2005;489:275–92.

assett T, Bach P, Chan HM. Effects of methylmercury on the secretion ofpro-inflammatory cytokines from primary microglial cells and astrocytes. Neu-roToxicology 2012;33:229–34.

levins G, Fedoroff S. Microglia in colony-stimulating factor 1-deficient op/op mice.J Neurosci Res 1995;40:535–44.

adosch D, Sutanto M, Chan E, Mhawi A, Gautschi OP, von Katterfeld B, et al. Titaniumuptake, induction of RANK-L expression, and enhanced proliferation of humanT-lymphocytes. J Orthop Res 2010;28:341–7.

hitu V, Stanley ER. Colony-stimulating factor-1 in immunity and inflammation.Curr Opin Immunol 2006;18:39–48.

olumba-Cabezas S, Serafini B, Ambrosini E, Sanchez M, Penna G, Adorini L, et al.Induction of macrophage-derived chemokine/CCL22 expression in experimen-tal autoimmune encephalomyelitis and cultured microglia: implications fordisease regulation. J Neuroimmunol 2002;130:10–21.

onductier G, Blondeau N, Guyon A, Nahon J-L, Rovère C. The role of monocytechemoattractant protein MCP1/CCL2 in neuroinflammatory diseases. J Neuroim-munol 2010;224:93–100.

raig MJ, Loberg RD. CCL2 (Monocyte Chemoattractant Protein-1) in cancer bonemetastases. Cancer Metastasis Rev 2006;25:611–9.

e Simone R, Ambrosini E, Carnevale D, Ajmone-Cat MA, Minghetti L. NGF pro-motes microglial migration through the activation of its high affinity receptor:modulation by TGF-[beta]. J Neuroimmunol 2007;190:53–60.

l Khoury J, Toft M, Hickman SE, Means TK, Terada K, Geula C, et al. Ccr2 deficiencyimpairs microglial accumulation and accelerates progression of Alzheimer-likedisease. Nat Med 2007;13:432–8.

lkabes S, Peng L, Black IB. Lipopolysaccharide differentially regulates microglial trkreceptor and neurotrophin expression. J Neurosci Res 1998;54:117–22.

ltayeb S, Berg A-L, Lassmann H, Wallstrom E, Nilsson M, Olsson T, et al. Temporalexpression and cellular origin of CC chemokine receptors CCR1, CCR2 and CCR5in the central nervous system: insight into mechanisms of MOG-induced EAE. JNeuroinflamm 2007;4:14.

sen N, Kielian T. Effects of low dose GM-CSF on microglial inflammatory profilesto diverse pathogen-associated molecular patterns (PAMPs). J Neuroinflamm2007;4:10.

arina C, Aloisi F, Meinl E. Astrocytes are active players in cerebral innate immunity.Trends Immunol 2007;28:138–45.

ilgueira L, Nestle F, Rittig M, Joller H, Groscurth P. Human dendritic cells phagocy-tose and process Borrelia burgdorferi. J Immunol 1996;157:2998–3005.

ischer H-G, Reichmann G. Brain dendritic cells and macrophages/microglia in cen-tral nervous system inflammation. J Immunol 2001;166:2717–26.

lynn G, Maru S, Loughlin J, Romero IA, Male D. Regulation of chemokinereceptor expression in human microglia and astrocytes. J Neuroimmunol2003;136:84–93.

ord A, Goodsall A, Hickey W, Sedgwick J. Normal adult ramified microglia separatedfrom other central nervous system macrophages by flow cytometric sorting.

nce Methods 209 (2012) 79– 89

Phenotypic differences defined and direct ex vivo antigen presentation to myelinbasic protein-reactive CD4+ T cells compared. J Immunol 1995;154:4309–21.

Friedman WJ. Neurotrophins induce death of hippocampal neurons via the p75receptor. J Neurosci 2000;20:6340–6.

Fujita H, Tanaka J, Toku K, Tateishi N, Suzuki Y, Matsuda S, et al. Effects of GM-CSF and ordinary supplements on the ramification of microglia in culture: amorphometrical study. Glia 1996;18:269–81.

Geissmann F, Jung S, Littman DR. Blood monocytes consist of two principal subsetswith distinct migratory properties. Immunity 2003;19:71–82.

Ginhoux F, Greter M, Leboeuf M, Nandi S, See P, Gokhan S, et al. Fate mapping anal-ysis reveals that adult microglia derive from primitive macrophages. Science2010;330:841–5.

Heese K, Hock C, Otten U. Inflammatory signals induce neurotrophin expression inhuman microglial cells. J Neurochem 1998;70:699–707.

Hinojosa AE, Garcia-Bueno B, Leza JC, Madrigal JL. CCL2/MCP-1 modulation ofmicroglial activation and proliferation. J Neuroinflamm 2011;8:77.

Hinze A, Stolzing A. Differentiation of mouse bone marrow derived stem cells towardmicroglia-like cells. BMC Cell Biol 2011;12:35.

Hooper C, Pinteaux-Jones F, Fry VAH, Sevastou IG, Baker D, Heales SJ, et al. Differentialeffects of albumin on microglia and macrophages; implications for neurodegen-eration following blood–brain barrier damage. J Neurochem 2009;109:694–705.

Howell OW, Rundle JL, Garg A, Komada M, Brophy PJ, Reynolds R. Acti-vated microglia mediate axoglial disruption that contributes to axonalinjury in multiple sclerosis. J Neuropathol Exp Neurol 2010;69:1017–33,http://dx.doi.org/10.1097/NEN.0b013e3181f3a5b1.

Izikson L, Klein RS, Charo IF, Weiner HL, Luster AD. Resistance to experimentalautoimmune encephalomyelitis in mice lacking the cc chemokine receptor(Ccr2). J Exp Med 2000;192:1075–80.

Janabi N, Peudenier S, Héron B, Ng KH, Tardieu M. Establishment of human microglialcell lines after transfection of primary cultures of embryonic microglial cells withthe SV40 large T antigen. Neurosci Lett 1995;195:105–8.

Kalla R, Liu Z, Xu S, Koppius A, Imai Y, Kloss CUA, et al. Microglia and the earlyphase of immune surveillance in the axotomized facial motor nucleus: impairedmicroglial activation and lymphocyte recruitment but no effect on neuronal sur-vival or axonal regeneration in macrophage-colony stimulating factor-deficientmice. J Comp Neurol 2001;436:182–201.

Lambert C, Desbarats J, Arbour N, Hall JA, Olivier A, Bar-Or A, et al. Dendritic celldifferentiation signals induce anti-inflammatory properties in human adultmicroglia. J Immunol 2008;181:8288–97.

Lambertsen KL, Clausen BH, Babcock AA, Gregersen R, Fenger C, Nielsen HH, et al.Microglia protect neurons against ischemia by synthesis of tumor necrosis fac-tor. J Neurosci 2009;29:1319–30.

Lee S, Varvel NH, Konerth ME, Xu G, Cardona AE, Ransohoff RM, et al. CX3CR1 defi-ciency alters microglial activation and reduces beta-amyloid deposition in twoAlzheimer’s disease mouse models. Am J Pathol 2010;177:2549–62.

Lee SC, Liu W, Brosnan CF, Dickson DW. GM-CSF promotes proliferation of humanfetal and adult microglia in primary cultures. Glia 1994;12:309–18.

Leonard EJ, Skeel A, Yoshimura T. Biological aspects of monocyte chemoattractantprotein-1 (MCP-1). Adv Exp Med Biol 1991;305:57–64.

Leone C, Le Pavec G, Même W, Porcheray F, Samah B, Dormont D, et al. Characteri-zation of human monocyte-derived microglia-like cells. Glia 2006;54:183–92.

Li B, Bedard K, Sorce S, Hinz B, Dubois-Dauphin M, Krause KH. NOX4 expression inhuman microglia leads to constitutive generation of reactive oxygen species andto constitutive IL-6 expression. J Innate Immun 2009;1:570–81.

Lutz MB, Suri RM, Niimi M, Ogilvie AL, Kukutsch NA, Rossner S, et al. Immaturedendritic cells generated with low doses of GM-CSF in the absence of IL-4are maturation resistant and prolong allograft survival in vivo. Eur J Immunol2000;30:1813–22.

Masliah E, Mallory M, Hansen L, Alford M, Albright T, Terry R, et al. Immunoreactivityof CD45, a protein phosphotyrosine phosphatase, in Alzheimer’s disease. ActaNeuropathol 1991;83:12–20.

Meagher J, Zellweger R, Filgueira L. Functional dissociation of the basolateral tran-scytotic compartment from the apical phago-lysosomal compartment in humanosteoclasts. J Histochem Cytochem 2005;53:665–70.

Nadal A, Fuentes E, Pastor J, McNaughton PA. Plasma albumin is a potent trig-ger of calcium signals and DNA synthesis in astrocytes. Proc Natl Acad Sci1995;92:1426–30.

Nagai A, Mishima S, Ishida Y, Ishikura H, Harada T, Kobayashi S, et al. Immor-talized human microglial cell line: phenotypic expression. J Neurosci Res2005;81:342–8.

Nakajima K, Honda S, Tohyama Y, Imai Y, Kohsaka S, Kurihara T. Neurotrophinsecretion from cultured microglia. J Neurosci Res 2001;65:322–31.

Nimmerjahn A, Kirchhoff F, Helmchen F. Resting microglial cells are highly dynamicsurveillants of brain parenchyma in vivo. Science 2005;308:1314–8.

Noto D, Takahashi K, Miyake S, Yamada M. In vitro differentiation of lineage-negativebone marrow cells into microglia-like cells. Eur J Neurosci 2010;31:1155–63.

Penninger JM, Irie-Sasaki J, Sasaki T, Oliveira-dos-Santos AJ. CD45: new jobs for anold acquaintance. Nat Immunol 2001;2:389–96.

Politis M, Pavese N, Tai YF, Kiferle L, Mason SL, Brooks DJ, et al. Microglialactivation in regions related to cognitive function predicts disease onset inHuntington’s disease: a multimodal imaging study. Hum Brain Mapp 2011;32:

258–70.

Re F, Belyanskaya SL, Riese RJ, Cipriani B, Fischer FR, Granucci F, et al. Granulocyte-macrophage colony-stimulating factor induces an expression program inneonatal microglia that primes them for antigen presentation. J Immunol2002;169:2264–73.

Page 223: Effects of Lead on Human Microglia and Neuronal … · microglia interaction, as well as during CNS development by mediating neurogenesis, migration and differentiation of neuronal

roscie

R

R

S

S

S

S

S

S

S

S

S

SS

S. Etemad et al. / Journal of Neu

ebenko-Moll NM, Liu L, Cardona A, Ransohoff RM. Chemokines, mononuclear cellsand the nervous system: heaven (or hell) is in the details. Curr Opin Immunol2006;18:683–9.

eiber H. Proteins in cerebrospinal fluid and blood: barriers, CSF flow rate andsource-related dynamics. Restor Neurol Neurosci 2003;21:79–96.

aez ET, Pehar M, Vargas MR, Barbeito L, Maccioni RB. Production of nerve growthfactor by beta-amyloid-stimulated astrocytes induces p75NTR-dependenttau hyperphosphorylation in cultured hippocampal neurons. J Neurosci Res2006;84:1098–106.

chilling T, Nitsch R, Heinemann U, Haas D, Eder C. Astrocyte-released cytokinesinduce ramification and outward K+ channel expression in microglia via distinctsignalling pathways. Eur J Neurosci 2001;14:463–73.

chmidtmayer J, Jacobsen C, Miksch G, Sievers J. Blood monocytes and spleenmacrophages differentiate into microglia-like cells on monolayers of astrocytes:membrane currents. Glia 1994;12:259–67.

ievers J, Parwaresch R, Wottge H-U. Blood monocytes and spleen macrophagesdifferentiate into microglia-like cells on monolayers of astrocytes: morphology.Glia 1994;12:245–58.

imard AR, Soulet D, Gowing G, Julien J-P, Rivest S. Bone marrow-derived microgliaplay a critical role in restricting senile plaque formation in Alzheimer’s disease.Neuron 2006;49:489–502.

impson J, Rezaie P, Newcombe J, Cuzner ML, Male D, Woodroofe MN. Expressionof the [beta]-chemokine receptors CCR2, CCR3 and CCR5 in multiple sclerosiscentral nervous system tissue. J Neuroimmunol 2000a;108:192–200.

impson J, Rezaie P, Newcombe J, Cuzner ML, Male D, Woodroofe MN. Expression ofthe �-chemokine receptors CCR2, CCR3 and CCR5 in multiple sclerosis centralnervous system tissue. J Neuroimmunol 2000b;108:192–200.

ˇkuljec J, Sun H, Pul R, Bénardais K, Ragancokova D, Moharregh-Khiabani D, et al.CCL5 induces a pro-inflammatory profile in microglia in vitro. Cell Immunol2011;270:164–71.

treit WJ. Microglia and macrophages in the developing CNS. Neurotoxicology

2001;22:619–24.

treit WJ. The role of microglia in brain injury. Neurotoxicology 1996;17:671–8.zczucinski A, Losy J. Chemokines and chemokine receptors in multiple scle-

rosis. Potential targets for new therapies. Acta Neurol Scand 2007;115:137–46.

nce Methods 209 (2012) 79– 89 89

Tambuyzer BR, Ponsaerts P, Nouwen EJ. Microglia: gatekeepers of central nervoussystem immunology. J Leukoc Biol 2009;85:352–70.

Tanuma N, Sakuma H, Sasaki A, Matsumoto Y. Chemokine expression by astrocytesplays a role in microglia/macrophage activation and subsequent neurodegen-eration in secondary progressive multiple sclerosis. Acta Neuropathol (Berl)2006;112:195–204.

Thacker MA, Clark AK, Bishop T, Grist J, Yip PK, Moon LDF, et al. CCL2 is a key mediatorof microglia activation in neuropathic pain states. Eur J Pain (London, England)2009;13:263–72.

Tonchev AB. The nerve growth factor in health and unhealth neurons. Arch Ital Biol2011;149:225–31.

Triantafilou M, Triantafilou K. Lipopolysaccharide recognition: CD14, TLRs and theLPS-activation cluster. Trends Immunol 2002;23:301–4.

Valdo P, Stegagno C, Mazzucco S, Zuliani E, Zanusso G, Moretto G, et al. Enhancedexpression of NGF receptors in multiple sclerosis lesions. J Neuropathol ExpNeurol 2002;61:91–8.

Wegiel J, Wisniewski HM, Dziewiatkowski J, Tarnawski M, Kozielski R, Trenkner E,et al. Reduced number and altered morphology of microglial cells in colony stim-ulating factor-1-deficient osteopetrotic op/op mice. Brain Res 1998;804:135–9.

Wirenfeldt M, Ladeby R, Dalmau I, Banati RB, Finsen B. Microglia – Biologyand relevance to disease. Mikroglia – Biologi og sygdomsmæssig relevans2005;167:3025–30.

Zhang J, Geula C, Lu C, Koziel H, Hatcher LM, Roisen FJ. Neurotrophins regu-late proliferation and survival of two microglial cell lines in vitro. Exp Neurol2003;183:469–81.

Zhang J, Shi XQ, Echeverry S, Mogil JS, De Koninck Y, Rivest S. Expression of CCR2in both resident and bone marrow-derived microglia plays a critical role inneuropathic pain. J Neurosci 2007;27:12396–406.

Zhang T, Somasundaram R, Berencsi K, Caputo L, Gimotty P, Rani P, et al. Migra-tion of cytotoxic T lymphocytes toward melanoma cells in three-dimensionalorganotypic culture is dependent on CCL2 and CCR4. Eur J Immunol 2006;36:

457–67.

Zuiderwijk-Sick EA, van der Putten C, Bsibsi M, Deuzing IP, de Boer W, Persoon-Deen C, et al. Differentiation of primary adult microglia alters their responseto TLR8-mediated activation but not their capacity as APC. Glia 2007;55:1589–600.