groningen international program of science in medicine...

40
Sepsis-induced acute kidney injury The role of the P2X7 receptor (P2X7R) in renal injury and sepsis through the local production of cytokines and chemokinesGroningen International Program of Science in Medicine (GIPS-M) Marije (ML) Sixma Supervisor London, United Kingdom Supervisor Groningen, Netherlands Professor Mervyn Singer Professor C.G.M. Kallenberg, Professor of Intensive Care Medicine, Head Professor Department of Rheumatology Research Department of Clinical Physiology and Clinical Immunology University College London University Medical Centre Groningen Division of Medicine, NIHR Senior Investigator ’09 Chairman of the GIPS-M Committee [email protected] [email protected] Co-supervisor Dr. Nish Arulkumaran, PhD at the division of Intensive Care Medicine, UCL [email protected] London, January June 2013

Upload: others

Post on 29-May-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Groningen International Program of Science in Medicine ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/... · (iii) Detection of macrophages 20 3. Enzyme-linked immunosorbent assay’s

Sepsis-induced acute kidney injury

“The role of the P2X7 receptor (P2X7R) in renal injury and sepsis through the local

production of cytokines and chemokines”

Groningen International Program of Science in Medicine (GIPS-M)

Marije (ML) Sixma

Supervisor London, United Kingdom Supervisor Groningen, Netherlands

Professor Mervyn Singer Professor C.G.M. Kallenberg,

Professor of Intensive Care Medicine, Head Professor Department of Rheumatology

Research Department of Clinical Physiology and Clinical Immunology

University College London University Medical Centre Groningen

Division of Medicine, NIHR Senior Investigator ’09 Chairman of the GIPS-M Committee

[email protected] [email protected]

Co-supervisor

Dr. Nish Arulkumaran, PhD at the division of

Intensive Care Medicine, UCL

[email protected]

London, January – June 2013

Page 2: Groningen International Program of Science in Medicine ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/... · (iii) Detection of macrophages 20 3. Enzyme-linked immunosorbent assay’s

2

Table of contents

ABBREVIATIONS 4

ENGLISH SUMMARY 5

INTRODUCTION

Sepsis and acute kidney injury 7

Current tools and limitations 7

Haemodynamic and histopathological findings 8

Inflammation and the inflammasome 9

The NLRP3 pathway 10

P2X7 receptor 11

Macrophages and monocyte chemoattractant protein 1 12

Aim of study 12

MATERIAL AND METHODS

1. In vivo methods 13

(i). Rat model of acute sepsis

(ii). Rat model of sepsis and recovery

2. Homogenization of Frozen Kidney Tissues 14

3. Bicinchoninic acid (BCA) protein estimation assay 14

4. Western blotting 14

5. Immunohistochemistry 15

6. Enzyme-linked immunosorbent assays (ELISAs) 15

7. Statistics 16

RESULTS

Polymicrobial model of acute sepsis

1. Analysis of renal dysfunction by use of serum creatinine 17

2. Immunohistochemistry

(i) Assessment of histological damage 17

(ii) P2X7 expression in kidney tissue 18

(iii) Detection of macrophages 20

3. Enzyme-linked immunosorbent assay’s

(i) Expression of serum and renal IL-1β 20

(ii) Expression of serum and renal MCP-1 21

4. Western Blotting 22

Recovery model of sepsis

1. Immunohistochemistry 23

3. ELISA; expression of renal MCP-1 24

DISCUSSION 25

CONCLUSIONS 27

FUTURE WORK 27

Page 3: Groningen International Program of Science in Medicine ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/... · (iii) Detection of macrophages 20 3. Enzyme-linked immunosorbent assay’s

3

DUTCH SUMMARY 29

ACKNOWLEDGMENTS 31

REFERENCES 32

APPENDICES

I. Buffers and solutions 36

ABSTRACTS 37

1. “P2X7 Receptor and Haemorrhage-reperfusion- Induced Acute Tubular Injury”

2. “Renal macrophage infiltration in a rat model of sepsis and recovery”

3. “Temporal changes in renal haemodynamics and oxygenation in a rat model of sepsis”

Page 4: Groningen International Program of Science in Medicine ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/... · (iii) Detection of macrophages 20 3. Enzyme-linked immunosorbent assay’s

4

Abbreviations

MOF multi organ failure

AKI acute kidney injury

ICU intensive care unit

RRT renal replacement therapy

ADQI Acute Dialysis Quality Initiative

RIFLE Risk- Injury- Failure- Loss and End stage renal disease criteria

sCr serum creatinine

AKIN Acute Kidney Injury Network

KDIGO Kidney Disease: Improving Global Outcomes guidelines

GFR glomerular filtration rate

ARF acute renal failure

RBF renal blood flow

MAP mean arterial pressure

ATN acute tubular necrosis

TECs tubular epithelial cell

LPS lipopolysaccharide

PRR pattern recognition receptor

PAMPs pathogen-associated molecular patterns

DAMPs danger-associated molecular patterns

ATP adenosine 5’-triphosphate

TLR toll-like receptor

NFkB nuclear factor kappa

IL-1β interleukin-1β

TNF-α tumor necrosis factor-α

COX-2 cyclooxygenase-2

NLRP-3 nod-like receptor protein 3

P2X7R P2X7 receptor

IFN-γ interferon-γ

mRNA messenger ribonucleic acid

NTN nephrotoxic nephritis

MCP-1 monocyte chemoattractant protein 1

CKD chronic kidney disease

NGAL neutrophil gelatinase-associated lipocalin

KIM-1 kidney injury molecule-1

SDS-PAGE Sodium dodecylsulphate-Polyacrylamide gel electrophoresis

PVDF Polyvinylidene difluoride

ELISA Enzyme-linked immunosorbent assay

PAS Periodic acid–Schiff (PAS)

ICE interleukin-1β converting enzyme

MAP kinase mitogen-activated protein kinase

Page 5: Groningen International Program of Science in Medicine ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/... · (iii) Detection of macrophages 20 3. Enzyme-linked immunosorbent assay’s

5

Summary

Introduction

Sepsis is an exaggerated inflammatory response to infection that may lead to multiple organ

failure and death. The kidney is commonly affected in the septic process leading to

dysfunction or complete failure. Of note, survivors of sepsis-induced acute kidney injury

(AKI) have an increased risk of developing end-stage chronic kidney disease. Release of pro-

inflammatory cytokines such as IL-1β and IL-18 are associated with the pro-inflammatory

cascade and immunological factors are likely to contribute to the development of sepsis-

induced AKI. However, initial release of these mediators is, in itself, insufficient. A

considerable role for the post-transcriptional processing and release of mature cytokines is via

the formation of a large multiprotein complex, the NLRP3 inflammasome. The cell surface

P2X7 receptor (P2X7R) facilitates assembly of NLRP3. As well as mediating inflammatory

responses, the P2X7R is also involved in apoptotic cell death. Upregulation of this receptor is

associated with renal injury. Moreover, A-438079, a selective P2X7 antagonist, attenuated this

response in a rat model of glomerulonephritis. These findings suggest that the P2X7R could

function as a potential therapeutic target. P2X7 expression is best known in immune cells such

as macrophages which play a critical role in the initiation, maintenance, and resolution of

inflammation. Macrophages both release and are influenced by chemokines such as MCP-1

that play a prominent role in the acute inflammatory response in several models of kidney

disease. Moreover, IL-1 β (after maturation and release by the P2X7/inflammasome) induces

expression of MCP-1 by epithelial cells.

Aims

With a working hypothesis that P2X7R upregulation causes renal injury through local

production of cytokines and chemokines, I sought to determine whether renal P2X7R

expression increases in a fluid-resuscitated three-day rat model of faecal peritonitis, and

specifically to define its relationship and co-localization to histopathological changes and

macrophage infiltration. In a separate longer term (2-week) rat model of zymosan-induced

peritonitis, I sought to describe renal P2X7R expression and pathological changes from the

early stages into the recovery phase.

Methods

For the acute model, sepsis was induced in instrumented, awake male Wistar rats by

intraperitoneal injection of faecal slurry followed by fluid resuscitation at 10 ml/kg/hr

commencing 2 hours post-induction of sepsis. Sham animals received a similar fluid regimen

but no i.p. injection. Septic and sham-operated animals were sacrificed post-sepsis at either 6

hours (n = 6 per group) or 24 hours (n= 8 per group). Kidneys were harvested and blood

analyzed for serum creatinine, histological evidence of renal injury, P2X7 expression,

macrophage infiltration, and cytokine and chemokine expression. Comparison was made

against naïve, non-instrumented animals (n=6).

In the long-term model, non-instrumented male Wistar rats were given i.p. zymosan (a yeast

cell wall product) as a more prolonged septic insult. Kidneys were harvested at day 2 (4 sham,

4 sepsis animals) and day 14 (4 sham, 8 sepsis), and analyzed for renal injury, macrophage

infiltration and chemokine expression.

Page 6: Groningen International Program of Science in Medicine ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/... · (iii) Detection of macrophages 20 3. Enzyme-linked immunosorbent assay’s

6

Continuous variables are presented as mean ± SD. Parametric and non-parametric data were

compared by unpaired t-test and Mann Whitney U tests, respectively. One-way ANOVA

assessed differences between more than two groups of continuous variables. Post-hoc Tukey’s

test ascertained between-group differences with P values <0.05 being taken as statistically

significant.

Results

In the acute septic AKI model, serum creatinine was significantly higher at 24 hours (p<0.05).

However, no significant renal histological damage was seen at either 6 or 24 hour timepoints.

In septic kidneys, P2X7R expression increased at 24 hours (p< 0.05) though a trend was

apparent even at 6 hours. Severely septic animals (with a 3-fold increase in serum creatinine)

showed evidence of intraluminal debris that stained intensely for P2X7. Increased expression

of P2X7 correlated with elevated serum and renal tissue IL-1β levels. Serum IL-1β were

elevated in septic animals, though levels were falling by 24 hours (p<0.05). By contrast, renal

tissue IL-1β levels were elevated at 6 hours after sepsis (p=0.05) but further increased at 24

hours (p<0.01). Consistent with these findings, septic AKI was associated with increased

renal expression of caspase-1 and pro-IL-18. No macrophage infiltration was seen at either

timepoint and there was only minimal elevation in renal MCP-1 levels.

In the long-term model of sepsis, there was also minimal evidence of acute renal injury at

Days 2 or 14. P2X7 staining has yet to be analyzed; however, macrophage infiltration was

evident in both glomeruli and interstitium in all Day 14 septic animals but in none of the sham

controls. Renal MCP-1 levels were also significantly elevated in septic animals (p<0.01) at

this timepoint. By contrast, macrophage infiltration was only seen in one of the four septic

animals on Day 2 and renal MCP-1 levels were similar to sham animals.

Conclusions

A significant increase in renal P2X7 expression was seen in septic animals after 24 hours,

correlating with elevated levels of IL-1β. In severely septic animals more renal injury was

apparent with a distinct pattern in staining, suggestive of a role in cell death. This finding,

together with an absence of early renal macrophage infiltration, suggests renal P2X7 is likely

to be expressed by intrinsic kidney cells. While significant histological damage was not seen,

reflecting findings in septic patients, there was biochemical evidence of renal dysfunction.

Further research is needed to evaluate whether P2X7 antagonism offers protection. By

contrast, the longer term model (in which P2X7 staining has not yet been analyzed) did show

significant renal macrophage infiltration within the glomeruli and interstitium with a

corresponding increase in renal MCP-1 at day 14. Whether this contributes to long-term renal

damage requires further study.

Page 7: Groningen International Program of Science in Medicine ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/... · (iii) Detection of macrophages 20 3. Enzyme-linked immunosorbent assay’s

7

Introduction

Sepsis is a life-threatening disorder and a leading cause of mortality among critically ill

patients. With an associated mortality rate exceeding 20%, sepsis is a major global health

problem (1-4). Sepsis represents an excessive systemic inflammatory host response to an

infectious agent that may result in multiple organ dysfunction (5), of which acute kidney

injury (AKI) is a common manifestation. The reported incidence of AKI in intensive care

units (ICU) may be as high as 70%, of which sepsis is associated with half the cases (1, 6, 7).

Overall mortality rates of patients with AKI remain high. Approximately one-third of patients

who require renal replacement therapy (RRT) die while still in the ICU (8-11). However, as

the biochemical abnormalities and fluid balance can be managed by renal replacement

therapy, it is uncertain whether or not AKI directly contributes to mortality. There is no

specific treatment for AKI once it occurs; current management is largely preventive or

supportive. Despite septic AKI being a commonplace problem, relatively little is known about

its pathogenesis. Clearly, outcomes should be improved with a better understanding of the

pathophysiology leading to more directed therapies.

Current tools and limitations

Although AKI is common, and even mild forms of AKI portend worse clinical outcomes, it is

often recognized late (7, 12). AKI defines a broad spectrum of renal impairment, ranging from

minimal elevations in serum creatinine to anuric renal failure. Because of great variations in

this clinical syndrome, a uniform definition was lacking until relatively recently. The Acute

Dialysis Quality Initiative (ADQI) proposed the RIFLE criteria (Risk, Injury, Failure, Loss

and End-stage renal disease) (13). This classification system for diagnosing and classifying

AKI is defined by criteria for serum creatinine (sCr) and urine output (Figure 1).

Subsequently, these criteria were refined by the Acute Kidney Injury Network (AKIN)

(Figure 1) (14). Both have been recently amalgamated into the ‘Kidney Disease: Improving

Global Outcomes (KDIGO)’ guidelines for Acute Kidney Injury (15), that offer a means of

standardizing epidemiological data and clinical studies.

Both AKIN and RIFLE criteria have been validated by a number of epidemiological studies.

Despite their merits, the criteria must be interpreted within their limitations. They lack detail

on the mechanisms and the precise timing of injury. Furthermore, knowledge of the patient’s

baseline serum creatinine is required, and monitoring of urine output is highly variable

depending on their hydration status and the use of diuretics.

The use of serum creatinine as a biomarker has several limitations. There may be a significant

loss of renal functional reserve (up to 70%) before serum creatinine rises. It may also not

accurately reflect the glomerular filtration rate (GFR) in acute illness due to fluctuations in

intravascular volume status (16). Alterations in creatinine production that accompany critical

illness, due in part to loss of muscle mass, further confound the use of serum creatinine as a

renal biomarker in acute illness. The need for more sensitive biomarkers of renal dysfunction

is therefore required, which may allow earlier initiation of appropriate management.

Page 8: Groningen International Program of Science in Medicine ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/... · (iii) Detection of macrophages 20 3. Enzyme-linked immunosorbent assay’s

8

Figure 1. RIFLE and AKIN classifications for acute kidney injury. Adapted from (13)

RIFLE: Risk–Injury–Failure–Loss–Endstage renal disease; AKIN: Acute Kidney Injury

Network; ARF: acute renal failure; Cr: creatinine; GFR: glomerular filtration rate.

Haemodynamic and histopathological findings

Knowledge of the pathophysiology of septic AKI is still in its infancy. It was generally

believed that, like other causes of shock, AKI was mainly due to renal hypoperfusion. Sepsis

results in hypovolaemia due to increased capillary leak, and hypotension due to arterial

vasodilatation and a loss in vascular tone. The resulting drop in renal blood flow (RBF) was

felt to cause renal ischaemia and, consequently, renal failure. However, Bellomo et al

systematically reviewed changes in RBF in sepsis and found that RBF was actually elevated

in human studies (17). Thereafter, they analyzed 159 animal studies and also found RBF was

either preserved or elevated during sepsis, provided the animals were fluid-resuscitated. They

confirmed these findings in a sheep model of septic shock which showed prominent renal

vasodilatation, accompanied by a marked increase in RBF, despite a three-fold increase in

serum creatinine and progressive oliguria (18, 19).

Another long-held paradigm in septic AKI was that acute tubular necrosis (ATN) would be

the predominant histological finding. Tubular injury evolves as a result of generalized or

localized impairment of oxygen and nutrient delivery to, and waste product removal from,

tubular epithelial cells (TECs) (20). TECs are metabolically very active and therefore

particularly sensitive to anoxia and vulnerable to toxins. This susceptibility partly derives

from the arrangement of the vascular supply to the outer medulla. In health, the pars recta of

the proximal tubule and the thick ascending limb are on the verge of hypoxia due to the

countercurrent exchange properties of the vasa recta, what increases by alterations of flow to

these areas (21). Other factors that predispose tubules to toxic injury include a vast

electrically-charged surface for tubular reabsorption, active transport systems for ions and

organic acids, and the capability for effective concentration. This potentially leads to

reabsorption of substances from the tubular lumen that may cause toxicity to epithelial cells

(22). Toxins may include drugs or high concentrations of endogenous substances such as

Page 9: Groningen International Program of Science in Medicine ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/... · (iii) Detection of macrophages 20 3. Enzyme-linked immunosorbent assay’s

9

myoglobin, potassium or urea (22). Tubular ischaemia initially leads to damage of TECs

followed by shedding. The resulting cellular debris is thought to exacerbate excretory failure

by blockage of the tubules. Despite this cascade being conceivable for septic AKI it does not

correspond with the histopathological findings normally found in septic AKI. While a local

inflammatory response with leukocyte infiltration and tubular apoptotic bodies has been

described in the kidney (23); acute tubular necrosis only occurs in 22% of renal

histopathological specimens, though usually to minimal degrees only, and fails to explain the

degree of functional impairment seen in vivo (24).

Other haemodynamic changes within the kidney such as efferent arteriolar vasodilatation, re-

distribution of blood flow due to periglomerular shunting, and late vasoconstriction may all

occur at different timepoints during the course of sepsis. However, the development of septic

AKI is likely not due to renal haemodynamic changes alone, particularly in view of the lack

of correlation between histopathological changes and functional abnormality. This has led to

other hypotheses including a role for immunological factors and other sequelae of

inflammation including endothelial dysfunction, coagulation abnormalities, endothelial

dysfunction, and oxidative stress (15).

Inflammation and the inflammasome

In response to inflammatory stimuli, a sequence of host-microbial interactions occur which

activates the innate immune response. Innate immunity coordinates the first-line defence

response against pathogens and involves both humoral and cellular components. Besides

being involved in the initiation of inflammation, it also has a warning function by alerting the

adaptive immune system. The components of innate immunity recognizes structures shared by

various classes of antigens, so-called pattern recognition receptors (PRRs) (25). PRRs

recognize pathogen-associated molecular patterns (PAMPs) on invading organisms, as well as

endogenous stress signals termed danger-associated molecular patterns (DAMPs). To date,

various DAMPs and PAMPs have been recognized. Examples of PAMPs include LPS,

peptidoglycan, bacterial DNA, viral RNA and fungi. Many DAMPs are nuclear or cytosolic

proteins that get released outside the cell or are exposed on the cell surface following stress,

tissue injury or cell death (26). Non-protein DAMPs include purines such as adenosine 5’-

triphosphate (ATP) (27), uric acid (28) and adenosine (29).

Toll-like receptors (TLRs), a class of PRRs, are transmembrane proteins primarily expressed

by innate immune cells, but also by endothelial and epithelial cells (30). Neutrophils and

macrophages recognize microbes via these surface receptors. During sepsis, there is a

significant upregulation in TLR-2 and TLR-4 expression (31). TLR-4 has been discovered as

a key mediator in renal inflammation (32). TLRs activate pro-inflammatory transcription

factors such as nuclear factor kappa B (NFkB) with subsequent production of cytokines (e.g.

interleukin-1 (IL-1), tumor necrosis factor-α (TNF-α) and IL-6, enzymes such as

cyclooxygenase-2 (COX-2), and a variety of chemokines, cellular adhesion molecules,

receptors and other proteins (33, 34) (Figure 2, i and ii).

IL-1β is a key initiator of the acute inflammatory response and plays a pivotal role in the

pathogenesis of septic shock (35, 36). However, initial IL-1β processing and release is

insufficient and a secondary stimulus is necessary (36). A considerable role for post-

transcriptional processing and release of mature cytokines is via formation of a large multi-

protein complex, the NLRP3 inflammasome. Initial activation of PRRs not only results in

NFκB-dependent transcription of pro-IL-1β but also upregulates NLRP3 (37, 38).The cell

surface P2X7 receptor (P2X7R) facilitates assembly of the NLRP3 inflammasome (39-41).

Page 10: Groningen International Program of Science in Medicine ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/... · (iii) Detection of macrophages 20 3. Enzyme-linked immunosorbent assay’s

10

ATP released into the extracellular milieu during inflammation is a potent stimulus for P2X7R

activation (38, 42, 43), leading to IL-1β processing from activated monocytes, macrophages

and microglia (44). Monocytes from transgenic mice lacking the P2X7R were unable to

release IL-1β in response to ATP (45). Pro-inflammatory molecules such as interferon-γ,

(IFN-γ), TNF-α and endotoxin can induce upregulation in P2X7R synthesis (46). ATP-driven

P2X7R activation results in formation of an ion pore causing potassium (K+) efflux (figure 2

iii). The following endogenous depletion of K+ is a key step in inflammasome activation (47,

48). The NLRP3 inflammasome recruits pro-caspase-1 which, by proteolysis to caspase-1, is

the final step in the processing and release of bioactive IL-1β (figure 2 iv).

Figure 2. Schematic figure of the inflammasome pathway.

i) Innate immunity is activated via

DAMP signalling with initial

activation of TLR-4.

ii) TLR-4 then activates pro-

inflammatory transcription factors

such as NFkB, leading to production

of pro-IL-1β and pro-IL-18 and

upregulation of NLRP3.

iii) ATP released into the extracellular

milieu during inflammation stimulates

P2X7R activation, a key step in

inflammasome activation.

iv) NLRP3 recruits pro-caspase-1

which is converted to caspase-1, the

final step in the processing and

release of bioactive IL-1β and IL-18.

Stimulation mediated through P2X7R/ NLRP3 rapidly releases large amounts of mature IL-1β

at rates up to 100x faster than with PAMP exposure alone (49, 50). Extended stimulation of

the P2X7 inflammasome cascade induces caspase-1-mediated death of monocytes and

macrophages (51-53). IL-1β may also be activated by P2X7-independent pathways. This

includes shedding via microvesicles (54), processing to biological active IL-1β by neutrophil-

derived serine proteases (cathepsin G, neutrophil elastase, and proteinase-3) (55, 56), or by

mast cell–derived serine proteases (granzyme A and chymase) (57, 58).

IL-18 is also an important inflammatory mediator. Excessive levels are associated with septic

shock and autoimmune syndromes (59). IL-18 shares structural features with IL-1, but is not

an endogenous pyrogen (60, 61). IL-18 binds to a receptor inducing synthesis of other pro-

inflammatory cytokines (IL-1β, IL-6, IL-8, TNF-α, IFN-γ) and chemokines (60). IL-18 is also

a substrate for the caspase-1 inflammasome, functioning via the P2X7R (37). IL-18 excretion

is higher in septic compared to non-septic AKI; increased levels predict deterioration of

kidney function 24-48 hours before AKI becomes clinically apparent (62, 63).

Page 11: Groningen International Program of Science in Medicine ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/... · (iii) Detection of macrophages 20 3. Enzyme-linked immunosorbent assay’s

11

NLRP3 pathway

Activation of the innate immunity occurs via DAMP and PAMP signalling, resulting in

release of the pro-inflammatory cytokines, IL-1β and IL-18. An additional stimulus is

necessary for extracellular release of active IL-1β and IL-18. Inflammasome-mediated post-

translational processing of IL-1β (and other inflammasome-inducing stimuli such as IL-18) is

linked to efficient export of these molecules to the extracellular compartments. Extracellular

ATP, acting on the P2X7R, is one of the best established stimuli for this pathway (59, 64).

How the inflammasome exactly mediates these series of events has yet to be fully resolved

since it can be triggered by different mechanisms in different cell types.

P2X7 receptor

The purine ATP is an important extracellular messenger in various physiological processes,

including synaptic transmission and inflammation (65, 66). Its effects are mediated through

activation of purinergic receptors, e.g. P1 adenosine and P2 nucleotide receptors (67). P2

receptors are classified into two subfamilies, the P2X ligand-gated ion channels and the P2Y

G protein-coupled receptors. P2 receptors are expressed differentially throughout the kidney.

Purinoceptor signaling has important roles in water and sodium transport, control of the renal

microcirculation and in autoregulatory responses (68-70). By its structural differences from

the other P2X members and the requirement of high concentrations of ATP for activation, the

P2X7 receptor (P2X7R) has several unique properties among the P2X family.

The P2X7R, first characterized in immune cells including monocytes, macrophages,

lymphocytes and bone-marrow derived cells, has a wide expression pattern, particularly in

epithelial cells of the gastrointestinal tract, skin, salivary glands and urinary tract (71, 72).

Demonstration of the extracellular release of ATP and the presence of P2X7R in the renal tract

are consistent with a functional role for nucleotide signaling. Attempts to define the functional

importance of this potential signaling system have not yet led to complete understanding.

The P2X7 membrane receptor functions as a ligand-gated ion channel; it activates cell

membrane permeabilization and various downstream-signaling pathways, including

inflammatory responses and modulation of cell turnover (72). This pro-inflammatory response

results in release of inflammatory cytokines such as IL-1β and IL-18 from macrophages,

changes in plasma membrane lipid distribution, and can lead to cell death by necrosis or

apoptosis (54, 70, 73). As discussed, the P2X7 subtype is the receptor responsible for ATP-

driven maturation and release of IL-β (44, 45, 74-76). Pro-inflammatory cytokines and

bacterial products (e.g. LPS) can also upregulate P2X7R expression and increase its sensitivity

to extracellular ATP (64). As the P2X7R is non-desensitizing, the pore stays open as long it is

bound by ATP. Sustained activation by extracellular ATP confers the ability of the molecule

to form a large plasma membrane ‘pore’, enabling the receptor to function not only as a non-

selective cation channel, but to also mediate cell membrane permeabilization by allowing

passage of large molecules up to 900Da (42). Should ATP stimulation be prolonged, cells

become irreversibly injured and death occurs (42, 70, 77).

The ability of the P2X7R to mediate inflammatory responses and its dual role in cell turnover

(by potentially effecting both apoptosis and proliferation) suggest its importance in situations

in which these processes are prominent. These include both normal and abnormal renal

function (78). In vitro studies where rat mesangial cells were stimulated by LPS, IFN-γ or

TNF-α showed significant upregulation in mRNA expression of P2X7. Exposure of these cells

to external ATP resulted in apoptotic cell death (79, 80). In vivo studies of healthy rat kidneys

however yielded very low levels of P2X7R expression (80, 81). A key role for P2X7R has

been suggested in glomerulonephritis in experiments using a rodent model of nephrotoxic

Page 12: Groningen International Program of Science in Medicine ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/... · (iii) Detection of macrophages 20 3. Enzyme-linked immunosorbent assay’s

12

nephritis (NTN) (70). A three-fold upregulation of glomerular P2X7R mRNA was seen by day

4, correlating with the onset of significant proteinuria and peak macrophage infiltration (71).

In the same model, P2X7-knockout mice were used, showing P2X7 deficiency was

significantly renoprotective as evidenced by better renal function, a striking reduction in

proteinuria and decreased histological glomerular injury (82). Other experiments with A-

438079, a selective P2X7 antagonist, prevented the development of antibody-mediated

glomerulonephritis in rats (82). These data support a pro-inflammatory role for P2X7 in

immune-mediated renal injury and suggest that the P2X7R is a potential therapeutic target.

Macrophages and monocyte chemoattractant protein 1 (MCP-1)

Macrophages, as major components of the mononuclear phagocyte system, play a significant

role in inflammation. A normal inflammatory response involves signals that initiate, maintain

and terminate this process. An imbalance leaves inflammation unchecked, resulting in cellular

and tissue damage. In inflammation, macrophages have three major functions, namely antigen

presentation, phagocytosis and immunomodulation via production of cytokines and growth

factors. Therefore, macrophages play a critical role in the initiation, maintenance, and

resolution of inflammation. Different cytokines can influence this inflammatory process by

activating and deactivating macrophages.

Monocyte chemoattractant protein 1 (MCP-1) belongs to a family of small cytokines called

chemokines. These play a major role in selectively recruiting monocytes, neutrophils and

lymphocytes, and in inducing chemotaxis. MCP-1 is a key chemokine that regulates migration

and infiltration of macrophages; it is not only expressed by macrophages but also by

endothelial, mesangial and epithelial cells (83). By eliciting and activating leukocytes, this

regulatory mediator plays an important role in the inflammatory cascade and, perhaps, the

pathogenesis of sepsis. P2X7R has a central role in macrophage IL-1β secretion via the

NLRP3 inflammasome (82). In an experimental model of glomerulonephritis a link was found

between P2X7, MCP-1 and macrophage infiltration (82). Moreover, MCP-1 was shown to be

important in glomerular recruitment of macrophages and crescent formation in NTN (84, 85);

it has been studied as a biomarker for the mononuclear inflammatory processes that occur

following ischaemia-induced AKI (86). Aim of study

My fellowship is aimed at researching the expression of renal P2X7R and any relationship

with the course and pathogenesis of septic AKI. I aimed to define (an early) role for the

P2X7R in a fluid-resuscitated three-day rat model of faecal peritonitis and, specifically, to

define its relationship and co-localization to histopathological changes seen in septic AKI. I

hypothesized that upregulation of the ATP-sensitive P2X7R causes renal injury in sepsis

through local production of cytokines and chemokines and mediation of cell death.

Despite minimal histological damage (87), survivors of acute kidney injury who require acute

renal replacement therapy have an up to 28-fold increased risk of developing stage 4 or 5

chronic kidney disease (CKD) (88, 89). Most of the focus of AKI has been on the acute injury

phase but little attention to date has been paid to recovery. Mechanisms underlying resolution

of AKI in sepsis are poorly understood and may be potentially injurious. Therefore, I also

aimed to describe temporal changes in renal inflammation and recovery following sepsis and

to establish the involvement of renal macrophages and cytokines in a long-term rat model of

zymosan-induced peritonitis.

Page 13: Groningen International Program of Science in Medicine ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/... · (iii) Detection of macrophages 20 3. Enzyme-linked immunosorbent assay’s

13

Materials & Methods

For buffers and solutions, see Appendices I

1. In vivo methods

Rat model of acute –polymicrobial- sepsis

Male Wistar rats weighing 300-350g were used. All invasive procedures and imaging

techniques were performed under isoflurane general anaesthesia. Twenty-four hours

following insertion of a tunnelled central venous catheter, sepsis was induced by

intraperitoneal injection of faecal slurry. Sham-operated animals had lines inserted but no

intraperitoneal injection. All animals were inspected four times daily and given a clinical

score based on their physical appearance (90). Fluid resuscitation with crystalloid was

initiated at 10 ml/kg/hr 2 hours following induction of sepsis. Sham animals received a similar

fluid regimen. At either 6 or 24 hours, animals were anaesthetized and an echocardiography

and renal ultrasound scan performed, followed by arterial blood gas analysis.

Subsequently, a laparotomy was performed. The urinary bladder was directly punctured with

a 22 gauge needle to drain and measure urine output. The left kidney was then resected. The

upper pole of the kidney was placed in 20% formalin and the remaining kidney was snap-

frozen into liquid nitrogen (LN2). A thoracotomy was performed followed by cardiac puncture

to obtain blood. This was centrifuged at 6500 rpm for 10 minutes, with subsequent snap

freezing in LN2. Sections of liver, spleen, lung, heart and aorta were collected and stored in

formalin and the remaining segments of these organs were snap-frozen in LN2. There were six

animals for each of the 6 hour timepoint groups and 8 animals for each of the 24 hour

timepoint groups. Each timepoint had a sham-operated and sepsis group. Separately, samples

were taken from non-instrumented naïve animals under anaesthesia.

Rat model of sepsis and recovery

Male Wistar rats weighing 300-350g were used. Rats were randomly divided into two groups:

sham or sepsis with timepoints at days 2 and 14. Animals in the septic group received

intraperitoneal zymosan* while sham animals received intraperitoneal saline. To assess their

clinical course, all animals were inspected daily and given a clinical score based on their

physical appearance. Body weight and food intake were also measured daily. Animals were

culled at days 2 or 14 post-zymosan injection and the left kidney harvested within 2 minutes

of the animal being culled. Sections of liver, spleen, lung, heart and aorta were collected and

stored in formalin and the remaining tissues were snap-frozen in LN2.

* Zymosan is a yeast-derived compound used to induce sterile inflammation. An advantage of

the zymosan-induced peritonitis (ZIP) model is that it generates a more prolonged

inflammatory insult than that seen in bacterial models. By adjusting the dose of zymosan,

insult severity can be varied. Clinical recovery in zymosan-treated animals commences from

48-72 hours, mimicking that seen in many immunocompetent individuals.

2. Homogenization of Frozen Kidney Tissues

Snap-frozen kidney sections (approx 2x3x2 mm) were put into a sterilin container containing

2 ml ice-cold radioimmunoprecipitation assay (RIPA) buffer. Tissue was homogenized on ice

using an electric homogenizer. Blades were rinsed in between with dH2O, 70% IMS and

Page 14: Groningen International Program of Science in Medicine ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/... · (iii) Detection of macrophages 20 3. Enzyme-linked immunosorbent assay’s

14

dH2O, respectively. Following thorough lysis, the homogenate was centrifuged for 5 minutes

at 16000g at 4°C to remove tissue debris. The supernatant was aliquotted and stored at -80°C.

3. Bicinchoninic acid (BCA) protein estimation assay

Following homogenization of the frozen kidney tissue, protein concentration was estimated

using a BCA Protein Assay kit (Novagen). A standard curve was prepared by diluting bovine

serum albumin (2 mg/ml) to achieve final concentrations of 2, 1.5, 1, 0.5, 0.25, 0.125 and

0.025 mg/ml using dH2O as a diluent. Tissue homogenate was diluted 1:10 in dH2O. To

prepare the BCA assay working reagent, 50 parts of reagent A were combined with 1 part of

reagent B. A blank 96-well plate was used and 25 μl of the standards or diluted samples were

added in duplicates. Two hundred μL of BCA working reagent were added to each well, and

placed on a shaker for 30 seconds. The plate was sealed and incubated at 37°C for 30 minutes.

Afterwards, the plate was left to cool to room temperature and the absorbance at 562 nm was

determined using a spectrophotometer (Synergy 2, Biotek). The recorded absorbance was

corrected for the dilution and the protein concentrations of the samples were extrapolated

from the standard curve.

4. Western blotting

Homogenization was performed as described previously, and protein concentrations estimated

using the BCA Protein Assay kit (Novagen). Kidney tissue homogenate was corrected for an

equal amount of 4 μg/μL protein per sample. SDS-sample buffer was added in a 1:1 ratio. To

reduce and denature the proteins, the cell lysate in sample buffer was heated at 90°C for 5

minutes, then quickly centrifuged at 16000g in a microcentrifuge for 1 minute and kept on ice

before use.

Equal amounts of protein were electrophoresed on 12% SDS-PAGE Tris-glycine minigels in

an electrode tank containing running buffer at a current of 80-120 Volt (for 2 gels: 1-1.5

hours). Subsequently, the gels were transferred onto pre-treated polyvinylidene difluoride

PVDF membrane (GE Healthcare). Semi-dry electroblotting was performed on a Bio-Rad

Trans-blot SD semi-dry transfer cell where the electrode plates are in direct contact with filter

paper as buffer reservoirs. Filter paper and membranes were trimmed to match the dimensions

of the gel and equilibrated in transfer buffer. The blotting assembly from anode to cathode

contained a filter paper sheet, the gel, PVDF membrane and a second filter paper. The

electroblotting was performed at a constant current of 15V for 45 minutes. Non-specific

antibody binding was minimized by blocking the membrane in TBST buffer (TBS with 0.1%

Tween 20) supplemented with 5% non-fat dry milk for 1 hour at room temperature. The

membranes were then incubated with the primary antibodies overnight at 4°C with gentle

agitation. The following antibodies were used; IL-18 (rabbit polyclonal, Santa Cruz) diluted

1:1000 in 5% milk/TBST and Caspase-1 (rabbit polyclonal, Santa Cruz), diluted 1:1000 in 5%

BSA in TBST. Membranes were then incubated with appropriate secondary antibodies

(Polyclonal Rabbit Anti-Goat IgG/HRP, DAKO or anti-rabbit IgG peroxidise labelled

polymer, DAKO), respectively, both diluted 1:3000 in 5% milk/TBST. Afterwards, the

membranes were washed three times for 8-10 minutes. Bands were visualized by enhanced

chemiluminescence (ECL) detection kit (GE-Amersham Biosciences). X-ray films were

manually developed at several incubation timepoints.

Page 15: Groningen International Program of Science in Medicine ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/... · (iii) Detection of macrophages 20 3. Enzyme-linked immunosorbent assay’s

15

5. Immunohistochemistry

To assess the amount and localization of P2X7, slides were stained with rabbit anti-rat P2X7

(Alamone). Signal development was performed using an anti-rabbit Envision kit (Dako

EnVision+ HRP-systems, DAB). Rat anti-mouse macrophage CD68 (AbD Serotec Inc.) was

used to assess the amount and localization of macrophages, visualization of these antibody

was obtained using an anti-mouse kit (Dako EnVision+ HRP-systems, DAB). Periodic acid-

Schiff (PAS) and Hematoxylin and eosin (H&E) staining are not described as they were

processed through the lab at Imperial College London, South Kensington.

Rat renal and spleen tissue samples were harvested and kept in 20% formalin prior to fixation.

After 24 hours, tissue was rinsed in 70% ethanol and embedded in paraffin wax. Tissue

sections, 5μm thick, were cut with a microtome and mounted onto coated glass slides.

Tissue sections were deparaffinized in xylene and rehydrated in decreasing concentrations of

ethanol to water. The slides were then heated in 0.01M citrate buffer [pH 6.0] at 90°C to

retrieve antigen exposure and allowed to cool for 5 minutes. Slides were washed three times

for 5 minutes each with excess PBS. In case of P2X7 staining, PBST (0.05% Tween 20 in

PBS) instead of PBS was used for all washes to reduce the amount of background staining.

To quench any endogenous peroxidase activity, sections were blocked with 3% H2O2 for 10-

30 minutes, followed by 3x5 min washes with excess PBS(T). Non-specific binding was

blocked by incubation with 10% milk in PBS (for P2X7) or 20% normal goat serum (NGS)

diluted in PBS (for CD68). Subsequently, the sections were incubated with the following

primary antibodies; P2X7 (rabbit anti-rat, 1:100, 004 Alamone) diluted in 1% BSA/0.05M

Tris-HCL [pH 7.2] or CD68 (mouse anti-rat, 1:500, AbD Serotec) diluted in PBS. The slides

were incubated for 1 hour at room temperature. One section from each sample was incubated

with antibody diluent instead of primary antibody and served as control. After washing, the

slides were incubated with the appropriate peroxidase-labelled polymer for 30 minutes at

room temperature. For P2X7 this was biotinylated goat anti-rabbit immunoglobulins in Tris-

HCL buffer (Dako), and for CD68 biotinylated goat anti-mouse immunoglobulins in Tris-

HCL buffer (Dako). After washing in PBS(T) to remove excessive unbound antibody, nickel

intensified 3-3′-diaminbenzidine (DAB, Dako) was used reveal antibody binding. Sections

were counterstained with Harris‘s haematoxylin solution, then dehydrated and mounted with

Eukitt (VWR International) and left to dry overnight.

The slides were examined with a Zeiss Axioplan light microscope (Carl Zeiss International).

Images were taken with a Leica DC200 digital camera (Leica Microsystems). Quantification

of staining was assessed by using Image Pro Plus software (Media Cybernetics) and each

slide was given an arbitrary score based on this method. For each section, 10 random fields of

view (x20 magnification) within the outer cortex were taken and the mean intensity of

staining was calculated.

6. Enzyme-linked immunosorbent assays (ELISAs)

IL-1β and MCP-1 estimations were performed to assess the presence of rat IL-1β and MCP-1

in renal tissue homogenate and serum. For the IL-1β and MCP-1 Sandwich ELISAs, rat IL-1β

DuoSet ELISA kit (R&D Systems) and rat MCP-1 ELISA set (BD Biosciences) were used.

Page 16: Groningen International Program of Science in Medicine ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/... · (iii) Detection of macrophages 20 3. Enzyme-linked immunosorbent assay’s

16

A 96 blank plate was coated with 100 μL of capture antibody, sealed and left to incubate

overnight. The following capture antibodies were used; goat anti-rat IL-1β (1:180 in PBS,

incubation at RT) and goat anti-rat MCP-1 (1:250 diluted in 0.1M Sodium Carbonate [pH

9.5], incubation at 4°C). Each well was aspirated and washed with wash buffer for three times

each. The plate was blocked with the appropriate assay diluent. For IL-1β this was 1% BSA in

PBS [pH 7.2-7.4], 300 μL/well. For MCP-1 this was 10% FBS in PBS, 200 μL/well. Both

plates were incubated for 1 hour at room temperature. After washing of the wells, again for 3

times each, 100 μL of the standards or samples were added in duplicates and incubated for 2

hours at room temperature, followed by 5x aspirating and washing of the wells.

For IL-1β, the method of antibody detection occurred in two subsequent steps:

(1) 100 μL of detection antibody (biotinylated goat anti-rat IL-1β, 1:180 in assay diluent) was

added to each well; the plate was then sealed and incubated for 2 hours at room temperature

(2) After washing (5x), 100μL of working dilution of Streptavidin-horseradish peroxidase

conjugate (SAv-HRP, 1:200 in assay diluent) was added to each well with an incubation

period of 20 minutes at room temperature.

For MCP-1, working detector was made containing biotinylated anti-rat MCP-1 (1:500

diluted in assay diluent) supplemented with SAv-HRP (1:250). One hundred μL of this

solution was added to each well with an incubation period of 1 hour at room temperature.

Afterwards, both plates were washed 7 times with soaking of the wells in wash buffer for 1

minute per wash. One hundred μL of substrate solution (1:1 mixture of Colour Reagent A

(H2O2) and Colour Reagent B (Tetramethylbenzidine) was added and incubated for 20

minutes in the dark at room temperature. Fifty μL of stop Solution (2N H2SO4) was added to

each well and absorbance was read at 450 nm using a spectrophotometric ELISA plate reader.

Using BCA protein estimation, kidney tissue homogenate was corrected for an equal amount

of protein per sample. Samples that were out of range were diluted in a 1:2-1:100 manner with

re-estimation of the protein concentration.

7. Statistics

All statistical analyses were performed using SPSS (IBM. Version 20) and graphs were drawn

using Graphpad Prism (GraphPad Software, Version 5.0d). Normality of continuous data was

assessed using the Shapiro-Wilk test. Continuous variables are presented as means (standard

deviation). Parametric data were compared using unpaired t-test, whereas non-parametric data

were compared using the Mann Whitney U test. One-way ANOVA (analysis of variance) was

used to assess difference between more than two groups of continuous variables. Post-hoc

Tukey’s test was performed to ascertain differences between individual groups. Pearson’s

correlation was used to assess linear association between two groups of continuous variables.

A p value <0.05 was taken as statistically significant.

Page 17: Groningen International Program of Science in Medicine ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/... · (iii) Detection of macrophages 20 3. Enzyme-linked immunosorbent assay’s

17

Results

I. Polymicrobial model of acute sepsis

1. Analysis of renal dysfunction by use of serum creatinine

Serum creatinine levels were analyzed as a measure of kidney dysfunction. At 6 hours, sham

and septic animals demonstrated similar, non-significantly raised, serum creatinine levels

(26±3 μmol/L sham vs 24±3 μmol/L septic; p = 0.399). In sepsis, a significant rise in serum

creatinine was seen at 24 hours (24±3 sham vs 30±5 μmol/L septic; p = 0.012) (Figure 1).

Serum creatinine levels in naives were within the normal range (20-25 μmol/L).

Figure 1. Creatinine

values expressed in

μmol/L; significantly

increased serum

creatinine levels were

seen in septic rats at 24 hours compared

to controls (sham) (indicated by asterisk,

p <0.05). Serum creatinine in naive and

sham-operated rats, are within normal

ranges (20-25 μmol/L). Data represent

mean±SD. N= 6 for naive and 6 hour

groups (sham and septic), and n=8 for

both 24 hour groups. sCR: serum

creatinine.

2. Immunohistochemistry

2a. Assessment of histological damage

To assess histopathological damage in kidneys during sepsis, I examined the Periodic acid–

Schiff (PAS) stained kidney sections. Acute renal histological injury was evaluated by a pre-

defined scoring system (82). This quantifies for tubular cell necrosis, loss of luminal brush

border, tubular cast formation and tubular dilatation by ascribing the following scales: mild-

moderate-severe. For each group, i.e. naive, sham (6 and 24 hr) and sepsis (6 and 24 hr), a

minimum of 6 slides were scored. Perimortem animals were excluded from analysis as such

animals had significant global haemodynamic compromise that may affect the histological

appearances.

No significant difference in overall acute renal histological injury was seen between sham and

septic kidneys at either 6 or 24 hours (figure 2). Both sham and septic animals had contained

areas with minimal dilated tubules and minimal loss of brush border. There was no evident

presence of necrosis or cast formation throughout the kidney. Looking at the time of sepsis,

there were no major histological changes at either 6 or 24 hours after initiation of sepsis.

Despite clinical, biochemical, and renal haemodynamic changes of septic AKI, histology

remains relatively preserved.

Creatinine (umol/L)

Naive

6hr s

ham

6hr s

epsis

24hr

sha

m

24hr

sep

sis

0

10

20

30

40

6 hrs 24 hrsNaive

*

Naive

Sham

Sepsis

Page 18: Groningen International Program of Science in Medicine ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/... · (iii) Detection of macrophages 20 3. Enzyme-linked immunosorbent assay’s

18

Figure 2: Histological assessment of rat kidneys for renal damage. A. Naive renal tissue

without any significant damage. B. Renal tissue obtained from a haemorrhage-reperfusion

model (A. Dyson, UCL), showing several characteristics of ATN such as dilated tubules,

ischaemic glomeruli and tubular casts. C. Kidney from a sham animal without any significant

damage. D. Kidney section from a 24 hour septic rat showing a similar pattern (no significant

damage) compared to shams. (PAS-staining, magnification x 20)

2b. P2X7 expression in kidney tissue

I then assessed P2X7 expression to see whether P2X7 correlates with severe sepsis at either 6

or 24 hour timepoints. In septic animals, P2X7-dense areas were seen in a patchy, discreet

manner with relatively more proximal than distal tubular P2X7 staining. As shown in picture

3A there was minimal P2X7 expression in naive kidneys. Picture 3b shows the minimal P2X7

expression seen in sham animals, and mainly located within the capillary loops of the

glomeruli and at the basolateral regions of the tubules. P2X7 expression was evident at both

timepoints in sham and septic animals (Figures 3B and 3C, illustrating P2X7 expression in 24

hour sham and septic animals respectively). In severely septic animals (with a serum

creatinine elevated at least three-fold compared to sham animals), the pattern of P2X7 staining

differed with evidence of intraluminal debris that stained intensely for P2X7 (3D). This pattern

of tubular P2X7 is suggestive of a role in cell death. No P2X7 staining was observed in naive,

sham or septic renal tissue when the primary antibody was replaced with non-immune serum.

For each group (naive, sham and sepsis) and timepoints (6 and 24 hours), ≥6 animals were

scored. Scoring was based on the area and intensity of staining as described previously.

Briefly, quantification of staining was assessed by using Image Pro Plus software, and each

slide was given an arbitrary score based on this method. For each section, 10 random fields of

view (x20 magnification) within the outer cortex were taken and the mean intensity of

staining was calculated.

Page 19: Groningen International Program of Science in Medicine ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/... · (iii) Detection of macrophages 20 3. Enzyme-linked immunosorbent assay’s

19

Figure 3; Histological assessment of rat kidney for P2X7. A: Naive kidney with minimal

P2X7; B: Kidney from a 24hour sham animal with minimal P2X7 mainly within the capillary

loops of the glomeruli and at the basolateral regions of tubules; C: Kidney from a 24 hour

septic animal showing a similar pattern as the 24 hour shams; D: Kidney from a severe septic

animal at 24 hours with overwhelming intraluminal, intraluminal debris, with intense staining

for P2X7. (P2X7-staining, magnification x 20)

P2X7 was visible in both tubules and glomeruli of naïve kidney tissue (Figure 3). At 6 hours,

there was a wide variation in the level of P2X7 expression. There was no significant difference

in the expression of renal P2X7 at 6 hours between sham and septic rats (0.011 vs 0.013, p =

0.31). At 24 hours, there was a significant difference in P2X7 in sham compared to septic rats

(0.0067 vs. 0.0138, p= 0.0025) (Figure 4).

Figure 4. Semi-quantitative analysis

of P2X7; a significant increase in

P2X7 was seen in septic rats at 24

hours compared to controls

(indicated by asterisk, p <0.05). Data

represent mean±SD. N= 6 for naive

and 6 hour groups (sham and septic),

and n=8 for both 24 hour groups.

P2X7 score

Naive

6hr S

ham

6hr S

epsis

24hr

Sha

m

24hr

Sep

sis

0.000

0.005

0.010

0.015

0.020

0.025

His

tolo

gy s

co

re

*

Naive 6 Hrs 24 Hrs

Naive

Sham

Sepsis

Page 20: Groningen International Program of Science in Medicine ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/... · (iii) Detection of macrophages 20 3. Enzyme-linked immunosorbent assay’s

20

2c. Detection of macrophages

P2X7 is predominantly expressed by macrophages. Macrophages are a major component of

the mononuclear phagocyte system and play a critical role in the initiation, maintenance, and

resolution of inflammation. Therefore, I assessed the level of macrophage expression

throughout the kidney to seek any correlation between macrophage expression and sepsis-

induced AKI. Furthermore, I wanted to determine the relative contribution of infiltrating

macrophages to renal P2X7 staining. The presence of macrophages was sought using CD68

staining as described. At least six animals were included in each group.

Naive kidneys showed no or minimal staining of macrophages. When present, macrophages

were found within the interstitium. A similar pattern was seen in sham and septic animals at 6

and 24 hours (Figure 5). The minimal presence of macrophages in the kidney tissue and the

pattern of staining of P2X7 suggest P2X7 is likely to be expressed by intrinsic renal cells.

Figure 5; Histological assessment of rat kidneys for presence of macrophages. A: Naive

kidney with no visible macrophage staining; B: Kidney from a 24 septic animal with minimal

staining for macrophages, as indicated by the arrow (CD68-staining, magnification x 20).

3. Enzyme-linked immunosorbent assays (ELISA)

3a. Expression of serum and renal IL-1β

Interleukin-1β (IL-1β) is a key initiator in acute inflammation and plays a pivotal role in the

pathogenesis of sepsis (35, 36). A pro-inflammatory response progressing to a “cytokine

storm” causes inflammatory cytokines such as IL-1β to be released from macrophages (54,

70, 73). IL-1β then binds to receptor target cells, eliciting a signalling cascade that enhances

the inflammatory response which may, in turn, lead to cell death (91). The P2X7 subtype is

identified as the receptor responsible for ATP-driven maturation and release of IL-β.

Therefore I sought to evaluate the role of IL-1β in this model of septic AKI. Serum and renal

tissue IL-1β levels were quantified by ELISA. Experiments were repeated in duplicate using

six animals for naïve studies and 6 hour timepoints, and 8 animals for the 24 hour timepoints.

Serum IL-1β is expressed as pg/mL and renal IL-1β as a ratio of μM/μg of protein.

There was no detectable serum IL-1β in naive animals, with minimal to none in sham animals

at both 6 and 24 hour timepoints (Figure 6A). There was a significant rise in serum IL-1β in

septic compared to sham animals at both 6 and 24 hour timepoints (Table 1). There was a

non-significant fall in serum IL-1β levels from 6 to 24 hrs in septic animals (p = 0.206). For

Page 21: Groningen International Program of Science in Medicine ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/... · (iii) Detection of macrophages 20 3. Enzyme-linked immunosorbent assay’s

21

renal IL-1β levels, a significant rise was seen in the septic animals at both 6 and 24 hours

(Figure 6b). In contrast to serum levels, renal IL-1β revealed a consistent rise in sepsis from 6

to 24 hours (p=0.0009).

Figure 6; Serum and renal expression of the pro-inflammatory cytokine IL-1β. Left-hand

graph shows a significant increase in serum IL-1β in the septic group at both 6 and 24 hours

(p<0.05), with a non-significant fall at 24 hours. Right-hand panel: renal IL-1β protein ratio

increases in septic animals at 6 hours and further at 24 hours (p<0.05). Significance is

indicated by the asterisk. Data represent mean ± SD. Six rats were used for naive and 6 hour

groups (sham and septic), and 8 for both 24 hour groups.

3b. Expression of serum and renal MCP-1

In view of the lack of renal macrophage infiltration, I measured renal MCP-1 levels. Renal

MCP-1 levels were not elevated at 6 hours (97±23 (sham) vs 125±37 (septic) vs 88±13 pg/μL

(naive); p = 0.3359). (Figure 7). At 24 hours, the difference in renal MCP-1 levels in septic

compared to sham animals neared significance (178±78 vs 92±41 pg/μL; p = 0.055).

Figure 7. Renal MCP-1 levels.

These were not elevated in sham or septic

animals at 6 hours. At 24 hours, a near

significant increase was seen in septic

animals compared to controls (p=0.0553).

Data represent mean ± SD. Six rats were used

naive and 6 hour groups (sham and septic),

and eight for both 24 hour groups.

Renal IL-1beta (uMol/ug Protein)

Naive

6hr s

ham

6hr s

epsis

24hr

sha

m

24hr

sep

sis

0

100

200

300

400

500

6hrs 24hrsNaive

*

*

Serum IL-1beta (pg/mL)

Naive

6hr s

ham

6hr s

epsis

24hr

sha

m

24hr

sep

sis

0

500

1000

6hrs 24hrsNaive

*

*

Renal MCP-1 (pg/uL)

Naive

6hr s

ham

6hr s

epsis

24hr

sha

m

24hr

sep

sis

0

50

100

150

200

250

Naive

Sham

Sepsis

Naive

Sham

Sepsis

Page 22: Groningen International Program of Science in Medicine ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/... · (iii) Detection of macrophages 20 3. Enzyme-linked immunosorbent assay’s

22

Table 1. Overview of cytokines and creatinine measured in this model of acute sepsis.

Significant differences are indicated by an asterisk.

4. Western Blotting

Formation of the NLRP3 inflammasome converts pro- to active caspase-1. Inactive caspase-1

(Pro-C1) has a weight of 45 kDa in rats, whereas the active fragment of caspase-1 (p10)

weights 10 kDa. Caspase-1, or interleukin-1β converting enzyme (ICE), then promotes

maturation of interleukin IL-1β and IL-18 by proteolytic cleavage of precursor forms into

biologically active pro-inflammatory cytokines. I determined the relative abundance of

precursors and active forms of caspase-1 and IL-18 in kidney tissue.

Similar quantity of total protein was loaded into each well (20 μg). Ten μL of pre-stained

protein ladder, Broad Range (10-230 kDa) (New England Biolabs) was added once and

served as a reference indicator. This ladder consists of 12 bands, namely; 10, 15, 20, 25, 30,

40, 50, 60, 80, 100, 150, 230.

Immunoblotting of renal tissue homogenate revealed a major band at 45 kDa, the predicted

size of pro-caspase-1 in all groups (Fig 8). There was no visible difference in the presence of

the precursor caspase-1 between naïve, 24 hours sham and septic animals. For active caspase-

1, with a predicted weight of 10kDa, no band was detected in naive renal tissue. Compared to

naives, there was a slight increase in active caspase-1 in septic compared to sham animals.

Figure 8. Western blot analysis of kidney homogenate obtained from naive, control and septic

rats. Migration of the 45 kDa active caspase-1 and the 10 kDa pro-caspase-1 molecular forms

are indicated (N=3 for naives and N=4 for sham and septic rats).

Naive

6 hours 24 hours

Sham Septic P Sham Septic P

Renal

IL-1β

0 (0-0) 22 ± 55 834 ± 451 0.005* 0 (0-0) 544 ± 534 <0.001*

Serum

IL-1β

20 ± 20 36 ± 21 117 ± 22 0.004* 92 ± 62 360 ± 101 0.002*

Renal

MCP-1

88 ± 13 97 ± 23 125 ± 37 0.339 92 ± 41 178 ± 78 0.055

Creatinine 23 ± 3 26 ± 3 24 ± 3 0.399 24 ± 3 30 ± 5 0.012*

Page 23: Groningen International Program of Science in Medicine ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/... · (iii) Detection of macrophages 20 3. Enzyme-linked immunosorbent assay’s

23

Immunoblotting of kidney homogenate for IL-18 was performed in naïve rats and 24 hour

sham and septic rats. Minimal levels of pro-IL-18 were detected in renal tissue, with a relative

increase of pro-IL-18 in septic compared to sham animals (Fig 9). Active IL-18 protein was

detected at very low levels in all groups with no visible difference between groups.

Figure 9. Western blot analysis of

kidney homogenate obtained from

naive, control and septic rats.

Migration of the 24 kDa pro-IL-18 and

the 18 kDa active IL-18 molecular

forms are indicated (N=3 for naïve rats

and N=4 for sham and septic rats).

II. Recovery model of sepsis: Results

1. Immunohistochemistry

Periodic acid–Schiff (PAS)-stained kidney sections were examined for histopathological

damage at days 2 and 14 in sham and septic animals. Acute renal injury was evaluated by a

pre-defined scoring system as described earlier. For each group, at least 4 slides were scored.

Minimal, if any, evidence of renal injury was seen in sham and septic kidneys at Days 2 or 14.

To evaluate the role of macrophages in this recovery model, I examined kidney sections for

CD68 in 4 sham and 4 septic at Day 2, and 4 sham and 8 septic kidneys at Day 14.

Macrophage infiltration was evident in glomeruli and interstitium in all Day 14 septic animals

but in no sham animal (Fig 10). At Day 2, 1 septic animal had renal macrophage infiltration

and 1 sham animal had minimal glomerular macrophage infiltration (Fig 11).

Figure 10; Histological assessment of rat kidney for presence of macrophages. On the left:

No presence of macrophages in sham animals. On the right: Evident staining for macrophages

in the glomeruli and interstitium (CD68-staining, magnification x 20).

Page 24: Groningen International Program of Science in Medicine ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/... · (iii) Detection of macrophages 20 3. Enzyme-linked immunosorbent assay’s

24

Figure 11; Kidneys showing macrophage

infiltration.

A significant difference was seen in septic

animals at day 14 (p=<0.05). N= 4 in all

groups except for sepsis day 14, N=8.

2. Enzyme-linked immunosorbent assays (ELISA)

At day 2 similar renal MCP-1 levels were seen in sham and septic animals (72±15 vs 78±14

pg/μL, p=0.875). By day 14, renal MCP-1 levels were significantly elevated in septic

compared to sham animals (187±55 vs. 86±18 pg/μL, p=0.009). A significant elevation in

renal MCP-1 was seen at day 14 in septic animals compared to sham animals. This coincides

with increased macrophage infiltration and clinical recovery seen at day 14.

Figure 12; Renal MCP-1 levels in long-

term model.

A significant increase was seen in septic

animals at day 14 as indicated by asterisk

(p<0.05). Data represent mean ± SD.

N= 4 in all groups except for sepsis day 14;

N=8.

Renal MCP-1 (pg/uL)

MC

P-1

(p

g/u

L)

Sham

Day

2

Sepsis

Day

2

Sham

day

14

Sepsis

day

14

0

50

100

150

200

250*

Day 2 Day 14

Clinical Score

6hrs

24hr

s

0

1

2

3

4Sham

Sepsis* *

Page 25: Groningen International Program of Science in Medicine ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/... · (iii) Detection of macrophages 20 3. Enzyme-linked immunosorbent assay’s

25

Discussion

Despite clinical, haemodynamic (data not shown) and biochemical alterations, no abundant

characteristic pathological features were found on renal histology. This finding is similar to

that predominantly reported in the literature (92). There is still no defined consensus on a

scoring system for histopathological damages seen in AKI. This is partly due to a wide

variation in histological features (varying from no damage, to minimal loss of brush border

and a few dilated tubules, to severe acute tubular necrosis) but also as a result of a poor

correlation between histopathological findings and in vivo functional impairment. To

determine AKI severity, I used a scoring system that grossly divides animals into mild-

moderate and severe groups. I received confirmation of a lack in difference from a blinded

histopathologist (Dr. Paul Bass, UCL). Despite minimal, if any histological damage, my

results revealed overall levels of increased P2X7 expression in this model of AKI.

I found increased P2X7 expression on both glomeruli and tubules, and showed a pattern in

P2X7 expression suggestive of a role in cell death. The physiological function of the P2X7R is

still under investigation but a number of roles have been proposed. Expression in shedding

epithelia such as skin (93), duodenum (94), vagina and uterus (95) suggest it may have a role

in normal cell turnover. There are several reports of P2X7R activation causing either necrosis

or apoptosis of cells of haemopoietic origin such as macrophages and lymphocytes (43, 77,

96). Furthermore, the ability of extracellular ATP to trigger apoptosis via the P2X7R has been

reported in other cell types including thymocytes (97), dendritic cells (98) and mesangial cells

(80). In HEK-293 cells, dramatic membrane blebbing and micro-vesciculation was observed

within seconds to minutes of receptor activation, a phenomenon in which large membrane-

bound vesicles protrude rapidly from the cell surface, associated with cells undergoing

apoptosis (96). Normally, apoptosis is a tightly regulated mechanism for maintaining normal

and healthy cell numbers; in kidney it occurs at a low level (99). However, apoptosis

increases following several forms of glomerular injury including ischaemia (100).

Besides P2X7, I also assessed the role of other components of the NLRP3 inflammasome

pathway including caspase-1, IL-1β and IL-18. Western blotting revealed a slight increase in

active caspase-1 and pro IL-18, but this needs re-evaluation with inclusion of a positive

control (spleen), and quantified in relation to a housekeeping protein’ (β-actin). Furthermore,

differences between bands should be formally assessed using relative densitometry.

As expected, serum IL-1β levels showed a clear rise during the acute phase of sepsis (35, 36).

Moreover, renal IL-1β showed a further rise at 24 hours compared to 6 hours in this model of

septic AKI. Glomerular mesangial cells and podocytes are capable of producing IL-1β (101,

102); this cytokine is considered largely responsible for leukocyte infiltration in anti-GBM

glomerulonephritis (103). IL-1β binds to receptor target cells and elicits a signalling cascade

that enhances the inflammatory response leading to cell death (91). Target cells include

endothelial cells which, when exposed to IL-1β, are induced to secrete chemokines such as

monocyte chemotactic peptide-1 (MCP-1) (104).

MCP-1 is not only expressed by macrophages but also by endothelial cells, mesangial cells

and epithelial cells (83). Macrophages play a critical role in the initiation, maintenance, and

resolution of inflammation. Therefore, they mediate both the pro and anti-inflammatory

cascades. MCP-1 is a key chemokine that regulates migration and infiltration of macrophages.

As it is produced by, and acts on macrophages, it has a dual role in eliciting and activating

Page 26: Groningen International Program of Science in Medicine ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/... · (iii) Detection of macrophages 20 3. Enzyme-linked immunosorbent assay’s

26

leukocytes. Chemokines play a prominent role in the acute inflammatory response in several

models of kidney disease, and a specific role has been revealed for MCP-1.

MCP-1 provides a stimulus for chemotaxis in glomerulonephritis by facilitating glomerular

recruitment of macrophages and crescent formation in both rats and mice (85, 105, 106). In a

rat model of ischaemia-induced AKI, increased levels of kidney MCP-1 mRNA, protein and

an increase in urinary MCP-1 excretion were reported (86). In addition, a correlation was seen

between areas of increased MCP-1 expression and infiltration of mononuclear cells into the

kidney. In cultured astrocytes P2X7R activation increased MCP-1 expression via a MAP

kinase-dependent mechanism which includes P38-MAP kinase. This pathway plays a role in

the cascade of programmed cell death (107) and may provide a link with the apoptotic

function of the P2X7. In an experimental glomerulonephritis model a link was seen between

P2X7, MCP-1 and macrophage infiltration. P2X7 knockout mice showed a 96% reduction in

urine MCP-1 levels correlating with a reduction of macrophage infiltration into the glomeruli,

but not the interstitium. Treatment with A-438079, a selective P2X7 antagonist, caused a

significant reduction in macrophage infiltration in renal tissue, and of urinary MCP-1 levels.

Although renal MCP-1 will not differentiate between different causes of AKI, it may be a

biomarker for differentiation between active inflammation and resolution. The correlation

between renal MCP-1 levels and the amount of macrophage infiltration in renal injury, as

described in the literature, corresponds with findings in our long-term model of sepsis and

recovery. However, in other models of renal injury, both MCP-1 and macrophages were

detected at early stages. At the very start of clinical recovery (day 2 in our long term model), a

minimal presence of macrophages was seen with correspondingly low basal levels of renal

MCP-1. Similarly, in our model of sepsis, no infiltration of macrophages was seen at either 6

or 24 hour timepoints. This may suggest that in sepsis, renal macrophages and MCP-1 are

likely to play more of a role in (late) resolution of inflammation rather than in the acute phase.

Activated macrophages can be classified into M1 and M2 phenotypes (108, 109). The M1

macrophages, or classically-activated macrophages, are immune effector cells with an acute

inflammatory phenotype. They are highly aggressive against microbes and can produce large

amounts of cytokines. The M2 macrophages, or alternatively-activated macrophages, possess

anti-inflammatory properties and can be divided into at least three subgroups. These subtypes

have various functions including regulation of immunity, maintenance of tolerance and tissue

repair. I propose that the upregulation of macrophages seen at day 14 in our model of

recovery of sepsis belongs to the M2 subtype. The exact pathophysiological role for

macrophages and MCP-1 at different timepoints in this rat model of sepsis remains to be

determined. Whether there is initial MCP-1 expression in the kidney that attracts

macrophages, or whether macrophages mainly induce expression of renal MCP-1 needs to be

elucidated. Most likely they are synergic partners that both play an important role in the

pathogenesis and recovery of sepsis-induced renal injury.

Page 27: Groningen International Program of Science in Medicine ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/... · (iii) Detection of macrophages 20 3. Enzyme-linked immunosorbent assay’s

27

Conclusions

A significant increase in renal P2X7 expression (in both glomeruli and tubuli) was seen in

septic animals after 24 hours, correlating with elevated levels of IL-1β. Consistent with these

findings, septic AKI was associated with increased renal expression of caspase-1 protein and

pro-IL-18. In peri-mortem animals, there was evidence of intraluminal cellular debris, which

stained intensely for P2X7. This pattern of tubular P2X7 suggests a possible role in cell death.

Despite upregulation of P2X7, there was absence of macrophage infiltration. This, in

combination with the pattern of staining of P2X7, suggests renal P2X7 is likely to be

expressed by intrinsic kidney cells. While significant histological damage was not seen,

reflecting findings in septic patients, there was biochemical evidence of renal dysfunction.

Further research is needed to evaluate whether P2X7 antagonism offers protection.

By contrast, the longer-term sepsis model that also incorporated a recovery phase did shown

significant renal macrophage infiltration within the glomeruli and interstitium with a

corresponding increase in renal MCP-1 at day 14. Whether this contributes to long-term renal

damage requires further study.

Future work

P2X7 antagonism

An important reason for looking at the P2X7 in septic AKI is its possible role in inflammation,

particularly as the P2X7R is considered a possible therapeutic target in inflammation.

Antagonists are currently in Phase II clinical trials. Therefore, our next step would be to

evaluate the effects of P2X7 antagonism on renal function and survival.

Define renal histological features in sepsis in detail and attempt to score injury

Because there were clear differences not only in the amount but also in the pattern and areas

of P2X7 expression, I have attempted to define a scoring system to differentiate between the

functional areas within the kidney and the pattern of expression. This scoring is based on 1:

the location of staining, which was subdivided into (i) glomerular, tubular, or interstitial, (ii)

cortical or medullary (iii) cytoplasmic, basolateral or apical expression (in tubules) 2: the

pattern of staining (discrete or diffuse) and 3: the amount of staining. We are awaiting

validation of this scoring system by a certified histopathologist. If possible, this could also be

used to score immunohistochemistry on renal tissue for NLRP3, macrophages and α-SMA.

The inflammasome and the role of other immunological mediators

Although different components of the inflammasome pathway have been assessed, these

results should be correlated against upregulation and/or presence of the NLRP3

inflammasome itself. We are currently performing immunohistochemistry for NLRP3. As

discussed, P2X7 is associated with cell apoptosis, so we would like to further investigate this

association with immunohistochemistry using Terminal deoxynucleotidyl transferase dUTP

nick end labelling (TUNEL)-staining. Furthermore, we propose to further investigate the role

of other immune cells in septic AKI.

Page 28: Groningen International Program of Science in Medicine ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/... · (iii) Detection of macrophages 20 3. Enzyme-linked immunosorbent assay’s

28

Temporal changes in renal biomarkers

As previously discussed, creatinine is a poor biomarker in early septic AKI. Although several

other biomarkers have been proposed (including neutrophil gelatinase-associated lipocalin

[NGAL], kidney injury molecule-1 [KIM-1], Cystatin C and IL-18), none have convincingly

proven reliability (110). There is a need for (new) biomarkers of renal cell injury that may

identify patients with AKI at an earlier stage, and may contribute to the prognosis of septic

AKI. A potential biomarker could be MCP-1. The kinetics and location of MCP-1 and the

early detectability of MCP-1 protein suggests MCP-1 could function as a potential early

biomarker for the mononuclear inflammatory processes that occurs in acute renal injury. In

mouse models with pre-, intra- and post-renal injury, its role as a biomarker was assessed

(111). During intrarenal injury, both urinary MCP-1 protein and MCP-1 gene activation

increased to a greater extent than NGAL. Moreover, uraemia in the absence of renal injury

induced the NGAL gene but not the MCP-1 gene. This suggests a possible higher specificity

for MCP-1 in AKI. Clinical findings in patients have endorsed this idea by discriminating

patients with and without AKI by use of urinary MCP-1 concentrations. It would therefore be

interesting to evaluate whether urine MCP-1 levels in our septic AKI models could also be an

early detector of septic AKI.

Determine inflammation and resolution at different time points in the long-term model

The source of MCP-1 and the role of the macrophage in renal injury and recovery require

further elucidation. To determine inflammation and resolution at different time points in our

long-term model, earlier and later timepoints (e.g. days 7 and 28) should be included.

Furthermore, it needs to be established if the macrophages belong to the M1 or M2

phenotypes. We propose to perform histological assessment of kidney sections and to confirm

macrophage specificity by double labelling with macrophage and immunofluorescent-labelled

M1 and M2 markers. To evaluate whether P2X7 correlates with inflammation or resolution in

this long-term model of sepsis and to assess fibrosis we are currently performing

immunohistochemistry with P2X7 and α-SMA-staining, and are awaiting formal

quantification.

Page 29: Groningen International Program of Science in Medicine ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/... · (iii) Detection of macrophages 20 3. Enzyme-linked immunosorbent assay’s

29

Dutch Summary

Sepsis is een overwelmende inflammatoire reactie op een infectie, wat kan leiden tot multi-

orgaan falen en de dood. Gedurende het septische proces raken de nieren vaak aangetast, wat

leidt tot dysfunctie of compleet nierfalen. Daarnaast hebben overlevers van sepsis-

geïnduceerd acuut nierfalen (AKI - acute kidney injury) een sterk verhoogd risico op het

ontwikkelen van eindstadium nierfalen. Het vrijkomen van pro-inflammatoire cytokines, zoals

IL-1β en IL-18, is geassocieerd met de pro-inflammatoire cascade die sepsis met zich

meebrengt en immunologische factoren lijken mee te dragen aan de ontwikkeling van

septisch-AKI. De mediatoren die aanvankelijk vrijkomen (in pro-vorm) zijn opzichzelf

insufficient. Een aanzienlijke rol voor de post-transcriptionele verwerking en het vrijlaten van

de actieve cytokines is via de formatie van een groot multi eiwitcomplex, het NLRP3

inflammasoom. De transmembrane cel receptor P2X7 (P2X7R) faciliteert de vorming van

NLRP3. Naast een belangrijke rol in de inflammatoire respons, speelt de P2X7R ook een rol

in apoptotische celdood. Opregulatie van deze receptor is geassocieerd met renale schade.

Daarentegen heeft A-438079, een selectieve P2X7 antagonist een reno-protectief effect in een

rat model met glomerulonefritis. Deze bevindingen wekken de suggestie dat P2X7R als een

potentieel therapeutisch doelwit kan worden gezien. P2X7 expressie is het best bekend in

immuuncellen zoals macrofagen, die een cruciale rol spelen in de initiatie, het onderhouden

van en de resolutie van inflammatie. Macrofagen komen vrij en worden beïnvloed door

chemokines zoals MCP-1. MCP-1 speelt een prominente rol in de acute inflammatoire

respons in meerdere modelen van nierziekten. Bovendien, induceert IL-1β (na maturatie en

vrijlating door de P2X7/inflammasome) expressie van MCP-1 door epitheelcellen.

Doel

Met de volgende werk hypothese; “P2X7R opregulatie veroorzaakt renale schade door lokale

productie van cytokines en chemokines”, heb ik onderzocht of renale P2X7 expressie

verhoogd is in een 3-daags rat-model met faecale peritonitis en vloeistofresuscitatie. Specifiek

heb ik gekeken naar een relatie met histopathologische veranderingen tijdens septisch-AKI en

naar de co-lokalisatie van macrofaag infiltratie.

In een ander lange termijn model (2 weken), met Zymosan-geinduceerde peritonitis, heb ik

onderzoek gedaan naar renale P2X7 expressie en naar de pathologische veranderingen vanuit

de vroege naar de ‘recovery’ fase.

Methoden

In het acute model werd sepsis geïnduceerd in geïnstrumenteerde, wakkere, mannelijke

Wistar ratten door middel van een intraperitoneale injectie van faecale ‘slurry’. Dit werd

gevolgd door vloeistofresuscitatie (10ml ml/kg/uur) vanaf 2 uur na het induceren van sepsis.

Sham (controle groep) ratten kregen hetzelfde vloeistof regime, maar geen i.p. injectie.

Septische en sham geopereerde ratten werden post-sepsis geofferd na 6 uur (n= 6 per groep)

of na 24 uur (n = 8 per groep). Bloed en nieren werden verkregen en geanalyseerd voor serum

creatinine, histologisch beoordeling van renale schade, P2X7 expressie, macrofaag infiltratie

en cytokine en chemokine expressie. Dit werd vergeleken met naïve, niet-geïnstrumenteerde

ratten (n=6).

In het lange termijn model, kregen niet-geinstrumenteerde mannelijke Wistar ratten een .i.p.

injectie met Zymosan (een glycaan afkomstig uit de celwand van een gist) voor een verlengd

septisch insult. Nieren werden verkregen op dag 2 (4 sham, 4 septische ratten) en dag 14 (4

sham, 8 sepsis) en geanalyseerd voor renale schade, macrofaag infiltratie en chemokine

expressie.

Page 30: Groningen International Program of Science in Medicine ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/... · (iii) Detection of macrophages 20 3. Enzyme-linked immunosorbent assay’s

30

Continue variabelen zijn weergegeven als gemiddelde ± SD. Parametrische data werd

geanalyseerd door middel van een unpaired T-test en non-parametrische data door middel van

de Mann Whitney U test. Verschillen tussen meer dan twee groepen continue variabelen werd

beoordeeld met one-way ANOVA. Door middel van de Post-hoc Tukey’s test werden

verschillen tussen groepen vastgesteld. Een p-waarde <0.05 werd als statistisch significant

beschouwd.

Resultaten

In het acute model werd een significant hoger serum creatinine gevonden in septisch-AKI na

24 uur (p<0.05). Echter, na zowel 6 en 24 uur werd geen significante histologische renale

schade gezien. In septische nieren was P2X7 expressie verhoogd na 24uur (p<0.05), een trend

was echter al zichtbaar na 6 uur. In ratten met een ernstige sepsis (met een 3x verhoogd serum

creatinine) was intraluminaal debris zichtbaar wat intens kleurde voor P2X7. Verhoogde

expressie van P2X7 correleerde met verhoogde niveau’s van IL-1β in zowel serum als in de

nieren. Serum IL-1β was verhoogd in septische dieren, deze niveaus daalden na 24 uur

(P<0.05). Daarentegen was renaal IL-1β verhoogd 6uur na sepsis, wat doorsteeg op 24 uur

(p<0.01). Tevens is septisch-AKI geassocieerd met verhoogde niveaus van renale expressie in

caspase-1 en pro IL-18. Na zowel 6 als 24 uur was er geen macrofaag infiltratie zichtbaar en

was er sprake van slechts minimale stijging in renale MCP-1 niveaus.

In het lange termijn model van sepsis werden tevens minimale aanwijzingen gevonden van

acuut renale schade op dag 2 en dag 14. P2X7 kleuring moet nog geanalyseerd worden, echter

macrofaag infiltratie was evident in zowel glomeruli en interstitium in alle septische dieren op

dag 14, terwijl macrofaag infiltratie afwezig was in alle dieren van de controlegroep.

De renale MCP-1 niveaus waren tevens significant verhoogd in de septische dieren (p<0.01)

op dit tijdpunt. Daarentegen, werd macrofaag infiltratie slechts in één van de 4 septische

dieren gezien op dag 2. Renale MCP-1 niveaus waren soortgelijk in shams.

Conclusies

Een significante verhoging in renale P2X7 expressie werd gezien in septische dieren na 24

uur, correlerend met verhoogde niveaus van IL-1β. In ernstig septische dieren werd een

distinctief kleurings patroon gevonden, suggestief voor een rol in celdood. Deze bevinding

tezamen met de afwezigheid van vroege macrofaag infiltratie, wekt de suggestie dat renaal

P2X7 hoogstwaarschijnlijk tot expressie gebracht wordt door intrinsieke cellen in de nier.

Ondanks biochemische afwijkingen van renale dysfunctie werd er geen significante

histologische renale schade gezien, dit reflecteert bevindingen in septische patienten. Verder

onderzoek is nodig om te evalueren of P2X7 antagonisme bescherming kan bieden.

In tegenstelling tot bevindingen in het acute model, werd in het lange termijn model van

sepsis significant macrofaag infiltratie gezien op dag 14, wat in zowel de glomeruli als het

interstitium zichtbaar was. Dit correspondeert met verhoogde niveaus van renaal MCP-1. Of

dit bijdraagt aan lange-termijn schade in de nieren behoeft verder onderzoek.

Page 31: Groningen International Program of Science in Medicine ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/... · (iii) Detection of macrophages 20 3. Enzyme-linked immunosorbent assay’s

31

Acknowledgments

I sincerely would like to thank all those who have given me their generous assistance, support and

guidance during the realization of my thesis. Not only could I not have performed my experiments

without their contributions but their uplifting feedback and energy gave me the opportunity to

start each day with renewed energy and kept me on my feet during the ups and downs of the entire

process.

I would particularly like to thank:

Prof. Mervyn Singer, for your guidance and support. It has been a great honour to be given the

opportunity to participate in the research with your group. You are a true role model of how to

lead a team; despite your tedious rushed schedules you would always make sure your door was

open for any questions or help in other sorts of ways. With your everlasting enthusiasm and

cheerful comments (even despite your awful taste for picking nicknames), I felt part of ‘the Singer

family’ straight away.

A special thanks to Dr. Nish Arulkumaran who provided me with great opportunities, both in and

outside the lab. I could not have succeeded in finishing this project without your generous

assistance. Your contagious ardour about science and uplifting motto kept me motivated, while

keeping my expectations right. It has been a real pleasure working with you!

I would also like to thank Dr. Frederick Tam and Prof. Charles Pusey for their hospitality in the

lab, Dr. Paul Bass, for his assistance and feedback regarding the histopathology and Prof. Robert

Unwin. Also a warm thanks to all the other members of the Research Department of Clinical

Physiology, UCL and the Renal Research Department at Hammersmith Hospital, Imperial for the

interesting and useful laboratory meetings and happy times working in the lab. Specifically I

would like to thank Gurtjeet Bangal and Clare Turner for their generous help in teaching my all

the ins and outs about immunohistochemistry.

I thank the GIPS-M committee who gave me the opportunity to participate in the GIPS-M

program, in specific Prof. Cees Kallenberg for his time and effort during my preparations and for

reviewing this thesis.

In particular, I would like to thank Maarten for his everlasting support, motivation and believe.

Your hardworking and positive mindset always 'boost' me to achieve my goals. Last but definitely

not least, I would like to thank my family for encouraging me throughout my life and education,

and my friends for the very much appreciated distraction and laughter in my spare time and for

always being there for me. I hope I can return you all with the same amount of love and laughter

on our continuing journey.

Sponsors I would like to thank the following sponsors for their financial support:

Nierstichting Nederland,

Fundatie van Vrijvrouwe van Renswoude,

Marco Polo Fonds,

JO Kolk studiefonds voor vrouwen,

Jan Kornelis de Cock Stichting,

Groninger Universiteisfonds.

Page 32: Groningen International Program of Science in Medicine ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/... · (iii) Detection of macrophages 20 3. Enzyme-linked immunosorbent assay’s

32

References

1. Angus DC, Linde-Zwirble WT, Lidicker J, Clermont G, Carcillo J, Pinsky MR. Epidemiology of severe sepsis in

the United States: analysis of incidence, outcome, and associated costs of care. Crit Care Med. 2001;29(7):1303-10.

2. Martin GS, Mannino DM, Eaton S, Moss M. The epidemiology of sepsis in the United States from 1979 through

2000. N Engl J Med. 2003;348(16):1546-54.

3. Linde-Zwirble WT, Angus DC. Severe sepsis epidemiology: sampling, selection, and society. Crit Care.

2004;8(4):222-6.

4. Dombrovskiy VY, Martin AA, Sunderram J, Paz HL. Rapid increase in hospitalization and mortality rates for

severe sepsis in the United States: a trend analysis from 1993 to 2003. Crit Care Med. 2007;35(5):1244-50.

5. Hotchkiss RS, Karl IE. The pathophysiology and treatment of sepsis. N Engl J Med. 2003;348(2):138-50.

6. Hoste EA, Kellum JA. Incidence, classification, and outcomes of acute kidney injury. Contrib Nephrol.

2007;156:32-8.

7. Hoste EA, Clermont G, Kersten A, Venkataraman R, Angus DC, De Bacquer D, et al. RIFLE criteria for acute

kidney injury are associated with hospital mortality in critically ill patients: a cohort analysis. Crit Care.

2006;10(3):12.

8. Metnitz PG, Krenn CG, Steltzer H, Lang T, Ploder J, Lenz K, et al. Effect of acute renal failure requiring renal

replacement therapy on outcome in critically ill patients. Crit Care Med. 2002;30(9):2051-8.

9. Bagshaw SM, George C, Bellomo R. A comparison of the RIFLE and AKIN criteria for acute kidney injury in

critically ill patients. Nephrol Dial Transplant. 2008;23(5):1569-74.

10. Uchino S, Kellum JA, Bellomo R, Doig GS, Morimatsu H, Morgera S, et al. Acute renal failure in critically ill

patients: a multinational, multicenter study. Jama. 2005;294(7):813-8.

11. Joannidis M, Metnitz PG. Epidemiology and natural history of acute renal failure in the ICU. Crit Care Clin.

2005;21(2):239-49.

12. Chertow GM, Burdick E, Honour M, Bonventre JV, Bates DW. Acute kidney injury, mortality, length of stay, and

costs in hospitalized patients. J Am Soc Nephrol. 2005;16(11):3365-70.

13. Bellomo R, Ronco C, Kellum JA, Mehta RL, Palevsky P. Acute renal failure - definition, outcome measures,

animal models, fluid therapy and information technology needs: the Second International Consensus Conference of

the Acute Dialysis Quality Initiative (ADQI) Group. Crit Care. 2004;8(4):24.

14. Mehta RL, Kellum JA, Shah SV, Molitoris BA, Ronco C, Warnock DG, et al. Acute Kidney Injury Network: report

of an initiative to improve outcomes in acute kidney injury: Crit Care. 2007;11(2):R31.

15. KDIGO Clinical Practice Guideline for Acute Kidney Injury. KDIGO Clinical Practice Guideline for Acute Kidney

Injury. PAPER 2012;2(1).

16. Doi K, Yuen PS, Eisner C, Hu X, Leelahavanichkul A, Schnermann J, et al. Reduced production of creatinine

limits its use as marker of kidney injury in sepsis. J Am Soc Nephrol. 2009;20(6):1217-21.

17. Langenberg C, Bellomo R, May C, Wan L, Egi M, Morgera S. Renal blood flow in sepsis. Crit Care. 2005;9(4):24.

18. Langenberg C, Wan L, Egi M, May CN, Bellomo R. Renal blood flow in experimental septic acute renal failure.

Kidney Int. 2006;69(11):1996-2002.

19. Langenberg C, Wan L, Egi M, May CN, Bellomo R. Renal blood flow and function during recovery from

experimental septic acute kidney injury. Intensive Care Med. 2007;33(9):1614-8.

20. Le Dorze M, Legrand M, Payen D, Ince C. The role of the microcirculation in acute kidney injury. Curr Opin Crit

Care. 2009;15(6):503-8.

21. Brezis M, Rosen S. Hypoxia of the renal medulla--its implications for disease. N Engl J Med. 1995;332(10):647-

55.

22. Vinay Kumar (Editor) RSCE, Stanley L. Robbins (Editor) Basic Pathology. 7th ed2002 june 15th.

23. Lerolle N, Nochy D, Guerot E, Bruneval P, Fagon JY, Diehl JL, et al. Histopathology of septic shock induced acute

kidney injury: apoptosis and leukocytic infiltration. Intensive Care Med. 2010;36(3):471-8. Epub 2009/11/20.

24. Langenberg C, Bagshaw SM, May CN, Bellomo R. The histopathology of septic acute kidney injury: a systematic

review. Crit Care. 2008;12(2):R38. Epub 2008/03/08.

25. Lichtman Ae. Basic Immunology, Functions & disorders of the immune system. 2nd ed.

26. Kono H, Rock KL. How dying cells alert the immune system to danger. Nat Rev Immunol. 2008;8(4):279-89.

27. Bours MJ, Swennen EL, Di Virgilio F, Cronstein BN, Dagnelie PC. Adenosine 5'-triphosphate and adenosine as

endogenous signaling molecules in immunity and inflammation. Pharmacol Ther. 2006;112(2):358-404.

28. Shi Y, Evans JE, Rock KL. Molecular identification of a danger signal that alerts the immune system to dying cells.

Nature. 2003;425(6957):516-21.

29. Drury AN, Szent-Gyorgyi A. The physiological activity of adenine compounds with especial reference to their

action upon the mammalian heart. J Physiol. 1929;68(3):213-37.

30. Kumar H, Kawai T, Akira S. Toll-like receptors and innate immunity. Biochem Biophys Res Commun.

2009;388(4):621-5.

31. Armstrong L, Medford AR, Hunter KJ, Uppington KM, Millar AB. Differential expression of Toll-like receptor

(TLR)-2 and TLR-4 on monocytes in human sepsis. Clin Exp Immunol. 2004;136(2):312-9.

32. Zhang B, Ramesh G, Uematsu S, Akira S, Reeves WB. TLR4 signaling mediates inflammation and tissue injury in

nephrotoxicity. J Am Soc Nephrol. 2008;19(5):923-32.

33. Aderem A, Ulevitch RJ. Toll-like receptors in the induction of the innate immune response. Nature.

2000;406(6797):782-7.

34. Anders HJ. Toll-like receptors and danger signaling in kidney injury. J Am Soc Nephrol. 2010;21(8):1270-4.

Page 33: Groningen International Program of Science in Medicine ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/... · (iii) Detection of macrophages 20 3. Enzyme-linked immunosorbent assay’s

33

35. Dinarello CA. Immunological and inflammatory functions of the interleukin-1 family. Annu Rev Immunol.

2009;27:519-50.

36. Ferrari D, Chiozzi P, Falzoni S, Hanau S, Di Virgilio F. Purinergic modulation of interleukin-1 beta release from

microglial cells stimulated with bacterial endotoxin. J Exp Med. 1997;185(3):579-82.

37. Mehta VB, Hart J, Wewers MD. ATP-stimulated release of interleukin (IL)-1beta and IL-18 requires priming by

lipopolysaccharide and is independent of caspase-1 cleavage. J Biol Chem. 2001;276(6):3820-6.

38. Bauernfeind FG, Horvath G, Stutz A, Alnemri ES, MacDonald K, Speert D, et al. Cutting edge: NF-kappaB

activating pattern recognition and cytokine receptors license NLRP3 inflammasome activation by regulating

NLRP3 expression. J Immunol. 2009;183(2):787-91.

39. Bauernfeind F, Bartok E, Rieger A, Franchi L, Nunez G, Hornung V. Cutting edge: reactive oxygen species

inhibitors block priming, but not activation, of the NLRP3 inflammasome. J Immunol. 2011;187(2):613-7.

40. Bergsbaken T, Fink SL, den Hartigh AB, Loomis WP, Cookson BT. Coordinated host responses during pyroptosis:

caspase-1-dependent lysosome exocytosis and inflammatory cytokine maturation. J Immunol. 2011;187(5):2748-

54.

41. Miao EA, Rajan JV, Aderem A. Caspase-1-induced pyroptotic cell death. Immunol Rev. 2011;243(1):206-14.

42. North RA. Molecular physiology of P2X receptors. Physiol Rev. 2002;82(4):1013-67.

43. Di Virgilio F. The P2Z purinoceptor: an intriguing role in immunity, inflammation and cell death. Immunol Today.

1995;16(11):524-8.

44. Ferrari D, Chiozzi P, Falzoni S, Dal Susino M, Melchiorri L, Baricordi OR, et al. Extracellular ATP triggers IL-1

beta release by activating the purinergic P2Z receptor of human macrophages. J Immunol. 1997;159(3):1451-8.

45. Solle M, Labasi J, Perregaux DG, Stam E, Petrushova N, Koller BH, et al. Altered cytokine production in mice

lacking P2X(7) receptors. J Biol Chem. 2001;276(1):125-32.

46. Humphreys BD, Dubyak GR. Modulation of P2X7 nucleotide receptor expression by pro- and anti-inflammatory

stimuli in THP-1 monocytes. J Leukoc Biol. 1998;64(2):265-73.

47. Walev I, Reske K, Palmer M, Valeva A, Bhakdi S. Potassium-inhibited processing of IL-1 beta in human

monocytes. Embo J. 1995;14(8):1607-14.

48. Bryan NB, Dorfleutner A, Rojanasakul Y, Stehlik C. Activation of inflammasomes requires intracellular

redistribution of the apoptotic speck-like protein containing a caspase recruitment domain. J Immunol.

2009;182(5):3173-82.

49. Perregaux D, Gabel CA. Interleukin-1 beta maturation and release in response to ATP and nigericin. Evidence that

potassium depletion mediated by these agents is a necessary and common feature of their activity. J Biol Chem.

1994;269(21):15195-203.

50. Laliberte RE, Eggler J, Gabel CA. ATP treatment of human monocytes promotes caspase-1 maturation and

externalization. J Biol Chem. 1999;274(52):36944-51.

51. Brough D, Le Feuvre RA, Iwakura Y, Rothwell NJ. Purinergic (P2X7) receptor activation of microglia induces cell

death via an interleukin-1-independent mechanism. Mol Cell Neurosci. 2002;19(2):272-80.

52. Verhoef PA, Kertesy SB, Lundberg K, Kahlenberg JM, Dubyak GR. Inhibitory effects of chloride on the activation

of caspase-1, IL-1beta secretion, and cytolysis by the P2X7 receptor. J Immunol. 2005;175(11):7623-34.

53. Brough D, Rothwell NJ. Caspase-1-dependent processing of pro-interleukin-1beta is cytosolic and precedes cell

death. J Cell Sci. 2007;120(Pt 5):772-81.

54. MacKenzie A, Wilson HL, Kiss-Toth E, Dower SK, North RA, Surprenant A. Rapid secretion of interleukin-1beta

by microvesicle shedding. Immunity. 2001;15(5):825-35.

55. Black R, Kronheim S, Sleath P, Greenstreet T, Virca GD, March C, et al. The proteolytic activation of interleukin-1

beta. Agents Actions Suppl. 1991;35:85-9.

56. Hazuda DJ, Strickler J, Kueppers F, Simon PL, Young PR. Processing of precursor interleukin 1 beta and

inflammatory disease. J Biol Chem. 1990;265(11):6318-22.

57. Mizutani H, Schechter N, Lazarus G, Black RA, Kupper TS. Rapid and specific conversion of precursor interleukin

1 beta (IL-1 beta) to an active IL-1 species by human mast cell chymase. J Exp Med. 1991;174(4):821-5.

58. Irmler M, Hertig S, MacDonald HR, Sadoul R, Becherer JD, Proudfoot A, et al. Granzyme A is an interleukin 1

beta-converting enzyme. J Exp Med. 1995;181(5):1917-22.

59. Dinarello CA. Interleukin-18 and the pathogenesis of inflammatory diseases. Semin Nephrol. 2007;27(1):98-114.

60. Dinarello CA. The IL-1 family and inflammatory diseases. Clin Exp Rheumatol. 2002;20(5 Suppl 27):S1-13.

61. Reddy P. Interleukin-18: recent advances. Curr Opin Hematol. 2004;11(6):405-10.

62. Bellomo R, Bagshaw S, Langenberg C, Ronco C. Pre-renal azotemia: a flawed paradigm in critically ill septic

patients? Contrib Nephrol. 2007;156:1-9.

63. Bagshaw SM, Langenberg C, Haase M, Wan L, May CN, Bellomo R. Urinary biomarkers in septic acute kidney

injury. Intensive Care Med. 2007;33(7):1285-96.

64. Ferrari D, Pizzirani C, Adinolfi E, Lemoli RM, Curti A, Idzko M, et al. The P2X7 receptor: a key player in IL-1

processing and release. J Immunol. 2006;176(7):3877-83.

65. Gallucci S, Matzinger P. Danger signals: SOS to the immune system. Curr Opin Immunol. 2001;13(1):114-9.

66. Di Virgilio F. Purinergic mechanism in the immune system: A signal of danger for dendritic cells. Purinergic

Signal. 2005;1(3):205-9.

67. Abbracchio MP, Burnstock G, Verkhratsky A, Zimmermann H. Purinergic signalling in the nervous system: an

overview. Trends Neurosci. 2009;32(1):19-29.

68. Turner CM, Vonend O, Chan C, Burnstock G, Unwin RJ. The pattern of distribution of selected ATP-sensitive P2

receptor subtypes in normal rat kidney: an immunohistological study. Cells Tissues Organs. 2003;175(2):105-17.

Page 34: Groningen International Program of Science in Medicine ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/... · (iii) Detection of macrophages 20 3. Enzyme-linked immunosorbent assay’s

34

69. Unwin RJ, Bailey MA, Burnstock G. Purinergic signaling along the renal tubule: the current state of play. News

Physiol Sci. 2003;18:237-41.

70. Booth JW, Tam FW, Unwin RJ. P2 purinoceptors: Renal pathophysiology and therapeutic potential. Clin Nephrol.

2012;78(2):154-63.

71. Turner CM, Tam FW, Lai PC, Tarzi RM, Burnstock G, Pusey CD, et al. Increased expression of the pro-apoptotic

ATP-sensitive P2X7 receptor in experimental and human glomerulonephritis. Nephrol Dial Transplant.

2007;22(2):386-95.

72. Hillman KA, Burnstock G, Unwin RJ. The P2X7 ATP receptor in the kidney: a matter of life or death? Nephron

Exp Nephrol. 2005;101(1):27.

73. Tam FW. Current pharmacotherapy for the treatment of crescentic glomerulonephritis. Expert Opin Investig Drugs.

2006;15(11):1353-69.

74. Ferrari D, Villalba M, Chiozzi P, Falzoni S, Ricciardi-Castagnoli P, Di Virgilio F. Mouse microglial cells express a

plasma membrane pore gated by extracellular ATP. J Immunol. 1996;156(4):1531-9.

75. Labasi JM, Petrushova N, Donovan C, McCurdy S, Lira P, Payette MM, et al. Absence of the P2X7 receptor alters

leukocyte function and attenuates an inflammatory response. J Immunol. 2002;168(12):6436-45.

76. Chessell IP, Hatcher JP, Bountra C, Michel AD, Hughes JP, Green P, et al. Disruption of the P2X7 purinoceptor

gene abolishes chronic inflammatory and neuropathic pain. Pain. 2005;114(3):386-96.

77. Surprenant A, Rassendren F, Kawashima E, North RA, Buell G. The cytolytic P2Z receptor for extracellular ATP

identified as a P2X receptor (P2X7). Science. 1996;272(5262):735-8.

78. Miller CM, Boulter NR, Fuller SJ, Zakrzewski AM, Lees MP, Saunders BM, et al. The role of the P2X(7) receptor

in infectious diseases. PLoS Pathog. 2011;7(11):10.

79. Schulze-Lohoff E, Hugo C, Rost S, Arnold S, Gruber A, Brune B, et al. Extracellular ATP causes apoptosis and

necrosis of cultured mesangial cells via P2Z/P2X7 receptors. Am J Physiol. 1998;275(6 Pt 2):F962-71.

80. Harada H, Chan CM, Loesch A, Unwin R, Burnstock G. Induction of proliferation and apoptotic cell death via P2Y

and P2X receptors, respectively, in rat glomerular mesangial cells. Kidney Int. 2000;57(3):949-58.

81. Rassendren F, Buell GN, Virginio C, Collo G, North RA, Surprenant A. The permeabilizing ATP receptor, P2X7.

Cloning and expression of a human cDNA. J Biol Chem. 1997;272(9):5482-6.

82. Taylor SR, Turner CM, Elliott JI, McDaid J, Hewitt R, Smith J, et al. P2X7 deficiency attenuates renal injury in

experimental glomerulonephritis. J Am Soc Nephrol. 2009;20(6):1275-81.

83. Tang WW, Qi M, Warren JS. Monocyte chemoattractant protein 1 mediates glomerular macrophage infiltration in

anti-GBM Ab GN. Kidney Int. 1996;50(2):665-71.

84. Wada T, Yokoyama H, Furuichi K, Kobayashi KI, Harada K, Naruto M, et al. Intervention of crescentic

glomerulonephritis by antibodies to monocyte chemotactic and activating factor (MCAF/MCP-1). Faseb J.

1996;10(12):1418-25.

85. Shimizu S, Nakashima H, Masutani K, Inoue Y, Miyake K, Akahoshi M, et al. Anti-monocyte chemoattractant

protein-1 gene therapy attenuates nephritis in MRL/lpr mice. Rheumatology. 2004;43(9):1121-8.

86. Rice JC, Spence JS, Yetman DL, Safirstein RL. Monocyte chemoattractant protein-1 expression correlates with

monocyte infiltration in the post-ischemic kidney. Ren Fail. 2002;24(6):703-23.

87. Takasu O, Gaut JP, Watanabe E, To K, Fagley RE, Sato B, et al. Mechanisms of cardiac and renal dysfunction in

patients dying of sepsis. Am J Respir Crit Care Med. 2013;187(5):509-17.

88. Chawla LS, Amdur RL, Amodeo S, Kimmel PL, Palant CE. The severity of acute kidney injury predicts

progression to chronic kidney disease. Kidney Int. 2011;79(12):1361-9.

89. Lo LJ, Go AS, Chertow GM, McCulloch CE, Fan D, Ordonez JD, et al. Dialysis-requiring acute renal failure

increases the risk of progressive chronic kidney disease. Kidney Int. 2009;76(8):893-9.

90. Brealey D, Karyampudi S, Jacques TS, Novelli M, Stidwill R, Taylor V, et al. Mitochondrial dysfunction in a long-

term rodent model of sepsis and organ failure. Am J Physiol Regul Integr Comp Physiol. 2004;286(3):6.

91. Dinarello CA. Interleukin-1 beta, interleukin-18, and the interleukin-1 beta converting enzyme. Ann N Y Acad Sci.

1998;856:1-11.

92. Rivers E, Nguyen B, Havstad S, Ressler J, Muzzin A, Knoblich B, et al. Early goal-directed therapy in the

treatment of severe sepsis and septic shock. N Engl J Med. 2001;345(19):1368-77.

93. Dyson A, Rudiger A, Singer M. Temporal changes in tissue cardiorespiratory function during faecal peritonitis.

Intensive Care Med. 2011;37(7):1192-200.

94. Dyson A, Cone S, Singer M, Ackland GL. Microvascular and macrovascular flow are uncoupled in early

polymicrobial sepsis. Br J Anaesth. 2012;108(6):973-8.

95. Langenberg C, Bagshaw SM, May CN, Bellomo R. The histopathology of septic acute kidney injury: a systematic

review. Crit Care. 2008;12(2):6.

96. Groschel-Stewart U, Bardini M, Robson T, Burnstock G. Localisation of P2X5 and P2X7 receptors by

immunohistochemistry in rat stratified squamous epithelia. Cell Tissue Res. 1999;296(3):599-605.

97. Groschel-Stewart U, Bardini M, Robson T, Burnstock G. P2X receptors in the rat duodenal villus. Cell Tissue Res.

1999;297(1):111-7.

98. Bardini M, Lee HY, Burnstock G. Distribution of P2X receptor subtypes in the rat female reproductive tract at late

pro-oestrus/early oestrus. Cell Tissue Res. 2000;299(1):105-13.

99. Grahames CB, Michel AD, Chessell IP, Humphrey PP. Pharmacological characterization of ATP- and LPS-induced

IL-1beta release in human monocytes. Br J Pharmacol. 1999;127(8):1915-21.

100. Zheng LM, Zychlinsky A, Liu CC, Ojcius DM, Young JD. Extracellular ATP as a trigger for apoptosis or

programmed cell death. J Cell Biol. 1991;112(2):279-88.

Page 35: Groningen International Program of Science in Medicine ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/... · (iii) Detection of macrophages 20 3. Enzyme-linked immunosorbent assay’s

35

101. Coutinho-Silva R, Persechini PM, Bisaggio RD, Perfettini JL, Neto AC, Kanellopoulos JM, et al. P2Z/P2X7

receptor-dependent apoptosis of dendritic cells. Am J Physiol. 1999;276(5 Pt 1):C1139-47.

102. Wolf G, Shankland SJ. Cell cycle control in glomerular disease. Prog Cell Cycle Res. 2003;5:71-9.

103. Sugiyama H, Kashihara N, Makino H. [Role of apoptosis in renal injury]. Nihon Rinsho. 1996;54(7):1975-81.

104. Niemir ZI, Stein H, Dworacki G, Mundel P, Koehl N, Koch B, et al. Podocytes are the major source of IL-1 alpha

and IL-1 beta in human glomerulonephritides. Kidney Int. 1997;52(2):393-403.

105. Tesch GH, Yang N, Yu H, Lan HY, Foti R, Chadban SJ, et al. Intrinsic renal cells are the major source of

interleukin-1 beta synthesis in normal and diseased rat kidney. Nephrol Dial Transplant. 1997;12(6):1109-15.

106. Tang WW, Feng L, Mathison JC, Wilson CB. Cytokine expression, upregulation of intercellular adhesion

molecule-1, and leukocyte infiltration in experimental tubulointerstitial nephritis. Lab Invest. 1994;70(5):631-8.

107. Sica A, Wang JM, Colotta F, Dejana E, Mantovani A, Oppenheim JJ, et al. Monocyte chemotactic and activating

factor gene expression induced in endothelial cells by IL-1 and tumor necrosis factor. J Immunol.

1990;144(8):3034-8.

108. Yamamoto-Shuda Y, Nakayama K, Saito T, Natori Y. Therapeutic effect of glucocorticoid on experimental

crescentic glomerulonephritis. J Lab Clin Med. 1999;134(4):410-8.

109. Tesch GH, Schwarting A, Kinoshita K, Lan HY, Rollins BJ, Kelley VR. Monocyte chemoattractant protein-1

promotes macrophage-mediated tubular injury, but not glomerular injury, in nephrotoxic serum nephritis. J Clin

Invest. 1999;103(1):73-80.

110. Koul HK. Role of p38 MAP kinase signal transduction in apoptosis and survival of renal epithelial cells. Ann N Y

Acad Sci. 2003:62-5.

111. Murray PJ, Wynn TA. Protective and pathogenic functions of macrophage subsets. Nat Rev Immunol.

2011;11(11):723-37.

112. Murray PJ, Wynn TA. Obstacles and opportunities for understanding macrophage polarization. J Leukoc Biol.

2011;89(4):557-63.

113. Rudiger A, Dyson A, Felsmann K, Carre JE, Taylor V, Hughes S, et al. Early functional and transcriptomic

changes in the myocardium predict outcome in a long-term rat model of sepsis. Clin Sci. 2013;124(6):391-401.

114. Vanmassenhove J, Vanholder R, Nagler E, Van Biesen W. Urinary and serum biomarkers for the diagnosis of acute

kidney injury: an in-depth review of the literature. Nephrol Dial Transplant. 2013;28(2):254-73.

115. Munshi R, Johnson A, Siew ED, Ikizler TA, Ware LB, Wurfel MM, et al. MCP-1 gene activation marks acute

kidney injury. J Am Soc Nephrol. 2011;22(1):165-75.

Page 36: Groningen International Program of Science in Medicine ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/... · (iii) Detection of macrophages 20 3. Enzyme-linked immunosorbent assay’s

36

Appendices I – Buffers and solutions

1. RadioImmunoPrecipitation Assay (RIPA) buffer: 50 mM Tris [pH 7.4], 150 mM

NaCl, 1% NP-40, 0.25% sodium deoxycholate, 0.1% , 1mM EDTA supplemented

with 10% protease inhibitors (Sigma-Aldrich Co)

2. SDS-sample buffer: 0.125M Tris-HCL [pH6.8], 4% SDS, 20% (v/v) glycerol, 0.6M

β-mercaptoethanol and 0.02% Bromophenol blue

3. 12% SDS-PAGE Tris-glycine minigels. The SDS-page gel consisted of a stacking

gel (i) overlaid onto a resolving gel (ii)

(i) 5% stacking gel: 30% Acrylamide/bis solution (ratio 37.5:1) in a buffer of 1.5M

Tric-HCL [pH 6.8] containing 0.1% (w/v) SDS . Polymerisation is initiated by

addition of 0.1% (w/v) ammonium persulphate and 0.05% (v/v)

tetramethylethylenediamine (TEMED).

(ii) 12% Resolving gel: 10% of a 30% Acrylamide/bis solution (ratio 37.5:1) in a

buffer of 0.375M Tris-HCL [pH 8.8] containing 0.1% SDS. Polymerisation is

initiated by addition of 0.1% (w/v) ammonium persulphate and 0.016% (v/v)

tetramethylethylenediamine (TEMED).

4. Running Buffer: 3,03g Tris base, 14,4g Glycine, 1g SDS make up to 1L with dH2O

5. Transfer (Towbin) buffer 3g 25mM Tris, 14,4g 192mM Glycine, 10% methanol

make up to 1L with dH2O

6. Tris-buffered saline (TBS): 2,42g 20mMTris, 8,2g 140mM NaCl, make up to 1L

with dH2O and adjust for [pH 7.6] with 1.0M HCL

7. 0.01M citrate buffer [pH 6.0]: 2.94g Tri-sodium citrate to 1L dH20, adjust for [pH

6.0] with 1.0M HCL

8. PBS: 8g NaCl + 1,15g Na2NPO4 + 0,2g KCL + 0,2g KH2PO4 make up to 1L with

dH2O

9. 0.05M Tris-HCL [pH 7.2]: 1.576g Trizma HCL to 200mL dH20 and adjust for [pH

7.2] with NaOH

10. 0.1M Sodium Carbonate [pH 9.5]: 7,13g NaHCO3 + 1,59g Na2CO3, make up to 1L

with dH20 and adjust for pH with 10N NaOH

Page 37: Groningen International Program of Science in Medicine ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/... · (iii) Detection of macrophages 20 3. Enzyme-linked immunosorbent assay’s

37

P2X7 Receptor and Haemorrhage-reperfusion- Induced Acute Tubular Injury

1M Sixma,

1A Dyson,

1, 2, 3N Arulkumaran,

2P Bass,

2RJ Unwin,

3F Tam,

1M Singer

1Bloomsbury

Institute of Intensive Care Medicine and

2Dept of Nephrology, University College London, UK and

3Imperial College Kidney and Transplant Institute, Hammersmith Hospital, London UK

INTRODUCTION

The P2X7 purinoreceptor (P2X7R) triggers activation of the inflammasome with release of pro-inflammatory

cytokines (e.g. IL-1β, IL-18) and the pro-apoptotic caspase-1. Constitutive expression of the P2X7R in the

kidney is minimal. However, in a rat glomerulonephritis model (1), expression, mainly localized to the

glomeruli, was upregulated, with selective P2X7R receptor antagonism being protective.

OBJECTIVES To determine the pattern of renal P2X7R expression in a rat model of severe haemorrhage-reperfusion injury.

METHODS

Anaesthetized male Wistar rats underwent insertion of carotid arterial and jugular venous lines. After 30 min

stabilization, 50% estimated circulating blood volume was removed from the arterial line over 15 min. Animals

were monitored for a further 90 min prior to resuscitation. This was immediately followed by administration of

shed blood over 15 min followed by a background infusion of n-saline (10 ml/kg/hr). Animals were culled at 6

hours post-reperfusion with kidneys taken for analysis. Paraffin-embedded kidney sections were stained for

periodic acid Schiff (PAS) and P2X7 (anti- P2X7 primary antibody, Alamone,).

RESULTS Major histological damage was seen, including loss of brush border from tubular epithelial cells, tubular casts,

dilated tubules and tubular cell death (Figure 1). Immunohistochemistry demonstrated widespread upregulation

of renal P2X7R in tubules of rats that underwent haemorrhage-reperfusion injury but none in naïve rats. P2X7R

expression was localized to areas of tubular damage (Figure 1). However, the glomeruli were negative for P2X7.

CONCLUSIONS

In this rat model of severe haemorrhage-reperfusion injury, there was histological evidence of significant tubular

injury. Coexistence of tubular injury and P2X7R upregulation suggests that P2X7R is implicated in the

pathophysiology of AKI. Further work is required to determine the functional significance of tubular P2X7R, and

the potential benefit of P2X7 receptor antagonism in haemorrhage-reperfusion injury-induced AKI.

References:

(1). Taylor SR et al. P2X7 deficiency attenuates renal injury in experimental glomerulonephritis. J Am Soc Neph

2009; 20:1275-81

Grant acknowledgement:

NA Wellcome, MS Dutch Kidney Foundation

Figure 1: Immunohistochemistry demonstrating severe renal tubular injury (left panel), with P2X7R

(stained brown) in kidneys taken from naïve (middle panel), and haemorrhage reperfusion rats (right

panel)

Page 38: Groningen International Program of Science in Medicine ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/... · (iii) Detection of macrophages 20 3. Enzyme-linked immunosorbent assay’s

38

Renal macrophage infiltration in a rat model of sepsis and recovery

N. Arulkumaran, M. Sixma, S Saeed, G Bangal, P Bass, F Tam, M. Singer

INTRODUCTION

Despite minimal histological damage (1), survivors of acute kidney injury (AKI) are at risk of developing

chronic kidney disease (2). Most of the focus of AKI has been on the acute injury phase but little attention to

date has been paid to recovery. Mechanisms underlying resolution of AKI in sepsis are poorly understood and

may be potentially injurious.

OBJECTIVES

To describe temporal changes in renal inflammation and recovery following sepsis in a long-term rat model of

zymosan-induced peritonitis

METHODS

Intraperitoneal zymosan was injected into male Wistar rats under isoflurane anaesthesia. Sham animals received

intraperitoneal saline. At day 2 or day 14, animals were culled and kidneys taken for analysis. Renal tissue was

homogenized and MCP-1 (monocyte chemoattractant protein-1) levels were measured by ELISA (R&D

-1 is a key chemokine regulating

monocyte/macrophage migration and infiltration. Paraffin-embedded kidney sections were stained for

macrophages (anti-ED-1 primary antibody, Abcam). Statistics were performed using independent t-tests. A p-

value of <0.05 was taken as being statistically significant. Data are presented as mean ± standard deviation, p-

value.

RESULTS In this model the severity of illness (clinical severity, organ dysfunction, weight loss, food intake) peaks at day 2

with gradual recovery over the subsequent 12 days. At Day 2 there was minimal renal cell damage with similar

renal MCP-1 -1

Macrophage infiltration was evident in glomeruli and interstitium in all day 14 septic animals but in none of the

sham animals (Figure 1). At day 2, just 1 septic animal demonstrated renal macrophage infiltration while 1 sham

animal had minimal glomerular macrophage infiltration.

CONCLUSIONS

In the recovery phase of sepsis in this long-term rodent model, significant renal macrophage infiltration was

present within the glomeruli and interstitium with a corresponding increase in renal MCP-1. These changes were

not seen during the acute injury phase. The source of MCP-1 and the role of the macrophage in renal injury and

recovery require further elucidation.

References

(1) Takasu O, et al. Mechanisms of cardiac and renal dysfunction in patients dying of sepsis. Am J Respir Crit

Care Med 2013;187:509–17. (2) Chawla LS, et al. The severity of acute kidney injury predicts progression to

chronic kidney disease. Kidney Int 2011; 79:1361-9.

Grant acknowledgment

SS is supported by a Medical Research Council (UK) training fellowship. NA is supported by a Wellcome Trust

training fellowship. MS is supported by the Dutch Kidney Foundation.

Figure 1: Immunohistochemistry for ED-1 (stained brown) in septic (Day 14) rat in the glomerulus (left

panel) and interstitium (right panel).

Page 39: Groningen International Program of Science in Medicine ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/... · (iii) Detection of macrophages 20 3. Enzyme-linked immunosorbent assay’s

39

Temporal changes in renal haemodynamics and oxygenation in a rat model of sepsis

1,2,3

N Arulkumaran, 1M Sixma,

2P Bass,

3F Tam,

2RJ Unwin,

1M Singer

1Bloomsbury

Institute of Intensive Care Medicine and

2Dept of Nephrology, University College London, UK and

3Imperial College Kidney and Transplant Institute, Hammersmith Hospital, London UK

INTRODUCTION

Postulated mechanisms for sepsis-induced acute kidney injury (AKI) include altered global and intra-renal

haemodynamics and bioenergetic dysfunction. However, temporal changes are not well elucidated.

OBJECTIVES

To characterize temporal changes in renal haemodynamics and tissue oxygenation in a long-term, fluid-

resuscitated rat model of faecal peritonitis.

METHODS

Tunnelled central venous lines were inserted into male Wistar rats under isoflurane anaesthesia. Twenty-four

hours later, sepsis was induced by intraperitoneal injection of faecal slurry (n=14). Sixteen animals served as

sham-operated controls. Fluid resuscitation (10 ml/kg/hr) was commenced at 2h post-slurry. At either 6hr or 24hr

animals were terminally anaesthetized and instrumented for measurement of cardiac output (echocardiography),

renal blood flow (ultrasonic flow probe), renal cortical tissue oxygen tension (Oxylite sensor), and renal oxygen

extraction ratio (from the renal arterio-venous oxygen difference). Statistics were performed using independent t-

tests. P values <0.05 were taken as statistically significant. Data are presented as mean ± standard deviation.

RESULTS

At 6 hours, septic animals had a lower renal vascular resistance (MAP/RBF) (0.072±0.001 vs. 0.084±0.011

sham, p<0.05) and a lower renal resistive index (peak systolic velocity/(peak systolic velocity-diastolic velocity))

(0.42±0.02 vs. 0.52±0.0.9 sham, p<0.05). No significant differences were seen between groups in global oxygen

delivery, renal blood flow, renal oxygen delivery, renal cortical oxygen tension or renal oxygen extraction.

By 24 hours, global oxygen delivery, renal vascular resistance and resistive index were similar between septic

and sham animals, whereas renal oxygen delivery was lower (285±68 vs. 371±41mL/min, p<0.05) in the septic

animals. Renal oxygen extraction ratio was however similar (43±7 vs 43±9% sham, p=0.876) and this was

associated with a significantly lower renal cortical oxygen tension (10.8 ± 3.2 septic vs 14.9±4.2 mmHg sham,

p<0.05).

CONCLUSIONS

In early sepsis (6h), renal autoregulation (fall in renal vascular resistance) maintains renal oxygenation.

However, at 24h, despite a maintained global DO2, renal autoregulation may be impaired. This results in reduced

renal O2 delivery and renal cortical hypoxia.

GRANT ACKNOWLEDGMENT.

NA is supported by a Welcome trust training fellowship. MS is supported by the Dutch Kidney Foundation.

Page 40: Groningen International Program of Science in Medicine ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/... · (iii) Detection of macrophages 20 3. Enzyme-linked immunosorbent assay’s

40

“If we knew what it was we were doing, it would not be called research, would it?”

― Albert Einstein