identification of molecular and functional heterogeneity

80
Identification of Molecular and Functional Heterogeneity of Epithelial Progenitor Cells in the Upper Airway by Monica Allison Clifford A thesis submitted in conformity with the requirements for the degree of Master of Science Medical Biophysics University of Toronto © Copyright by Monica Clifford 2013

Upload: others

Post on 28-Jul-2022

8 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Identification of Molecular and Functional Heterogeneity

Identification of Molecular and Functional Heterogeneity of Epithelial Progenitor Cells in the

Upper Airway

by

Monica Allison Clifford

A thesis submitted in conformity with the requirements

for the degree of Master of Science

Medical Biophysics

University of Toronto

© Copyright by Monica Clifford 2013

Page 2: Identification of Molecular and Functional Heterogeneity

ii

Identification of Molecular and Functional Heterogeneity of Epithelial Progenitor Cells in the

Upper Airway

Monica Allison Clifford

Master of Science

Medical Biophysics University of Toronto

2013

Abstract

Upper airways are lined with a pseudostratified mucociliary epithelium

maintained by basal cells. To investigate functional and phenotypic heterogeneity

within the human basal cell compartment, we used a combination of limiting dilution

assays and surface marker profiling on primary cultures of basal cells with verified

progenitor activity. The limiting dilution assay suggested functional heterogeneity in the

ability of basal cells to repopulate a filter and maintain a barrier at ALI. The frequency of

cells with this activity varied between patient strains and ranged from 0.08%-1% of basal

cells. Validation of large-scale comprehensive surface marker profiling on basal cells led

to identification of 74 antigens demarking consistent subpopulations. Preliminary

functional analyses suggest differences in differentiation potential of some

subpopulations. This work supports the idea that the basal cell compartment may be

functionally heterogeneous, and provides new molecular tools for interrogation of

human basal cells.

Page 3: Identification of Molecular and Functional Heterogeneity

iii

Acknowledgments

It is a great pleasure to thank the many people who have made this thesis possible.

It is difficult to overstate my gratitude to my supervisor Dr. Nadeem Moghal. With his

enthusiasm and passion for science he inspired me and with his patience and knowledge he

taught me. Throughout my degree and thesis preparation he provided encouragement, sound

advice and lots of good ideas. I would have been lost without him.

I wish to thank my lab mates, Boram Kim and Emily van de Laar, and my many colleagues for

their support, guidance, assistance and companionship.

I am grateful to my committee Dr. Sean Egan and Dr. Norman Iscove for their wisdom and

guidance throughout my degree.

Finally, I wish to thank my family, friends and the many people who provided me with the

support I needed to succeed and who made this experience memorable.

Page 4: Identification of Molecular and Functional Heterogeneity

iv

Contents

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

Acknowledgments ........................................................................................................................... iii

List of Tables .................................................................................................................................... iv

List of Figures .................................................................................................................................. vii

List of Appendices.......................................................................................................................... viii

List of Abbreviations ........................................................................................................................ ix

1. Introduction ................................................................................................................................. 1

1.1 Structure of Normal Adult Airway ......................................................................................... 1

1.2 Classical and Non-Classical Stem Cell Theory ........................................................................ 3

1.3 Candidates for Lung Stem Cells ............................................................................................. 4

1.3.1 Progenitor Populations in the Distal Airways and Respiratory Region .......................... 5

1.3.2 Progenitor Populations in the Proximal Airway ............................................................. 7

1.4 Evidence for Heterogeneity within the Basal Cell Compartment ....................................... 10

2. Experimental Design and Methods ........................................................................................... 13

2.1 Isolation of Human Tracheal Epithelial Cells ....................................................................... 13

2.2 Growth and purification of HTECs by culturing ................................................................... 14

2.3 In vitro differentiation using ALI culture ............................................................................. 15

2.5 Antigen retrieval .................................................................................................................. 17

2.6 Antibody Staining ................................................................................................................ 17

2.7 Enface staining..................................................................................................................... 18

2.8 High- Throughput Antibody Screen ..................................................................................... 19

2.9 RNA Extraction and cDNA Synthesis .................................................................................... 19

2.10 Real-Time PCR ................................................................................................................... 19

2.11 Statistical Analysis of Limiting Dilution Analysis Results ................................................... 20

3. Results ....................................................................................................................................... 20

3.1 Early passage HTECs are p63-positive and retain regenerative properties ........................ 20

3.2 Ciliogenesis can be used to evaluate culture performance at ALI. ..................................... 24

3.3 Kinetic analysis of mucociliary differentiation of P0-plastic cells seeded at ALI. ................ 27

3.4 ALI cultures may maintain a progenitor population during differentiation. ....................... 33

3.5 Functional heterogeneity is suggested by limiting dilution analysis ................................... 36

Page 5: Identification of Molecular and Functional Heterogeneity

v

3.6 Culture-isolated basal cells have heterogeneous surface marker expression. ................... 40

3.7 Differences in isolated cell populations can be determined using ALI culture. .................. 42

4. Discussion .................................................................................................................................. 46

4.1 Establishment of culture systems and Assays ..................................................................... 48

4.2 Evidence for Functional Heterogeneity ............................................................................... 52

4.3 Evidence for Molecular Heterogeneity ............................................................................... 54

7. References ................................................................................................................................. 60

Appendices .................................................................................................................................... 64

Appendix A ................................................................................................................................ 64

Appendix B................................................................................................................................. 67

Page 6: Identification of Molecular and Functional Heterogeneity

vi

List of Tables

Table 1: LDA results are unable to support that differentiation at ALI is dependent on a

single factor.

Table 2: LDA results show frequency of a cell capable of repopulating an ALI culture.

Table 3: Frequency of a cell capable of repopulating an ALI culture is significantly

different between patient strains.

Page 7: Identification of Molecular and Functional Heterogeneity

vii

List of Figures

Figure 1. P0plastic HTECs are a purified population of basal cells

Figure 2. P0p cells retain the ability to differentiate in xenograft and ALI cultures (Str. 37

and 38)

Figure 3. P0p cells retain the ability to differentiate in ALI cultures (Str. 23)

Figure 4. Accurate evaluation of ciliogenesis is possible by enface staining and FOXJ1

expression but not by 2D ALI sections.

Figure 5. Qualitative assessment of differentiation in ALI cultures at multiple time points

by immunofluorescence.

Figure 6. Significant changes in gene expression occur during differentiation at ALI and

vary between patient samples.

Figure 7. Differentiation of basal cells at ALI can maintain a progenitor cell population.

Figure 8. Cell surface markers identify subpopulation of P0p basal cells.

Figure 9. P0p basal cells sorted for expression of CD54 resulted in morphologically

distinct ALI cultures.

Figure 10. P0p basal cells sorted for expression of podoplanin gave rise to

indistinguishable ALI cultures.

Page 8: Identification of Molecular and Functional Heterogeneity

viii

List of Appendices

Appendix A: List of subpopulations identified by HTS

Appendix B: Primer sequences

Page 9: Identification of Molecular and Functional Heterogeneity

ix

List of Abbreviations

ALI air liquid interface

BASC bronchioalveolar stem cell

BrdU 5-bromo-2-deoxyuridine

BSA bovine serum albumin

βTUBIV beta tubulin IV

C Celsius

CCA Clara Cell-specific marker

CD cluster of differentiation

cDNA complementary DNA

cm centimeter

CO2 Carbon dioxide

DAPI 4',6-diamidino-2-phenylindole

DASCs Distal airways stem cells

DMSO dimethyl sulfoxide

DNA Deoxyribonucleic acid

Np63 delta-N p63 isoform

EDTA Ethylenediaminetetraacetic acid

EGF epidermal growth factor

ELDA extreme limiting dilution analysis

EtOH ethanol

FACS Fluorescent activated cell sorting

FBS fetal bovine serum

H&E hematoxylin and eosin

Page 10: Identification of Molecular and Functional Heterogeneity

x

HCl Hydrochloric acid

HTEC Human Tracheal Epithelial Cells

HTS high-throughput screen

IF immunofluorescence

K14 keratin 14

K5 keratin 5

LHC-9 Laboratory of Human Carcinogenesis-9

MEM minimal essential media

ml milliliter

mm millimeter

MUC16 Mucin 16

MUC5ac Mucin 5ac

N normal

ng nanogram

nM nanomolar

NO2 nitric dioxide

NOD/SCID

non-obese diabetic/severe combined

immunodeficiency

P0p passage 0, plastic

P0pA passage 0 plastic to ALI

P0plastic passage 0, plastic

P1 passage 1

P2 passage 2

P2pA passage 2 plastic to ALI

P3 passage 3

Page 11: Identification of Molecular and Functional Heterogeneity

xi

p63 tumour protein 63

PAP plastic, ALI, plastic

PAPA plastic, ALI, plastic, ALI

PBS phosphate buffered saline

PSG penicillin streptomycin L-Glutamine

qPCR quantitative polymerase chain reaction

RA retinoic acid

RNA ribonucleic acid

SMG submucousal gland

SO2 Sulfur dioxide

SP-C prosurfactant apoprotein-C

str. strain

TAC transit amplifying cell

TAp63 transactivating p63 isoform

TASCs tracheal airway stem cells

TF Tissue factor

TNS trypsin neutralizing solution

µg microgram

UHN University Health Network

µm micrometer

Page 12: Identification of Molecular and Functional Heterogeneity

1

1. Introduction

1.1 Structure of Normal Adult Airway

The human lung is a complex organ that delivers oxygen to the circulatory

system. It is composed of a series of airways that lead ultimately to the respiratory

region where gas exchange occurs. To facilitate gas exchange the structure of the lung

provides an extensive interface between the environment and organism. This extensive

interaction highlights the airway epithelium’s essential barrier function and role in

defense against infectious and harmful inhaled agents [1-4]. Air entering the lungs

initially passes through proximal and conducting airways where it is conditioned prior to

entering the respiratory region. The numerous cell types found in lung epithelium

contribute to the function and maintenance of this barrier [1-4].

The proximal airways, comprised of trachea and main stem bronchi, are lined

with a pseudostratified columnar epithelium [5]. This epithelium contains three main

cell types: basal, secretory, and ciliated [2]. Ciliated cells are most prevalent, accounting

for more than 50% of the total cell number [3,6]. They are involved in tight junction

formation, and function to transport mucus from the lung to the throat by means of

ciliary beating [2,3]. Goblet cells are the predominant secretory cell in proximal airways

and account for the majority of mucin production [3]. They are also present at the

luminal surface and are involved in barrier maintenance through tight junction

formation [2]. Other secretory cells include serous cells and clara cells, though these are

present at a very low frequency [2]. A major difference between human and murine

Page 13: Identification of Molecular and Functional Heterogeneity

2

airways is the frequency and distribution of clara cells. In mice, clara cells are the

predominant secretory cell and are found throughout proximal and distal airways [7]. In

humans, clara cells are largely restricted to bronchioles [8]. Basal cells account for up to

30% of the total cell number in human trachea but decrease in number in bronchi and

bronchioles [2]. They are present at the basement membrane and serve as an

anchoring point for the epithelium by formation of hemidesmosomes [9,10]. Basal cells

are cuboidal non-columnar cells, distinguished by their expression of tumour protein 63

(p63), cytokeratins 5 and 14 (K5/K14), as well as the cell surface marker CD44 [10-12].

P63 exists in different isoforms in the cell. Emerging data has suggested that the

isoform lacking an N-terminal transactivating domain (Np63) regulates expression of

K5 and K14 in keratinocytes. It may therefore be essential for maintaining basal cell

identity [13]. The isoform possessing the transactivating domain (TAp63) is thought to

play a role in maturation and differentiation of keratinocytes and esophageal cells. It

may therefore also have a role in maturation of lung basal cells into other, differentiated

cell types [14]. Basal cells represent a progenitor cell compartment for proximal

airways, giving rise to secretory and ciliated cell lineages [4,15-18]. Neuroendocrine

cells and submucosal glands (SMG) are also found in this region of the airway [19,20].

Epithelial composition of the human distal airway is distinct from the

pseudostratified columnar epithelium of the proximal airway [21]. The bronchioles are

lined with a simple cuboidal epithelium composed mostly of ciliated and clara cells; they

are devoid of cartilaginous rings and SMGs [5]. Moving distally, bronchioles connect to

terminal bronchioles, which have a simple cuboidal epithelial structure composed

Page 14: Identification of Molecular and Functional Heterogeneity

3

mostly of clara cells with a large decline in the number of ciliated cells [5]. This is the

final region of conducting airways and leads into alveoli, where gas exchange occurs.

Alveoli are lined with two cell types: type I and type II alveolar cells [21]. Type I alveolar

cells are thin squamous cells through which gas exchange occurs [21]. Type II alveolar

cells are thought to be progenitor cells of alveoli with the ability to give rise to type I

alveolar cells [21]. Additionally, they secrete pulmonary surfactant proteins essential for

alveoli to overcome the surface tension of water and fill with air upon inhalation [21].

1.2 Classical and Non-Classical Stem Cell Theory

Highly proliferative tissues such as the intestinal epithelium or the hematopoietic

compartment of the bone marrow rely on a classical stem cell hierarchy to maintain

homeostasis [22,23]. In these tissues a stem cell undergoes asymmetric division,

resulting in self-renewal (i.e. production of another stem cell) and creation of a daughter

cell. The daughter cell is still relatively undifferentiated but proliferates much more

rapidly than the stem cell; these are referred to as progenitor cells, or transit amplifying

cells (TACs). TACs have a finite capacity for self-renewal and are responsible for the

generation of the differentiated cell types that compose the mature tissue, such as

columnar cells in the intestinal epithelium, or circulating blood [22,23]. In classical stem

cell hierarchies the stem cells cycle rarely and possess the capacity for life-long self-

renewal.

Lung epithelium is a relatively quiescent tissue compared to blood, or intestinal

epithelium [24]. The low turnover rate required for homeostasis has made it difficult for

Page 15: Identification of Molecular and Functional Heterogeneity

4

researchers to identify stem/ progenitor cells involved in long-term maintenance.

Therefore, most research has focused on characterizing proliferation that occurs in

response to injury. Several cell types have been identified as facultative progenitors

including: type II alveolar cells and clara cells [25,26]. The transit amplifying properties

of these relatively differentiated cells suggests that lung epithelium may have a non-

classical stem cell hierarchy similar to that found in other endoderm derived tissues

such as the liver [27]. Mature hepatocytes appear to be responsible for repair following

injury [27]. However, the liver also contains relatively undifferentiated oval cells, which

have been shown to act as facultative stem cells under some conditions [27]. Basal cells

in the proximal airway epithelium may play an analogous role, as they possess capacities

for regeneration as well as for differentiation into columnar cells. Such ‘non-classical’

stem cell hierarchies may represent an alternative means for homeostasis and repair, or

may be reflective of the difficulty of identifying rare stem cells in a relatively quiescent

tissue. The presence of facultative stem cells in lung epithelium does not preclude the

possibility of a classical stem cell hierarchy. Recent findings suggest that more primitive

progenitors, and possibly stem cells, do exist in the lung [28].

1.3 Candidates for Lung Stem Cells

Exposure of lung epithelium to damaging and infectious agents in the

environment necessitates repair. In a recent study, Kajstura et al. proposed the

existence of a primitive CD117+, KLF4+, NANOG+, OCT3/4+ and SOX2+ stem cell that

facilitates extensive repair, supporting a classical stem cell hierarchy in the lung [28].

Page 16: Identification of Molecular and Functional Heterogeneity

5

They showed that cells isolated from human lung tissue specimens, expanded in F12

media supplemented with 10% serum and sorted for CD117 were self-renewing,

clonogenic, and multipotent in vitro [28]. A cryogenic injury model was used to ablate

cell populations from regions of the mouse lung, after which, CD117+ cells isolated from

human tissue and expanded in vitro were transplanted into the region. These

transplanted cells regenerated human bronchioles, alveoli and pulmonary vessels in the

mouse lung. These cells showed multipotency and were serially transplantable, two key

features of stem cells [28]. These cells are found throughout the human lung, but are

present in higher numbers in the distal airway. The proposed CD117+ stem cell would

represent a novel population upstream of previously described regional progenitors.

The majority of current evidence suggests that repair processes call on regional

progenitor cells present in the airway epithelium. The study of airway maintenance and

repair, as well as the population of progenitor cells responsible for these processes,

relies on ex vivo cultures of human epithelial cells and in vivo mouse models. Studies in

these systems have led to an understanding of some of the hierarchical relationships

that exist; basal cells give rise to secretory as well as ciliated cells in the proximal airway;

clara cells give rise to ciliated cells in bronchioles; and type II alveolar cells give rise to

type I alveolar cells in respiratory regions [16,25,26].

1.3.1 Progenitor Populations in the Distal Airways and Respiratory Region

The mouse airway epithelium can be used to study lung injury repair. Several

mouse injury models based on inhalation of cytotoxic agents have been established. In

Page 17: Identification of Molecular and Functional Heterogeneity

6

the respiratory region, type I alveolar cells are selectively destroyed by inhalation of

hyperoxic agents such as ozone and nitrogen dioxide (NO2) or the chemotherapeutic

agent blyeomycin [29,30]. In a study of alveolar repair, Evans et al. labeled dividing

cells with tritiated thymidine following NO2 treatment [25]. They found that type II

alveolar cells took up label, but over a 14 day chase period there was an increase in the

proportion of labeled type I cells [25]. This suggests that type II alveolar cells serve as a

regional progenitor for type I cells. Although clara cells and type II alveolar cells

demonstrate some capacity for regeneration, they also have a more differentiated

phenotype, suggesting they may be more mature, or facultative progenitors.

A naphthalene induced injury model is also widely used. Administration of

naphthalene selectively ablates clara cells based on their expression of cytochrome

P450F2 [31]. Once inside the cell, naphthalene is converted to a cytotoxic epoxide by

P450, leading to cell death [31]. Studies of mouse bronchioles have revealed a rare

population of clara cells which don’t express cytochrome P450F2, and are thus resistant

to naphthalene injury [32,33]. Following treatment with naphthalene, these rare clara

cells may act as a progenitor population to regenerate bronchiolar epithelium [33].

However, in proximal airways, the contribution of these resistant clara cells is minimal.

The basal cells of the trachea and bronchi regenerate the clara cell compartment

following naphthalene injury [16].

A putative stem cell residing at the bronchioalveolar junction has been described

in distal airways of the mouse lung [34]. A cell that was double positive for the type II

Page 18: Identification of Molecular and Functional Heterogeneity

7

alveolar cell marker, prosurfactant apoprotein-C (SP-C) and Clara Cell-specific marker

(CCA) was found in a murine adenocarcinoma model as a population present only in

tumours [34,35]. Kim et al. defined this double-positive cell population as the

bronchioalveolar stem cell (BASC) [34,35]. BASCs are characterized as Sca1-positive,

CD34-positive, CD45-negative, Pecam-negative cells. When FACS-purified on the basis

of these markers, and expanded on a mouse fibroblast feeder layer, BASCs gave rise to

cells expressing clara cell, and type I alveolar cell markers in Matrigel [34]. These

findings suggest that BASCs have multipotential differentiation capacity and may

represent a rare population of stem/progenitor cells in distal airways. However, the in

vivo importance of BASCs in lung homeostasis has not supported by lineage-tracing

assay [26]. Indeed, by following the fate of lineage-labeled Scgb1a1 (CCA+) cells, they

found no evidence for a contribution of CCA-positive cells, including BASCs, to alveolar

homeostasis or repair [26]. Also, BASCs have not been found in humans.

1.3.2 Progenitor Populations in the Proximal Airway

The study of human tracheal epithelial cells has relied primarily on two ex vivo

systems; the rat tracheal xenograft model and the Air-Liquid Interface (ALI) culture

system [36-38]. In the xenograft assay, a rat trachea is decellularized by multiple rounds

of freeze-thaw. The trachea is then assembled into an open-ended cassette and

inserted subcutaneously on the back of an immunocompromised mouse [37]. Human

epithelial cells are injected into the lumen of the rat trachea and over the course of 5-6

weeks, these cells regenerate a fully differentiated epithelium which includes SMGs

Page 19: Identification of Molecular and Functional Heterogeneity

8

[37]. Alternatively, ALI culture is an in vitro system where cells are seeded onto the

surface of a porous Transwell membrane and grown under submerged conditions until

confluence is reached [36,39]. Once the cells have formed a confluent monolayer,

media from the upper chamber is removed and cells are fed basolaterally, leaving the

apical surface exposed to air [36,39]. The ALI system allows for differentiation into

ciliated cells, and secretory cells, and maintains cells in a basal position.

Using the xenograft model Englhardt et al. interrogated cultured primary human

tracheal/bronchial epithelial cells [40]. The cells were infected with retrovirus carrying

either ß-galactosidase or alkaline phosphatase at an efficiency of 5-10% [40]. Xenografts

where then established with between 0.5-1x106 cells. After several weeks, the resulting

epithelium contained labeled colonies of different sizes and of distinct composition [40].

Over 40% of labeled colonies contained basal, intermediate, ciliated and goblet cells.

However, the majority of colonies were composed of a subset of these cell types. For

example nearly 30% of colonies were composed of basal, intermediate and ciliated cells,

but lacked goblet cells, while less than 3% of colonies were composed of basal cells

alone. Interestingly, no colonies were found that lacked both basal and intermediate

cells. These data demonstrate that human tracheal epithelial cells, expanded transiently

in vitro as P0plastic cells, possess the potential to give rise to a new, fully differentiated

epithelium in vivo. Additionally, the varying clonal output from individual labeled cells

suggest heterogeneous lineage potential of cells in the initial population. However, it is

also possible that the initial population was homogeneous, with individual cells giving

rise to distinct clonal outcomes as a varying stochastic response to environmental cues.

Page 20: Identification of Molecular and Functional Heterogeneity

9

Basal cells of the trachea were speculated to be progenitors as early as 1963,

based on lack of discernible functional features and their location in the epithelium [11].

In 2007, Hajj et al. proved this by using CD151 and Tissue Factor (TF) surface markers to

purify human basal cells [41]. The CD151/TF double positive population was isolated

from a suspension of primary human airway epithelial cells by Fluorescence Activated

Cell Sorting (FACS). The ability of double positive cells to generate a well-differentiated,

functional epithelium relative to the marker negative fraction was assessed both by

xenograft assay and ALI culture. Only the CD151/TF-positive basal cell fraction

possessed the ability to proliferate and differentiate in these assays [41]. These results

show that basal cells can act as progenitors for the proximal airway.

Complimentary in vivo murine experiments using injury repair models also

support basal cells as being the main regenerative cell for proximal airways [16]. Using

SO2 to induce epithelial damage, Rock et al. studied repair by following the fate of

lineage-labeled basal cells [16]. A transgenic mouse was developed to follow the fate of

K5+ basal cells by combining K5-CreER and a Rosa26-lacZ reporter. Following tamoxifen

injections to activate Cre, the fate of labeled cells was tracked during homeostasis or

during repair from SO2 injury. K5+ basal cells gave rise to label containing ciliated and

clara cells, showing that basal cells function as progenitors for columnar cells during

homeostasis and repair [16].

Together these results show that the proximal airway epithelium has capacity for

extensive repair and regeneration, that this capacity is found within the basal cell

Page 21: Identification of Molecular and Functional Heterogeneity

10

compartment, and that basal cells regenerate non-basal cell types. These data further

suggest that there is heterogeneity of lineage potential of within the basal cell

compratment. However, while it is accepted that cells present in the basal

compartment act as progenitors, the degree of heterogeneity or hierarchical

organization within this compartment is unknown.

1.4 Evidence for Heterogeneity within the Basal Cell Compartment

Based on the understanding that basal cells house regenerative potential, the

distribution of this potential within the basal compartment has been the focus of a

number of studies. Heterogeneity within the basal cell population could be described

as: i) functional differences between basal cells, in terms of lineage potential or

proliferative capacity, ii) molecular differences between populations of basal cells, or iii)

anatomical differences in location, such as the residence of stem cells in specific niches.

Using murine adapted influenza H1N1 infection as a novel injury model, Kumar

et al. provided evidence for functionally distinct classes of p63+ basal cells [42]. Airway

cells were isolated from nasal, tracheal and distal airways of healthy human subjects.

Cells with in vitro clonogenic potential were found at frequencies ranging from 1:500 to

1:2000. Clonogenic cells were p63+ and K5+ basal cells, but given the frequency of p63+

K5+ basal cells is far greater than 1:500 in some airway segments, these data suggest

not all basal cells have the same ability to give rise to colonies in vitro [42]. Lineage

potential of these cells was also investigated using in vitro differentiation assays.

Clonogenic cells isolated from the tracheal airway, termed tracheal airway stem cells

Page 22: Identification of Molecular and Functional Heterogeneity

11

(TASCs), formed pseudostratified epithelia, and differentiated into goblet and ciliated

cells in ALI culture [42]. Clonogenic distal airway stem cells (DASCs) formed monolayer

epithelia and showed only limited induction of mucin expression and ciliated cell

formation in ALI culture [42]. These findings suggest that intrinsic differences exist

between regionally distinct p63+ progenitor cells, and that only rare cells in the basal

cell compartment (represents approximately 30% of all tracheal epithelial cells) possess

clonogenic potential. The presence of functional heterogeneity within the basal cell

compartment is further supported by distinct lineage composition of colonies derived

from single labeled basal cells during tracheal xenograft regeneration [43].

Potential molecular heterogeneity within the basal cell compartment has been

suggested based on differential cytokeratin expression. While expression of K5 and K14

are associated with basal cells in the airway epithelium [10], it has been reported that

these keratins may not be uniformly expressed in all basal cells [16,44-46]. A number of

studies in murine models have shown enrichment for K5 or K14 promoter activity in a

subset of basal cells in the upper trachea, suggesting there are subpopulations of basal

cells which have higher levels of cytokeratin expression [44-46]. An in vitro study by

Schoch et al. suggests basal cells expressing different levels of K5 could be functionally

distinct [44]. In this study, by using a transgenic mouse in which the K5 promoter drives

expression of enhanced green fluorescent protein (EGFP), they isolated high K5/EGFP-

expressing tracheal basal cells by FACS and evaluated their colony forming potential

under ALI conditions. Interestingly, K5 high expressing cells (EGFP bright) showed a 4.5-

Page 23: Identification of Molecular and Functional Heterogeneity

12

fold greater efficiency of colony formation and a 12-fold greater ability to form large

colonies relative to the K5 low-expressing basal cells [44].

The stem cell compartment in a number of tissues, including the intestinal

epithelium, exists in a specific anatomical niche [23]. There is some evidence that

stem/progenitor activity might be enriched in SMGs in the upper airway of mice [44-46].

Label retaining cells at the opening of SMGs have been described by Borthwick et al. in a

pulse chase experiment [46]. Using sulfur dioxide inhalation to injure the lung

epithelium, followed by intraperitoneal injections of BrdU to label cycling cells, they

observed an enrichment of label retaining cells (LRCs) in SMGs. LRCs are thought to

possess stem- or progenitor-like characteristics as they undergo a limited number of

divisions in response to injury, and then enter a quiescent state. However, the

relationship of these LRCs to basal cells is not known [46]. To investigate the

contributions of SMG basal cells to regeneration of damaged epithelia, Hegab et al.

developed a method to separate surface epithelial basal cells from SMG basal cells [45].

In vitro, the cells isolated from surface epithelium gave rise to a distinct colony

morphology in 3D cultures compared to SMG cells, suggesting that these two

populations possess inherent biological differences [45]. Furthermore, they performed

a K14 lineage trace showing that K14+ basal cells, which are enriched in SMGs, can

generate SMGs and surface epithelium during repair following severe ischemic injury

[45]. Taken together their results suggest that functionally distinct, K14+ SMG cells may

be multipotent progenitors capable of regenerating SMGs and surface epithelia [45].

Page 24: Identification of Molecular and Functional Heterogeneity

13

Current evidence supports p63+ basal cells being an important progenitor

population of the proximal, and possibly, distal airway epithelium. These cells have

critical roles in repair, and their study could lead to novel stem cell based therapies for

proximal airway diseases like cystic fibrosis. Furthermore, basal cells are thought to be

the cell of origin for squamous cell carcinomas (SCCs) of the lung as well as a subset of

adenocarcinomas. Given the important role of basal cells in health and disease states,

we seek to study molecular and functional heterogeneity within the basal cell

compartment of the healthy adult human airway epithelium. Such studies may help

uncover hierarchical organization within the basal cell compartment or may help to

define states associated with distinct basal cell behaviours. To perform this work we

use functionally verified populations of cultured, primary, unpassaged healthy human

airway basal cells (P0plastic cells). We predict that P0plastic cells contain identifiable

molecular and functional heterogeneity which is reflective of the in vivo basal cell

compartment.

2. Experimental Design and Methods

2.1 Isolation of Human Tracheal Epithelial Cells

Epithelial cells are isolated from healthy organ donor tracheas obtained as

surgical waste from lung transplants with informed patient consent from the Trillium

Gift of Life Network and Research Ethics Board approval (UHN). Tissue is stored in a

solution of Minimal Essential Media (MEM) with antibiotics and placed at 4°C until

retrieved for epithelial cell isolation. All steps of the dissociation are carried out on ice,

Page 25: Identification of Molecular and Functional Heterogeneity

14

or at 4°C. Initially, fatty tissue is removed and the trachea is rinsed in a solution of

MEM with antibiotics. It is then cut into 1-2 cm2 pieces and digested in a solution of

MEM with Protease type XIV (Sigma P5147), DNase (5 mg) containing L-Glutamine Pen-

Strep (PSG), Fungizone (Amphotericin B) and Gentamycin. Digestion is carried out at 4°C

for 24 hours; after 24 hours, tissue pieces are moved to fresh protease solution. Upon

collection, isolated human tracheal epithelial cells (HTECs) are counted using a

hemocytometer and either plated directly for use in experiments or suspended at 1x106

cells/ml in freezing media composed of LHC-9 with 10% fetal bovine serum (FBS) and

10% Dimethylsulfoxide (DMSO). Suspensions are frozen slowly overnight at -80°C then

transferred to a liquid nitrogen freezer for long-term storage.

2.2 Growth and purification of HTECs by culturing

Cell culture and propagation methods used for primary HTECs were first

established in 1981 by Lechner et al. [47]. Prior to initiation of culture, dishes are

prepared by coating with type 1 collagen (PureCol, Advanced BioMatrix). PureCol is

diluted to 48ug/ml in 0.01N hydrochloric acid (HCl); 10ml of this solution is added to a

10 cm dish and incubated at 37°C with 5% carbon dioxide for 2 hours. After incubation,

the solution is removed and plates are ready for use in HTEC culture. Initially, frozen

vials of HTECs are thawed into a collagen coated culture dish in 10 ml of LHC-9

supplemented with Amphotericin B; LHC-9 media is formulated for isolation and

expansion of p63-positive basal cells [47]. Media is changed 16 hours after thawing to

remove DMSO and FBS. Media is changed every other day and cells are maintained in

Page 26: Identification of Molecular and Functional Heterogeneity

15

culture for approximately 10 days or until 70% confluence is reached, at which point

cells are referred to as P0plastic cells and are used for subsequent experiments.

Subculturing of cells is performed by treating cultures with 0.025% trypsin/EDTA, trypsin

is then neutralized with Trypsin Neutralizing Solution (TNS) (ReagentPack, Subculturing

reagent kit, Lonza, CC-5034). Cells are typically replated at a density of 1500-3000

cells/cm and maintained in LHC-9 without Amphotericin.

2.3 In vitro differentiation using ALI culture

Growth and differentiation of P0plastic cells on porous transwell membranes is

performed as previously described [36]. Matsui et al. initially described ‘Matsui’ media,

used for expansion and differentiation of P0plastic cells at ALI [39]. Transwell

membranes are coated with 48ug/ml PureCol in 0.01N HCl for 2 hours at 37C. PureCol

is removed and 1.5 ml Matsui expansion media (0.33nM Retinoic acid (RA) and 5ng/ml

epidermal growth factor (EGF)) is added to the bottom chamber. Cells are suspended

at required concentration in Matsui expansion media to seed the desired number in a

0.5 ml aliquot in the upper chamber. Cells are grown at 37C and 5% CO2, media is

changed every other day. Once confluence is reached, media is removed from the

upper chamber and media in the lower chamber is switched to Matsui differentiation

media with high RA (50nM) and low EGF (0.5ng/ml). Cells are maintained in culture at

an air-liquid interface for a minimum of 3 weeks at which point they are evaluated for

differentiation.

2.4 Histology of ALI Cultures

Page 27: Identification of Molecular and Functional Heterogeneity

16

An agar embedding method described by the UHN Pathology Research Program

was adopted to facilitate cutting sections from paraffin embedded samples. At the

point of harvest, cultures are washed twice with PBS and 10% buffered formalin is

added above and below the membrane to a total volume of 1 ml. Fixation is carried out

at room temperature for 1 hour, formalin is removed, and the cultures are rinsed an

additional three times with PBS. Membranes supporting the fixed ALI culture are

removed from the Transwell insert using a scalpel to cut carefully around the edges

while keeping the membrane pressed flat against the cutting surface. The membrane is

placed tissue side down in an embedding mold containing molten agar (3% low gelling

temperature agar in distilled water) ensuring the membrane is well away from all edges

and completely covered with agar. The agar is allowed to set at room temperature. The

agar block containing a fixed culture is removed from the mold and excess agar is

trimmed so the block will fit into an embedding cassette, leaving at least 1 mm of agar

on all sides. Agar blocks can be stored in 70% ethanol until processed.

Once prepared, agar blocks can be processed like a regular block of tissue.

Embedding cassettes are loaded into a Shandon Excelsior tissue processor and

processed overnight. Briefly, this machine takes the blocks through stages of

dehydration in an alcohol series, alcohol is displaced with xylene, and xylene is

subsequently replaced with increasing concentration of paraffin in xylene until the

blocks contain only paraffin.

Page 28: Identification of Molecular and Functional Heterogeneity

17

Using a microtome, 6 µm sections are cut from paraffin embedded samples

perpendicular to the direction of the membrane in the block. Sections are floated on a

42C water bath to flatten and soften the paraffin, and then scooped onto pre-cleaned

charged glass slides. The slides with cut sections are placed in a 58C oven until dry and

the paraffin is melted onto the slide. This can be carried out overnight.

Prior to H&E or antibody staining, the paraffin must be cleared from the sample.

Paraffin is initially removed by soaking slides in xylene for 10 minutes; this step is

repeated with fresh xylene. The slides are then taken through an ethanol series of: 2x

100% for 10 minutes, 90% for 5 minutes, 70% for 3 minutes, 50% for 3 minutes, 35% for

3 minutes and PBS for 3 or more minutes.

2.5 Antigen retrieval

Antigen retrieval must be performed on formalin fixed samples prior to antibody

staining [48,49]. The optimal retrieval process depends on the antigen. We find heat

retrieval in citrate buffer is sufficient for our antigens of interest [48,49]. Deparaffinized

samples are placed in containers of 10mM citrate buffer pH 6 in an antigen retriever

(2100 Retriever). The assembly is brought to over 120C under pressure for at least 3

minutes. The cycle takes approximately 20 minutes to complete; the whole apparatus is

allowed to cool for 100 minutes before removing samples for a total run time of 2 hours.

2.6 Antibody Staining

Following antigen retrieval, cells are permeablized in PBS with 0.1% Triton X-100

for 5 minutes. Samples are then blocked in 3% bovine serum albumin (BSA)/ 0.1%

Page 29: Identification of Molecular and Functional Heterogeneity

18

Triton X-100 in PBS for 30 minutes and incubated with primary antibody diluted in BSA/

0.1% Triton X-100 in PBS for 1 hour at room temperature. Primary antibodies used

were: CD44 (IM7) cat# 17-0441-81, MUC16 (oc125) cat# sc-33344, Muc5ac (45M1) cat#

MS145-p1, p63 (BC4A4) cat# sc-56188, and βTUBIV (ONS.1A6) cat# T7941. After

incubation with primary, samples were washed 3 times with 0.1% Triton X-100 in PBS

and incubated with secondary antibody in BSA/ 0.1% Triton X-100 in PBS for 2 hours at

room temperature or overnight at 4°C. The secondary antibodies were alexafluor

(Invitrogen) 488 goat anti-rabbit (1:500), 555 Goat anti-rabbit (1:500) and 568 goat anti-

mouse (1:500). Samples are washed 3 times with 0.1% Triton X-100 in PBS. Tissue

sections and cytospun preparations were mounted in vectashield mounting media with

DAPI.

2.7 Enface staining

For a quantitative assessment of differentiation, enface staining technique was

established. ALI membranes are formalin fixed as described above. ALI membranes are

stained as described above; antigen retrieval is not required for βTUBIV staining. Upon

completion of staining membranes are washed three times in 0.1% Triton X-100 in PBS

then dehydrated through an alcohol series from 70% for approximately 1 minute, to

90% ETOH briefly and rinsed in 100% ETOH to remove any residual water from the

surface of the ALI. The membrane is carefully cut from the well insert with a sharp

scalpel while keeping the membrane pressed against the cutting surface. In a fume

hood, the membrane is placed in a drop of 30% Permount in xylene on a pre-cleaned

glass microscope slide, epithelial side up. More Permount-xylene solution is added on

Page 30: Identification of Molecular and Functional Heterogeneity

19

top of the membrane and a cover glass is carefully placed to avoid bubbles and flatten

the membrane, the slide is dried in a fume hood overnight. Imaging of enface staining

was performed using Metamorph® tiling utility.

2.8 High- Throughput Antibody Screen

Cells were expanded under submerged conditions to 70% confluence as

P0plastic. Cells were trypsinized with 0.0125% trypsin+EDTA (Clonetics), neutralized

with TNS, pelleted and resuspended in Hanks buffered salt solution with 1% FBS. Cells

were submitted to the antibody core facility. Cells were then stained with the panel of

antibodies in a 96-well plate format; marker expression was analyzed by flow cytometry.

2.9 RNA Extraction and cDNA Synthesis

Total RNA was extracted using Ambion Micro RNA kit, RNA isolate was treated

with DNase I to remove contaminating genomic DNA. A total of 1ug of RNA was

converted into cDNA in a 20ul reaction volume using the High Capacity cDNA Reverse

Transcriptase kit (ABS). An automated thermocycler was used to provide the following

conditions: 10 minutes at 25°C, 120 minutes at 37°C, 5 seconds at 85°C and 4°C until

transferred to -20°C for long-term storage.

2.10 Real-Time PCR

cDNA stock solution was prepared by diluting cDNA to a concentration of 2 ng/µl

in autoclaved miliQ water. The SYBR Green system (Biorad) was used for qPCR

reactions. 4 µl of diluted cDNA was added to a reaction mixture containing 5 µl of SYBR

green super mix (Biorad) and 1 µl of primer mix containing 3µM of forward and reverse

Page 31: Identification of Molecular and Functional Heterogeneity

20

primers (Appendix B). Reaction conditions for detection of amplification were 95°C for 3

minutes, 95°C for 10 seconds, and 60°C for 30 seconds for 40 cycles.

2.11 Statistical Analysis of Limiting Dilution Analysis Results

Stem/progenitor cell occurrence follows a Poisson distribution. Limiting dilution

analysis results are analyzed by a webtool: Extreme limiting dilution analysis [50]. The

program enables inclusion of densities that result in all failures or all successes. Results

are based on the assumption of a linear relationship between seeding density and

outcome.

3. Results

3.1 Early passage HTECs are p63-positive and retain regenerative properties

To begin to address the degree of molecular heterogeneity within the human basal cell

compartment, we used previously described methods to extract large airway progenitor

cells from adult human tracheal tissue [51-53]. Although our goal was to analyze basal

cells in as close to an in vivo state as possible, we found that the most effective means

of isolating these cells also cleaved many cell surface markers. To bypass this problem

and increase the yield of cells for high-throughput assays we expanded enzymatically

digested, P0 tracheal suspensions without passaging, in a serum free medium previously

described to maintain large airway progenitor activity [47]. After expansion, we

evaluated the purity of this P0-plastic population by immunofluorescent (IF) antibody

staining for basal and columnar cell markers. We used α-p63, α-βTUBIV, α-MUC16 and

Page 32: Identification of Molecular and Functional Heterogeneity

21

α-Muc5ac to assess the presence of basal cells, ciliated cells, mucinous cells and goblet

cells respectively. Staining showed approximately 90% of cells were positive for p63

(Fig. 1a). No cells had polarized apical βTUBIV staining, a hallmark feature of ciliated

cells, though a small percentage stained weakly for βTUBIV in cytoplasmic foci (Fig.

1b) [54,55]. No cells expressed MUC16 (fig. 1c) or Muc5ac (data not shown). To

assess if p63-negative nuclei represented contaminating cell types, we examined

another basal cell marker, CD44 by FACS analysis, showing that P0plastic cells are

~100% positive for CD44 (fig. 1e) [12]. Based on CD44 staining data, we re-

examined p63 expression by IF and found a number of nuclei initially scored

negative actually had low levels of p63. Together these data indicate that P0plastic

cells represent a close to pure population of basal cells, with a low amount of

heterogeneity in p63 expression.

To verify that proliferating P0plastic basal cells retained progenitor activity,

we seeded cells derived from different donor tracheas into rat tracheal xenograft

(Strain 37)[37] and ALI culture (Strain 38 and 23)[38] systems, which support the

differentiation of basal cells into columnar cells. These cells formed well-

differentiated epithelia in xenograft and ALI cultures showing they retained

functional in vivo progenitor properties (fig. 2a, b; fig 3a). The presence of ciliated,

secretory and basal cells was confirmed in ALI-culture sections stained for βTUBIV

(fig. 2c; fig. 3b), MUC16 (fig. 2d; fig. 3c), and p63/CD44 (fig. 2e; fig. 3d). Although

other studies have reported differentiation of basal cells into Muc5ac-positive goblet

cells under ALI culture conditions, we did not detect this lineage (fig. 2f; fig. 3e)[39].

Page 33: Identification of Molecular and Functional Heterogeneity

22

Instead, we found robust differentiation into MUC16-positive mucinous cells,

another abundant in vivo mucinous large airway lineage [56].

Figure 1. P0plastic HTECs are a purified population of basal cells. Cytospun

unpassaged, HTECs expanded in submerged condition on collagen coated plastic (P0p)

stain positively for (A) basal cell marker p63 (86.3 +1.2%), but do not express (B) tubIV

(0%) or (C) Muc16 (0%). (D) Percentage of cells expressing each marker. (E)

Expression of basal cell marker, CD44, was evaluated by FACS on P0p cells.

Page 34: Identification of Molecular and Functional Heterogeneity

23

Figure 2. P0p cells retain the ability to differentiate in xenograft and ALI cultures.

(A) P0p cells were seeded into denuded rat tracheas and implanted into NOD/SCID

mice. After 33 days, xenografts were fixed, sectioned and stained by H&E (Str. 37). (B-

F) P0p cells were grown at ALI cultures for 21 days, after which filters were fixed,

sectioned and stained with H&E and indicated antibodies (Str. 38). All scale bars

represent 50 m.

Page 35: Identification of Molecular and Functional Heterogeneity

24

Figure 3. P0p cells retain the ability to differentiate in ALI cultures (Str.23). (A-E)

P0p cells were maintained at ALI for 21 days, after which filters were fixed, sectioned

and stained with H&E and indicated antibodies (Str. 38). All scale bars represent 50 m.

3.2 Ciliogenesis can be used to evaluate culture performance at ALI.

To quantify p63-positive progenitor activity in ex vivo cultures we focused on the

ciliated cell lineage, which is a major lineage derived from p63-positive progenitors [16].

Three methods to evaluate the extent of ciliogenesis were explored: 2D sections, enface

staining and gene expression analysis.

Intracellular localization of βTUBIV at the apical surface is a definitive marker for

mature ciliated cells [54,55]. However, the distribution of βTUBIV-positive cells is not

uniform in differentiated ALI cultures (fig. 4a). This can be seen in sections from the

Page 36: Identification of Molecular and Functional Heterogeneity

25

same membrane where in some sections ciliated cells are sparsely distributed (fig. 4a),

while in others they are densely distributed (fig. 4b). In contrast, basal cells, as marked

by p63, appear uniformly distributed as assessed by random sections of the same

membrane (fig. 4c and d). The impact of uniformity of cellular distribution on

estimating the mean number of cells is demonstrated by comparing standard deviations

in the percentages of ciliated cells and basal cells (fig. 4e). The percentage of total cells

having undergone ciliogenesis was 10.4 ±5.8%, compared to 25.4 ±2.7% for evenly

distributed basal cells. Thus, without extensive sectioning, random sections do not

readily allow for accurate extrapolation of the extent of differentiation for an entire

membrane, making it difficult to quantify differences in ciliogenesis between samples.

To circumvent this problem, we sought to establish a quantitative method to assess the

entire membrane for ciliated cells.

An enface staining technique was adopted as a non-biased method of

determining the extent of ciliogenesis. The membrane is stained for βTUBIV and images

of the entire membrane are captured and stitched together using Metamorph® tiling

software (fig. 4f). Software analysis of the enface image provides either: (1) a count of

ciliated cells, which is useful under conditions where limited differentiation occurs; or

(2) a percentage of reconstituted epithelium that is covered by cilia. In theory, this

approach accurately evaluates the extent of ciliated cell differentiation across the entire

culture, and should allow for quantitative comparison between cultures.

Page 37: Identification of Molecular and Functional Heterogeneity

26

Figure 4: Accurate evaluation of ciliogenesis is possible by enface staining and

FoxJ1 expression, but not by 2D ALI sections. (A,B) 2D sections stained for tubIV show irregular distribution of ciliated cells, in contrast, (C,D) p63-positive cells are evenly distributed. (E)The variability in percentage and frequency of positive cells as calculated

based on staining of 2D sections gives low confidence in values for tubIV when compared to normally distributed p63. (F) Enface staining allows quantitative determination of total ciliogenesis. (G) Analysis of gene expression provides a quantitative method to compare induction of FoxJ1 between cultures, physiologic controls (tracheal cells) and baseline controls (P0plastic basal cells). Scale bars in IF

images: 50 m; enface image: 1 mm.

Page 38: Identification of Molecular and Functional Heterogeneity

27

In addition to βTUBIV staining, we also evaluated FOXJ1 gene expression in

differentiated cultures (fig. 4g). FOXJ1 is a transcription factor involved in ciliogenesis in

various tissues; in the airway epithelium, its expression is restricted to ciliated cells [57].

Gene expression analysis by qPCR has the advantage of being very sensitive and, like

enface staining, uses the entire population of cells from the culture. qPCR analysis does

not permit determination of ciliated cell number, but we have found that induction of

FOXJ1 gene expression tracks with emergence of ciliated cells (fig 6g-l, and 7c). Gene

expression analysis allows for direct quantitative comparison between samples, as well

as comparison to physiological (tracheal cells) and base line control cells (P0plastic,

starting population). Our qPCR analysis of FOXJ1 in ALI cultures revealed that P0plastic

basal cells retained a robust ability to induce FOXJ1 to in vivo, tracheal levels (fig. 4g).

3.3 Kinetic analysis of mucociliary differentiation of P0-plastic cells seeded at

ALI.

In order to characterize the differentiation potential of putative subpopulations

of basal cells, we analyzed the kinetics of mucociliary differentiation from bulk P0-plastic

cultures at ALI. P0plastic progenitors isolated from three different donor tracheas were

grown at ALI, and cultures evaluated for differentiation markers at six stages of

repopulation and differentiation. The stages examined were: proliferating subconfluent

cultures, covering 50-70% of filter, cultures just achieving confluence, which is the point

when media was removed from the upper chamber and cells were exposed to air, 3

days post-confluence, 7 days post-confluence, 14 days post-confluence, and 21 days

Page 39: Identification of Molecular and Functional Heterogeneity

28

post-confluence. Cultures were evaluated by IF staining on sections, gene expression

analysis, and enface staining.

As p63 progenitors differentiate, we expect to see a shift from a population of

basal cells to a heterogeneous culture containing mixed lineages. Thus, we first

examined changes in the proportion of basal cells by immunostaining sections for p63.

We found that at subconfluence and at confluence nearly all nuclei are p63-positive (fig

5a, b). As early as 3 days post-confluence, a p63-negative layer of nuclei was seen

above the basally located p63-positive nuclei, consistent with pseudostratification (fig.

5c). This staining pattern of basally located p63-positive nuclei and apical p63-negative

nuclei persisted until the cultures reached maturity at 21 days post-confluence (fig. 5d,

e, and f). To investigate the contributions of TAp63 and Np63 to immunostaining and

to quantify p63 expression, we looked at gene expression levels of both p63 isoforms.

We saw the expected downward trend in expression of Np63; however, in one patient

strain there was an initial increase leading up to confluence, prior to its eventual decline

(fig. 6a). In contrast, TAp63 initially increased in all patient strains, and declined

following confluence in 2 of 3 patient strains (fig. 6b). All progenitor strains showed the

expected decline in Np63 expression that would occur during differentiation.

However, our data also suggest there may be some strain-dependent differences in

regulation of p63 isoforms during differentiation at ALI. The significance of these

differences is not known.

Page 40: Identification of Molecular and Functional Heterogeneity

29

Page 41: Identification of Molecular and Functional Heterogeneity

30

Figure 5: Qualitative assessment of differentiation in ALI culture at multiple time

points by immunofluorescence. (A-F) p63, (G-L) tubIV, and (M-R) Muc16 expression is assessed at multiple time points during repopulation and differentiation of ALI cultures by P0p cells. Expression of p63 in: (A) subconfluent cultures, (B) at confluence, and (C-F) in post confluent cultures, p63 staining is seen only in basally located cells. (G) Some

cells in subconfluent cultures have cytoplasmic tubIV foci, (H-I) these are not observed at confluence or 3 days post-confluence, (J) appearance of pre-ciliated cells is seen at 7 days post-confluence, and (K-L) mature ciliated cells are present at 14, and 21 days post-confluence. (M-O) Muc16 is not seen until 3 days post confluence, (P-R) and is

maintained until 21 days post-confluence. Scale bars: 25 m.

Polymerization of βTUBIV into apically projecting microtubules of cilia is a

definitive marker of the airway ciliated cell lineage [54,55,58]. In sections of

subconfluent, confluent, and 3 day post-confluence ALI cultures, we observed some

cytoplasmic βTUBIV staining, but no apical polymerization (fig. 5g, h, and i). At 7 days

post-confluence, intracellular accumulation of high levels of βTUBIV was seen in some

cells, presumably those undergoing ciliogenesis (fig. 5j). However, only at 14- days and

21-days post-confluence did apical localization of βTUBIV appear, indicating mature

ciliated cells (fig. 5k, l).

Similar kinetics for ciliogenesis were observed by following FOXJ1 expression by

qPCR. FOXJ1 expression was first detectable at 3-7 days post-confluence (fig. 6c), and

reached over 700% of physiological levels in two of three patient samples by 21 days

post-confluence (Fig 6c). Although the extent of induction varied substantially between

patient strains, the kinetics of ciliogenesis were similar. Enface staining for βTUBIV also

showed kinetics similar to the emergence of FOXJ1 which is consistent with FOXJ1’s late

role in ciliogenesis [59]. By enface staining, 3-day post-confluent cultures also showed

intracellular βTUBIV staining, but no mature ciliated cells (data not shown). At 7 days

Page 42: Identification of Molecular and Functional Heterogeneity

31

post-confluence, a limited number of mature ciliated cells were apparent, and by 14

days and 21 days post-confluence, cultures had large numbers of ciliated cells (fig. 7e-g).

Monitoring culture progression by light microscopy, we consistently observed

mucous-like secretions on the surface of differentiating cultures prior to detecting any

sign of ciliogenesis. To examine for emergence of mucinous lineages, we followed

expression of MUC16, which in vivo, marks abundant surface and glandular tracheal

cells [56]. In subconfluent cultures, and when confluence was first achieved, we saw no

reactivity with an α-MUC16 antibody (fig. 5m, n). However, as early as 3 days post-

confluence, we observed cell-specific staining at the apical surface of the epithelium (fig.

5o), which became more intense and more wide-spread by 1 week post confluence (fig.

5p). After this period it became difficult to determine which cells were unambiguously

MUC16 positive, possibly due to the antigen being clipped and spread across the

epithelial surface (fig. 5q, r). By qPCR, MUC16 gene expression was first detectable at

confluence, but at less than 1% of physiological levels (fig. 6d). At 3 days post-

confluence, expression in two of three patient samples reached over 10% of physiologic

levels, while in the third strain expression had reached less than 2% of these levels. In

subsequent weeks there was strain to strain variability in gene expression. In Str.

30277, MUC16 expression peaked at 7 days, reaching ~80% of physiologic levels but

then declined (fig. 6d). In Str. 78297, MUC16 levels rose to over 200% of physiological

expression by day 21 (fig. 6d). In Str. 39, MUC16 expression plateaued on day 14, at

25% of physiologic levels (fig. 6d). Our data indicate that for quantification of mucinous

cell numbers by antibody staining, cultures between 3 and 7 days post confluence are

Page 43: Identification of Molecular and Functional Heterogeneity

32

optimal. However, for greater sensitivity, qPCR on bulk populations of cultures older

than 7 days post confluence is better suited.

Page 44: Identification of Molecular and Functional Heterogeneity

33

Figure 6: Significant changes in gene expression occur during differentiation at

ALI and vary between patient samples. Gene expression changes in (A) Np63, (B)

TAp63, (C) FOXJ1 and (D) MUC16 were monitored during repopulation and

differentiation of P0p cells in ALI cultures. Enface staining for TUBIV showing (E) a

limited number of ciliated cells at 7 days post-confluence, (F, G) but substantial ciliated

cell coverage at both 14 and 21 days post-confluence.

3.4 ALI cultures may maintain a progenitor population during differentiation.

Serial passaging of HTECs on 2D plastic, under submerged culture conditions has

been reported to result in attenuation of basal cell progenitor capacity as assessed in ALI

culture [38]. Gray et al. demonstrated that NHTBE cells grown to a P3 plastic stage

retain the ability to functionally differentiate at ALI into an epithelium possessing many

properties of the in vivo epithelium including mucociliary differentiation and tight

junction formation [38]. For large scale or long-term studies it would be of great

interest to preserve progenitor activity ex vivo. The ALI culture system recapitulates an

in vivo environment by creating an air-liquid interface where cells are fed basolaterally

and exposed to air at the apical surface. This environment, combined with factors in the

media, leads to mucociliary differentiation of p63-positive basal cells, and formation of a

pseudostratified epithelial layer. It is unknown if this in vivo-like environment is better

able to maintain stem/progenitor cells than 2D-culture. Alternatively, differentiation

signals could push the stem/ progenitor cell population toward a terminally

differentiated fate, where basal cells seen in ALI cultures serve only a structural role in

epithelial anchoring.

Page 45: Identification of Molecular and Functional Heterogeneity

34

Page 46: Identification of Molecular and Functional Heterogeneity

35

Figure 7. Differentiation of basal cells at ALI can maintain a progenitor

population. (A) P0plastic cells were passaged on either ALI or plastic, re-equilibrated

on plastic and their ability to differentiate at ALI was re-assessed; arrows indicated

points where samples were collected. Differentiated cultures grown from either plastic-

passaged (P2pA) or ALI-passaged (PAPA) cells were assessed for expression of: (B)

FoxJ1, (C) Muc16, (D) Np63 and (E) TAp63. Enface staining for tubIV show extent of

ciliogenesis in (F) P2pA, plastic-passaged and (G) PAPA, ALI-passaged cultures.

To address this question, we compared progenitor activity of ALI-passaged and

plastic-passaged cells relative to the P0plastic progenitor population. Initially, after

expansion, P0p cells were seeded in replicate at ALI or passaged on plastic as P1 cells

(fig. 7a). After a well differentiated epithelium was generated at ALI (3 weeks), or P1

plastic cells reached 70% confluence, CD44+ basal cells were FACS purified from the

cultures (fig. 7a). To control for potential changes in progenitor activity due to culturing

in the different media associated with ALI vs. plastic culture we re-equilibrated both ALI

and plastic sorted cells in LHC-9 media on 2D plastic before seeding the P3 cells into ALI

for final analysis (fig. 7a). Both PAP and P2p cells proliferated in ALI culture wells at

comparable rates to form a confluent monolayer. After establishment of the ALI, both

populations maintained a barrier.

We next examined ciliated cell lineage potential of PAP and P2p progenitor cells.

In general, passaging under both conditions reduced ciliogenic differentiation. FOXJ1

induction was significantly reduced under both conditions in all three stains relative to

P0pA (fig. 7b). However, in Str. 30277, P2p basal cells had significantly larger induction

of FOXJ1 gene expression at ALI. These data indicate the ability to give rise to ciliated

cells is significantly impacted by prolonged culture under both conditions (fig. 7b). A

Page 47: Identification of Molecular and Functional Heterogeneity

36

dramatic decline in extent of ciliogenesis from P0plastic to P2plastic cells was seen by

enface staining (fig. 7f and g); given the deterioration of P2pA cultures in 2 of 3 patient

strains, we were unable to attribute failure of PAPA cultures in these 2 strains to their

previous differentiation at ALI. In Str. 39 we observed ciliary differentiation of PAPA

cultures at levels comparable to P2pA cultures (fig. 7g, h). MUC16 was induced at levels

similar to P0pA under both treatment conditions in all strains, suggesting that MUC16

lineage potential is maintained during the culture period examined (fig. 7c). Evaluation

ofNp63 and TAp63 showed no consistent difference following treatment conditions

(fig. 7d and e). These results demonstrate that both plastic and ALI culture can maintain

MUC16 competent lineage potential. However ALI is not better than plastic for

maintaining ciliogenic potential, and may be worse.

3.5 Functional heterogeneity is suggested by limiting dilution analysis

While some work supports the concept of functional heterogeneity within the

basal cell compartment, definitive evidence for a hierarchical organization is still lacking

[28,40,45]. To examine potential functional heterogeneity in human basal cells, we

attempted to quantify the frequency of ALI repopulating cells in limiting dilution assays.

We hypothesized that cells which could: proliferate, form tight junctions, and

differentiate on ALI, would have properties equivalent to in vivo progenitors. P0plastic

cells were seeded on ALI filters at seeding densities ranging from 10 000 to less than 100

cells per membrane. Cultures were assessed on their ability to: form a barrier, to

Page 48: Identification of Molecular and Functional Heterogeneity

37

maintain a barrier once exposed to air, and to differentiate as assessed by the presence

of beating cilia. We employed the ‘Extreme Limiting Dilution Analysis’ (ELDA) webtool

(http://bioinf.wehi.edu.au/software/elda/) to determine the frequency of a cell that

met these criteria [50].

In order for ELDA to provide an accurate estimate of frequency, it requires a

mixed outcome at a single seeding density on replicate filters. Some filters must show

success, while others show failure [50]. Furthermore, ELDA analysis assumes a single-hit

model, in which a single cell, the stem cell, is the only factor that affects success of the

culture; therefore the single hit hypothesis must be met before frequency of a

stem/progenitor cell can be estimated [50]. If additional factors or parameters that

affect the success of a culture can also be diluted, ELDA cannot be used to quantify the

frequency of a single progenitor population [50].

We initially used presence of functional cilia, directly assessed by light

microscopy, as the output for success and the indicator of presence of ALI-repopulating

cells. However, in experiments using basal cells derived from 11 donor tracheas, none

provided sufficient data to test the single-hit criteria (table 1) and allow estimation of

the frequency of ciliogenic progenitors. In no case did we observe a mixed response of

cultures successfully differentiating and failing to differentiate at a single seeding

density (Appendix C). Although there was strain to strain variability, we did determine

that in all cases, a seeding density of 6 000 cells/ filter was sufficient for success at ALI

(table 1). When success was re-defined to include those cultures that only reached

Page 49: Identification of Molecular and Functional Heterogeneity

38

confluence and formed tight junctions, but did not undergo differentiation, 5 patient

strains gave sufficient data to test the single hit hypothesis. In all of these patient

strains, the single hit criterion was met. The estimated frequency of a repopulating cell

ranged from as high as 1 in 96 in one patient strain, to as low as 1 in 1297 in another

(Table 2), suggesting human basal cells are functionally heterogeneous in their ability to

repopulate an ALI filter (Table 3).

Table 1: LDA results are unable to support that differentiation at ALI is

dependent on a single factor. P0plastic cells were seeded in replicate filters at

multiple dilutions and grown to confluence while being fed apically and

basolaterally. If confluence was reached, cultures were exposed to air apically

and fed basolaterally until differentiation was observed or culture failed to

differentiate. Those cultures which underwent mucociliary differentiation were

scored as successes and ELDA [50] was used to determine frequency of cell that

leads to success. No experiment satisfied the criteria of the statistical test and

frequency could not be estimated. Experimental data is available in Appendix C.

Lowest density at which differentiation occurred is presented.

Patient Sample

Reject single-hit hypothesis (p<0.05)

Lowest successful seeding density

18 Insufficient data 10 94214 Insufficient data 100 78297 Insufficient data 250 79168 Insufficient data 500 39 Insufficient data 500 23 Insufficient data 1000 11111 Insufficient data 1000 38 Insufficient data 1500 29945 Insufficient data 2000 32 Insufficient data 6000 55 Insufficient data 6000

Page 50: Identification of Molecular and Functional Heterogeneity

39

Table 2: LDA results show frequency of a cell capable of repopulating an

ALI culture. P0plastic cells were seeded in replicate filters at multiple dilutions

and grown to confluence while being fed apically and basolaterally. If confluence

was reached, cultures were exposed to air apically and fed basolaterally until

differentiation was observed or culture failed to differentiate. Those cultures

which reached confluence and/or underwent differentiation were scored as

successes and ELDA [50] was used to determine frequency of cell that leads to

success. Lower and Upper bounds represent 95% confidence intervals.

Patient Sample

Lower Estimated frequency

Upper Reject single-hit hypothesis (p<0.05)

78297 511 164 53 No (0.483) 79168 203 96 45 No (0.906) 29945 1030 519 261 No (0.267) 32 398 182 83 No (0.823) 55 3441 1297 489 No (0.297)

Table 3: Frequency of a cell capable of repopulating an ALI culture is

significantly different between patient strains. Pairwise comparison of

estimated repopulating cell frequency between patient samples was performed

by Pearson Chi square test using ELDA tool [50].

Patient Sample 1

Patient Sample 2

P-value Significance

29945 32 0.0502 29945 55 0.152 29945 78297 0.0966 29945 79168 0.00249 * 32 55 0.00228 * 32 78297 0.884 32 79168 0.25 55 78297 0.00837 * 55 79168 8.56e-05 * 78297 79168 0.461 *

Page 51: Identification of Molecular and Functional Heterogeneity

40

3.6 Culture-isolated basal cells have heterogeneous surface marker

expression.

The functional heterogeneity in basal cells observed here and in other work,

suggests there might be identifiable molecular heterogeneity within basal cells

[28,40,44-46]. To investigate the degree of molecular heterogeneity in human basal

cells, we stained tracheal basal cells with a panel of 339 antibodies raised against cell

surface markers. We chose a P0plastic population for this analysis for several reasons.

First, efficient isolation of lung epithelial cells required enzymatic digestion that clipped

off many surface proteins, potentially compromising our ability to detect some

subpopulations. Second, from the amount of donor tissue typically received, insufficient

cell numbers were obtained for such a screen without expansion. Third, one round of

expansion in LHC-9 media had the additional benefit of helping to remove columnar

cells as well as contaminating blood and stromal cells. To minimize potential artifactual

surface marker expression due to prolonged cell culture, we limited our analysis to

unpassaged P0plastic cell populations with verified progenitor activity (fig. 2 and 3).

High-throughput screens (HTSs) were carried out on two different patient

strains. Any distinctly shifted population that was reproduced across both patient

strains was considered a hit (fig. 8a-c). If the regenerative potential of lung epithelium

follows a classical stem cell hierarchy, as observed in blood, rare cells may represent the

stem or progenitor cell. These P0p populations could be identified as rare marker-

positive or rare-marker negative cells. Alternatively, if a non-classical stem cell model is

followed, in which a large number of facultative stem/progenitor cells are

Page 52: Identification of Molecular and Functional Heterogeneity

41

Figure 8: Cell surface markers identify subpopulations of P0plastic basal cells.

(A) Markers represent percentage of marker-positive cells in Str. 38, plotted against

percentage marker-positive in Str. 23 for a given cell-surface protein. Insets show (B)

small subpopulation, present on less than 2% or (C) those on greater than 98% of cells.

(D) A summary of all markers, categorized based on highest level of expression in any of

the three screens into: 0-1.9%, 2-9.9%, 10-89.9%, 90-97.9%, 98-100% (Total), the

number selected for further screening based on HTS (HTS), and the number confirmed

by single tube staining (validated) are shown (for list of markers selected based on HTS

see Appendix A).

Page 53: Identification of Molecular and Functional Heterogeneity

42

present in the tissue, we might expect to see more prevalent populations of cells

representing progenitors. These cells may also exist as marker-positive or marker-

negative subpopulations.

From the HTS, 112 markers were heterogeneously detected in both patient

strains of basal cells (fig. 8a-c, appendix A). These markers were examined in a third

patient strain of basal cells by FACs staining in a single tube format. Single tube staining

confirmed subpopulations denoted by 74 of these 112 markers; 35 markers were not

detectable, and 3 markers were detected on 100% of cells. Of the 74 markers of

heterogeneity that validated, 28 marked rare subpopulations consistently representing

fewer than 2% of the bulk population (fig. 8b, c). An additional 20 markers were present

on 2- 10% of the bulk population (fig. 8d). The remaining 26 markers were detected on

a relatively high percentage of cells, demarking more than 10% of the bulk population

(fig. 8d). Although the hits were similar between the two strains examined by HTS,

there was large variability in the percentage of cells expressing these markers between

patient samples (Appendix A). Single tube FACs analysis consistently identified smaller

populations (Appendix A), which might be due to technical differences in staining

between HTS and single tube format.

3.7 Differences in isolated cell populations can be determined using ALI

culture.

While comprehensive functional evaluation of all reproducible subpopulations

would be beyond the scope of this work, we chose a few candidate subpopulations for

functional studies. Bulk population cells were FACS sorted based on expression of CD54

Page 54: Identification of Molecular and Functional Heterogeneity

43

(fig. 9a, 5.4%) or podoplanin (fig. 10a, 85%), and seeded in replicate at ALI (10 000 cells

per well).

Monitoring progress of CD54-positive and -negative cultures revealed a marked

difference in epithelial morphology arising immediately after confluence and persisting

until 3 weeks post confluence when cultures had fully differentiated (fig. 9b-d). CD54-

positive cultures resembled control cultures, with many gland-like structures. In

contrast, CD54-negative cells formed very smooth cultures, with few gland-like

structures. During the three week period following establishment of ALI, CD54-positive

cultures appeared to slough a significant amount of debris or cells. This process was not

observed in CD54-negative cultures. These differences did not impact the extent of

ciliogenesis by enface staining (fig. 9e-g). Although gene expression analysis for

induction of FOXJ1 showed high variability between replicate cultures of each

subpopulation, no significant difference between CD54-positive and CD54-negative

cultures and unsorted controls were detected (fig. 9h). Evaluation of MUC16 induction

also showed no reproducible difference between populations (fig. 9i).

Cells sorted for the expression of podoplanin (fig. 10a) and grown at ALI showed

no differences during culture maturation at ALI. Enface staining revealed similar levels

of ciliogenesis in podoplanin-positive, podoplanin-negative and unsorted cultures (fig.

10b-d). Similar to our analysis with CD54 subpopulations, there were large differences

in FOXJ1 expression between replicate cultures, but overall, no reproducible difference

Page 55: Identification of Molecular and Functional Heterogeneity

44

Figure 9: P0p basal cells sorted for expression of CD54 resulted in morphologically distinct cultures. (A) FACS plot showing gating used to sort P0plastic cells based on podoplanin expression. Phase contrast microscopy showing morphology of: (B) CD54-positive, (C) CD54-negative, and (D) unsorted ALI cultures at 3

weeks post-confluence (scale bars: 50 m). Enface staining of: (E) CD54-positive, (F) CD54-negative, and (G) unsorted ALI cultures at 21 days post-confluence (scale bars: 1 mm). QPCR analysis showing (H) FoxJ1 and (I) Muc16 gene expression between CD54-positive and CD54-negative cultures.

Page 56: Identification of Molecular and Functional Heterogeneity

45

Figure 10: P0p basal cells sorted for expression of podoplanin gave rise to

indistinguishable cultures. (A) FACS plot showing gating used to sort P0plastic cells

based on podoplanin expression. Enface staining of: (B) podoplanin-positive, (C)

podoplanin-negative, and (D) unsorted ALI cultures at 21 days post-confluence (scale

bars: 1 mm). QPCR analysis showing (E) FoxJ1 and (F) Muc16 gene expression

between podoplanin-positive and podoplanin-negative cultures.

Page 57: Identification of Molecular and Functional Heterogeneity

46

between podoplanin-positive and –negative subpopulations (fig. 10e). Similarly, no

significant differences were observed in MUC16 induction between the podoplanin-

positive and podoplanin-negative subpopulations (fig. 10f).

Rare populations of cells are of great interest, as they may represent stem or

progenitor cells. However, these populations pose a challenge for study due to their

limiting number. To bypass this issue, we adopted the strategy of depleting the bulk

population of such rare cell populations. If the depleted cells represent an essential

population it is expected that cultures would perform poorly even at high seeding

densities. Depletion of populations expressing: CD116, CD117, CD127 or CD337 showed

no noticeable decline in performance (data not shown). These data suggest that such

subpopulations do not represent essential populations for success at ALI, or that the

culture conditions employed here do not require such populations.

4. Discussion

Under steady state conditions the airway epithelium is relatively quiescent.

However, it is regularly exposed to harmful environmental agents and pathogens, and is

the site of a number of prevalent diseases such as lung cancer, and cystic fibrosis.

Hence, a mechanism is required for normal, long term maintenance that can also

respond rapidly to injury and disease conditions. Presumably, the regenerative process

responsible for maintaining epithelial integrity involves a local stem cell population. This

population could follow a classical stem cell hierarchy as seen in several highly

proliferative tissues including the intestinal epithelium and blood, where a rare stem cell

Page 58: Identification of Molecular and Functional Heterogeneity

47

gives rise to progeny cells [22]. Alternatively, the lung epithelium could follow a non-

classical stem cell hierarchy as proposed in the liver [27,60]; in this model a large

number of equipotent, facultative stem cells would act in steady state and following

injury to repair or maintain the epithelium. Arguments can be made for either model in

the lung epithelium; a classical stem cell model might provide for long term

regenerative potential to maintain a tissue over the lifespan of the organism. A non-

classical model could be sufficient for long term maintenance of a tissue with a low

turnover rate, while providing a large number of progenitor cells capable of responding

quickly to acute injury. No consensus has been reached as current evidence supports

either model existing in the lung.

Basal cells are known to possess the regenerative capacity of the tracheal

epithelium [16,40,41], but also serve to anchor the entire epithelium to the basement

membrane through their unique formation of hemidesmosomes [9,10]. In light of the

multiple known functions for basal cells, in either a classical or non-classical model, we

might expect to find heterogeneity amongst basal cells at varying stages of maturity,

lineage commitment, or functional commitment. In any case, basal cell subpopulations

could be molecularly and functionally distinct.

In this work I explored cellular heterogeneity of the human trachea progenitor

cell compartment. Some evidence from murine studies has supported the idea of

functional difference in subsets of basal cells based on expression of cytokeratins, K5

and K14 [16,44-46]. Other work suggests that the anatomical location of basal cells is a

Page 59: Identification of Molecular and Functional Heterogeneity

48

predictor of progenitor capacity [40,45,46]. In humans, the existence of cellular

heterogeneity has been supported by several lines of evidence [28,40,42]. In vitro

studies suggest there are differences in the differentiation potential of human p63+

progenitor cells that reside at different locations within airways. P63+ cells isolated

from the trachea were shown to differentiate into ciliated and goblet cells at ALI, while

those from the distal airway formed monolayers with very little mucociliary

differentiation [42]. Differences in clonogenic lineage potential of tracheal basal cells

have been shown in tracheal xenografts [40]. Furthermore, 1.6% of cells isolated from

murine trachea have some clonogenic potential, and 0.05- 0.1% of human and murine

tracheal cells can give rise to large colonies in vitro [42,44]. Since basal cells represent

approximately one third of all epithelial cells in the proximal airway [2], these numbers

suggest that rare basal cells possess clonogenic potential, while even fewer possess a

large capacity for proliferation. These results support the existence of subpopulations

within the basal lineage with distinct functional properties. Based on this evidence, we

set out to discover the frequency of such a population of cells in the human trachea, and

to find molecular markers which could be used to identify and separate functionally

distinct subpopulations.

4.1 Establishment of culture systems and assays

To begin to investigate heterogeneity in the basal cell compartment we first had

to obtain functional progenitor populations and establish techniques to evaluate them.

Using previously described methods, human tracheal epithelial cells (HTECs) were

dissociated from tracheal tissue and cultured to selectively expand basal cells while not

Page 60: Identification of Molecular and Functional Heterogeneity

49

supporting growth or survival of fibroblasts or columnar cells [47]. These culture-

purified basal cells, grown on plastic without passaging (P0plastic), possess the capacity

to differentiate into other functional tracheal lineages [38]. Using these methods, we

derived basal cells from a number of donor tracheas and confirmed their purity and

progenitor function.

The basal cell purity of P0plastic cells was initially assessed by FACS analysis

showing over 99% expressed CD44, an in vivo basal cell specific marker (fig. 1). Purity

was also evaluated by IF staining; nearly 90% of P0plastic cells stained positively for p63,

another basal cell-specific marker, and no cells expressed columnar cell markers MUC16

or polarized apical βTUBIV, markers for mucinous and ciliated cells respectively (fig. 1).

Given these data and our observation that with longer exposure times we could detect

low levels of p63 in “p63 negative” nuclei, we conclude that virtually all P0plastic cells

are basal cells. The reduced levels of p63 immunoreactivity observed in some cells

could be a staining artifact, or could represent cells undergoing a cellular process which

corresponds with decreased expression of p63; these changes may occur during cell

division or migration. Such processes may also account for the observed βTUBIV

cytoplasmic foci, which could represent cells in a particular stage of the cell cycle, or

cells undergoing specific morphological changes [58].

To verify progenitor activity of the P0plastic basal cells and to establish methods

to functionally interrogate basal cell subpopulations, we characterized the behaviour of

basal cells in ALI cultures. Basal cells were initially seeded subconfluently at ALI, grown

Page 61: Identification of Molecular and Functional Heterogeneity

50

to confluence while being fed both basolaterally and apically, and then fed only

basolaterally for 3 weeks. Assessment of p63 immunostaining and qPCR showed that

basal cells grown at ALI retain their basal identity until cultures reach confluence. By

three days post-confluence there were many p63 negative nuclei, and secretory cells

were detected by MUC16 protein and gene expression suggesting that progenitors were

transitioning into other lineages. Emergence of ciliated cells was apparent by 7 days

post confluence as assessed by IF staining. At 14 days post confluence, the presence of

beating cilia was visible by light microscopy, which corresponds with large increases in

FOXJ1 expression. These data demonstrate that P0plastic basal cells are a pure

population of progenitors that have retained mucociliary potential.

Having purified a population of functional progenitors we sought to develop a

method to functionally evaluate progenitor cells that is: consistent between samples,

easily monitored and compared, and gives good differentiation. Using the ALI culture

system to differentiate progenitor cells, we looked at ciliogenesis as a progenitor

output. Ciliogenesis is an ideal lineage readout as: it is robust; it can be observed

microscopically in living culture by light microscopy; and it can be measured

quantitatively at the level of gene and protein expression.

Histological sections have historically been used to examine ciliogenesis. They

reveal epithelial morphology, and IF staining provides a qualitative view of cell types

present and may allow some cell types to be quantified [41,61-63]. However, we found

histological sections to have limited value for quantitative analysis when limited

Page 62: Identification of Molecular and Functional Heterogeneity

51

differentiation is achieved. This is largely due to uneven distribution of ciliated cells. To

resolve this issue, we investigated two alternative methods to quantitatively assess

differentiation: qPCR and enface staining. We show that FOXJ1 gene induction tracks

well with emergence of ciliated cells as determined by IF staining for βTUBIV, (fig. 5 and

6). Furthermore, using IF staining, ciliated cells are easily delineated from non-ciliated

cells because βTUBIV is contained within membrane bound cilia projections. The enface

staining and tiling method described here allows the extent of differentiation to be

measured across the entire ALI culture, removing any variability due to uneven

distribution of ciliated cells.

We observed macroscopic secretions on the apical surface of the ALI culture

preceding appearance of functional cilia, suggesting the early emergence of secretory

cells. Surprisingly, antibody and gene expression analysis suggest these secretions are

due to MUC16 secretory cells, not due Muc5ac+ goblet cells (fig. 2, 3). Although we

found that P0plastic cells can differentiate into goblet cells in xenograft cultures, we

have not observed the emergence of goblet cells in ALI cultures, suggesting our ALI

conditions lack required signaling for this fate. Instead, our ALI conditions drive

physiologic levels of MUC16 secretory cell differentiation, another abundant secretory

lineage in the upper airway [56] (fig. 2, 3). We observed that Muc16 gene and protein

expression show similar kinetics at ALI, and are induced in all samples. However, trends

in induction and expression are not as robust or consistent between patient strains as

those observed with ciliated cell markers (fig. 5 and 6).

Page 63: Identification of Molecular and Functional Heterogeneity

52

Previous work has indicated that serial passage of human basal cells on plastic

leads to significant decline in progenitor potential [38]. We asked whether serial

passage of cells on ALI culture might provide better preservation of progenitor activity.

This could be important for maintaining progenitor cells for long term experiments or

expanding limited primary cells to acquire large numbers required for high-throughput

assays. We compared the progenitor activity of plastic-passaged cells to ALI-passaged

cells by comparing their ability to differentiate at ALI (fig. 7). Consistent with previous

reports, we found that the progenitor potential of plastic passaged cells fell drastically

over 2 passages. A similar decline was observed in the activity of ALI-passaged cells.

The extent of ciliogenesis was substantially lower in P2pA and PAPA cultures than in

P0pA from all patient strains examined. However, there was little impact on MUC16

lineage potential following P2pA and PAPA cultures, which showed similar MUC16

expression to P0pA cultures. These data show that ALI passaging is not better than

submerged LHC-9 culture at preserving mucociliary progenitor activity. The differential

impact of ciliary and mucinous potential could suggest these lineages are derived from

different progenitor cell populations, which are differentially maintained during culture.

Alternatively, the ciliogenic potential may be maintained but the signaling environment

of culture may preferentially drive a mucinous fate. Further development of both ALI

and submerged culture conditions could permit better maintenance or detection of

progenitor potential.

4.2 Evidence for functional heterogeneity

Page 64: Identification of Molecular and Functional Heterogeneity

53

To investigate functional heterogeneity within the basal cell compartment, we

employed a limiting dilution assay (LDA). In theory, if rare stem cells exist in our

P0plastic basal cell population, they would be diluted out as cultures were seeded at

lower densities. The distribution of these cells would follow a Poisson model, meaning

at some critical seeding density we would have a high probability of seeding one or zero

stem cells. At this density we would see some cultures succeed, while those cultures

not containing a stem cell would fail. Alternatively, we could see a similar result if the

cells were equipotent, but produced a paracrine factor required for growth, since a

minimum number of cells might be required for sufficient concentrations of that factor

to accumulate.

Using this technique, we were able to obtain sufficient data from 5 patient

strains to confirm that a single factor may be responsible for success of the cultures, as

defined by the cultures reaching confluence and maintaining a barrier (table 2). We

found that basal cells isolated from different patients performed significantly differently

in ALI cultures (table 3). The estimated frequency of a basal cell capable of

repopulating an ALI culture ranged from 1 in 96 to 1 in 1297 (0.08%-1%) (fig. 3). These

values are similar to those previously reported in human and mouse for the frequency

of cell capable of giving rise to large colonies (0.05- 0.1%) [42,44]. The slightly lower

frequency of clonogenic cell observed in these studies may be due to failure to sort

basal cells from columnar cells, differences between species, damage from the sorting

process [44], or culture conditions [42] used in these studies. Alternatively, this could

Page 65: Identification of Molecular and Functional Heterogeneity

54

reflect natural variation between patients based on biological differences such as age, or

gender.

The data presented here supports the existence of subsets of ALI repopulating

cells rather than the existence of a dilutable paracrine factor essential for growth. First,

the complete failure of one replicate culture at the same seeding density that results in

successful repopulation in another replicate culture is difficult to reconcile with dilution

of a paracrine factor unless an extreme error in seeding density occurred. Second, the

sometimes large differences in induction of differentiation genes between replicate

cultures (fig. 6, 7, 10 and 11), seeded at high initial densities, are consistent with the

notion that there are rare cells that give rise to differentiated progeny, and that these

cells are variably diluted during seeding. This observation is also difficult to reconcile

with the model that equipotent cells simply produce a limiting paracrine factor,

especially when cells are seeded at high densities. Our data support a model where rare

cells, representing less than 1% of P0plastic basal cells, have the ability to undergo

massive expansion and repopulate an ALI filter. However, the mucociliary

differentiation of these cultures may be dependent on additional factors or cell types.

None of the LDA experiments yielded sufficient results to test whether a single factor

was responsible for successful mucociliary differentiation. However, the difference in

our ability to test the single hit hypothesis for repopulation but not differentiation

suggests a different factor, or number of factors, is required for each process.

4.3 Evidence for molecular heterogeneity

Page 66: Identification of Molecular and Functional Heterogeneity

55

Large scale comprehensive surface marker profiling has not been reported for

lung basal cells in any species. To comprehensively investigate molecular heterogeneity

within the basal cell population we employed a high-throughput screen consisting of

339 antibodies. Here we consider a subpopulation to be any subset of P0plastic cells

that have differential expression of the examined surface marker. We initially identified

115 cell surface markers that denoted subpopulations of basal cells in 2 independent

patient strains. Of these markers, 74 were confirmed in a non-high throughput test on

cells from different patient strains, indicating reproducible surface marker

heterogeneity among human basal cells. The observed heterogeneity could be due to

the presence of hierarchical relationships between basal cells with different proliferative

and lineage potential, or result from equipotent basal cells that are in different states.

For example, these states could reflect different stages of maturation or differentiation

or stress response to in vitro culture. The cell surface marker signature of any single

subpopulation of basal cells could be comprised of single or multiple markers within the

group of 74. At present, it is unclear how many distinct populations are represented by

the 74 markers.

While the extent to which this surface marker heterogeneity reflects in vivo

populations has not been addressed in this work, our results provide tools for future

directed in vivo analyses. A previous study by Atsuta et al. looking at integrin expression

on human bronchial epithelial cells supports some of our findings. They also found

evidence for expression of 12 surface markers identified in our screen on P0plastic

bronchial epithelial cells [64]. However, they did not examine potential heterogeneity in

Page 67: Identification of Molecular and Functional Heterogeneity

56

expression of these markers. Our data suggest that 9 of these 12 markers are actually

heterogeneously expressed among basal cells.

Little work has been done to identify normal lung stem cell markers. However,

one recent report by Kajstura et al. proposed the existence of rare human CD117+,

KLF4+, NANOG+, OCT3/4+ and SOX2+ stem cells in the lung. While most of these cells

are p63 negative and lack epithelial markers, some evidence was provided that

indicated a fraction of these cells could acquire a p63 positive fate [28]. In our work, we

found inconsistent expression of CD117 across different patient samples. Furthermore,

we were unable to expand viable primary unpassaged HTECs using the culture

conditions that were reported to promote expansion of the CD117+ stem cells [28].

While it is possible that our failure to consistently detect such a cell could be due to its

relative enrichment in the distal rather than primary airways, the existence of CD117+

stem cells has yet to be confirmed by other labs.

It is possible that distinct progenitor cells throughout the airways share some

surface antigens. Studies of lung progenitor cells in distal regions of the airway have led

to the identification of several surface markers. Kim et al. described a bronchioalveolar

stem cell as Sca1+ and CD34+. Sca-1 is a murine specific antigen and we did not detect

CD34 immunoreactivity in our basal cells.

In some cases, cancer stem cells share phenotypic properties with normal stem

cells [65]. Several surface markers have been identified for lung cancer stem cells, also

known as tumour initiating cells (TIC). Tirino et al. proposed that a CD133+ cell fraction

Page 68: Identification of Molecular and Functional Heterogeneity

57

of non-small cell lung cancers (NSCLC) includes TICs [66]. They found a CD133+ fraction

in 72% of NSCLC, and showed that culturing these cells as non-adherent spheres

increased the proportion of CD133+ cells, which resulted in a nearly 4-fold increase in

tumour initiating capacity of these cultures in NOD/SCID mice [66]. However, CD133+

cells were not detected in our screen, which suggests that such cells may only arise in

disease states, or may reflect non-basal cell progenitors. More recent work by Zhang et

al. did not support CD133 as a marker for NSCLC TICs, but provided evidence for the

CD166+ fraction as marking the majority of TICs [67]. In our screen, all basal cells

expressed CD166, which could be consistent with some TICs arising from the p63+ basal

cell compartment.

Putative stem cell populations have been characterized in a number of other

epithelia. Notably, mammary glands are composed of a stratified epithelium where

p63+ basal cells are thought to contain the progenitor activity. CD49f, CD24 and CD29

have been identified as potential markers of mammary epithelial stem cells [68,69], and

CD49f has also been used to enrich progenitor activity in p63+ population of

keratinocytes [70]. All three of these markers were identified in our screen as being

heterogeneously expressed in lung basal cells and are promising candidates for further

study.

To investigate the existence of potential functional differences between marker-

positive and marker-negative subpopulations of P0plastic basal cells we looked at CD54

and podoplanin, markers of relatively abundant subpopulations which showed

Page 69: Identification of Molecular and Functional Heterogeneity

58

promising results in preliminary experiments. Cells were sorted based on expression of

CD54 or podoplanin, and fractions were assessed at ALI for: proliferative potential,

extent of differentiation, and emergence of mature cell types. For both CD54 and

podoplanin, the positive and negative fractions had indistinguishable lineage potential

as assessed by qPCR and enface staining of 21 day post confluent cultures. However,

monitoring of CD54 subpopulations revealed marked, reproducible differences in

culture morphology during epithelial regeneration (fig. 10). CD54 expression on airway

epithelial cells has been linked to local molecular events in inflammation in response to

allergen exposure, playing a role in eosinophil recruitment and infiltration [71]. Thus,

these morphological differences between CD54+ and CD54- generated ALI epithelia

could reflect functional differences in the ability to promote specific inflammatory

processes (fig. 10) [71,72]. The failure to detect a difference between podoplanin

subpopulations could reflect stochastic changes in ex vivo expression without functional

significance, or could reflect limitations of our assays in detecting functional differences

between subpopulations of basal cells. Notably, we only examined two lineage markers

and our assays do not extensively evaluate the self-renewal potential of basal cells.

The markers we have identified can now be used to help distinguish a classical

from non-classical stem cell model for epithelial regeneration in the trachea. Candidate

subpopulations can be sorted and re-cultured, and the proliferative ability of these cells,

as well as their ability to regenerate marker positive and marker negative states can be

assessed. Sorted cells can also be interrogated in ALI models employing serial

passaging, as well as tracheal xenograft models, which may help uncover functional

Page 70: Identification of Molecular and Functional Heterogeneity

59

differences between basal cell subpopulations. Additionally, antibody staining of

tracheal tissue could be used to distinguish relevant in vivo subpopulations from those

generated artifactually by ex vivo culture. Finally, multi-parametric FACS analysis of

P0plastic basal cells will be useful to determine how many subpopulations are

represented by the markers identified here.

Here I present data demonstrating human tracheal P0plastic cells are a purified

population of basal cells that retain progenitor activity (Fig. 1, 2 and 3). I provided

detailed analysis of the differentiation process of P0plastic basal cells on ALI, showing

robust induction of markers for both ciliogenic and secretory differentiation programs,

as well as optimized methods for assessing progenitor output and differentiation (Fig. 4,

5 and 6). I demonstrated that ALI culture conditions are able to maintain a progenitor

population at an extent similar to LHC-9 conditions (Fig. 7). Performing a limiting

dilution assay on basal cells with in vitro and in vivo validated progenitor activity, I

determined the frequency of clonogenic cells capable of repopulating ALI ranges from

0.08%-1%, and varies significantly between patients. Importantly, I contributed a list of

74 validated cell surface markers that are heterogeneously expressed amongst P0plastic

basal cells. Subpopulations varied in frequency from <1% to ~50%. Preliminary

functional analyses suggest there may be differences in the differentiation potential of

some of the subpopulations. This work supports the notion that the basal cell

compartment may be functionally heterogeneous, and provides a new arsenal of

molecular tools for the directed investigation of heterogeneity among human basal

cells.

Page 71: Identification of Molecular and Functional Heterogeneity

60

7. References

1. Message SD, Johnston SL (2004) Host defense function of the airway epithelium in health and disease: clinical background. J Leukoc Biol 75: 5-17.

2. Knight DA, Holgate ST (2003) The airway epithelium: structural and functional properties in health and disease. Respirology 8: 432-446.

3. Wanner A, Salathe M, O'Riordan TG (1996) Mucociliary clearance in the airways. Am J Respir Crit Care Med 154: 1868-1902.

4. Puchelle E, Zahm JM, Tournier JM, Coraux C (2006) Airway epithelial repair, regeneration, and remodeling after injury in chronic obstructive pulmonary disease. Proc Am Thorac Soc 3: 726-733.

5. Crystal RG, Randell SH, Engelhardt JF, Voynow J, Sunday ME (2008) Airway epithelial cells: current concepts and challenges. Proc Am Thorac Soc 5: 772-777.

6. Spina D (1998) Epithelium smooth muscle regulation and interactions. Am J Respir Crit Care Med 158: S141-145.

7. Evans CM, Williams OW, Tuvim MJ, Nigam R, Mixides GP, et al. (2004) Mucin is produced by clara cells in the proximal airways of antigen-challenged mice. Am J Respir Cell Mol Biol 31: 382-394.

8. Boers JE, Ambergen AW, Thunnissen FB (1999) Number and proliferation of clara cells in normal human airway epithelium. Am J Respir Crit Care Med 159: 1585-1591.

9. Evans MJ, Cox RA, Shami SG, Plopper CG (1990) Junctional adhesion mechanisms in airway basal cells. Am J Respir Cell Mol Biol 3: 341-347.

10. Evans MJ, Van Winkle LS, Fanucchi MV, Plopper CG (2001) Cellular and molecular characteristics of basal cells in airway epithelium. Exp Lung Res 27: 401-415.

11. Rhodin JA (1966) The ciliated cell. Ultrastructure and function of the human tracheal mucosa. Am Rev Respir Dis 93: Suppl:1-15.

12. Lackie PM, Baker JE, Gunthert U, Holgate ST (1997) Expression of CD44 isoforms is increased in the airway epithelium of asthmatic subjects. Am J Respir Cell Mol Biol 16: 14-22.

13. Romano RA, Ortt K, Birkaya B, Smalley K, Sinha S (2009) An active role of the DeltaN isoform of p63 in regulating basal keratin genes K5 and K14 and directing epidermal cell fate. PLoS One 4: e5623.

14. Guo X, Keyes WM, Papazoglu C, Zuber J, Li W, et al. (2009) TAp63 induces senescence and suppresses tumorigenesis in vivo. Nat Cell Biol 11: 1451-1457.

15. Cole BB, Smith RW, Jenkins KM, Graham BB, Reynolds PR, et al. (2010) Tracheal Basal cells: a facultative progenitor cell pool. Am J Pathol 177: 362-376.

16. Rock JR, Onaitis MW, Rawlins EL, Lu Y, Clark CP, et al. (2009) Basal cells as stem cells of the mouse trachea and human airway epithelium. Proc Natl Acad Sci U S A 106: 12771-12775.

17. Rawlins EL, Hogan BL (2006) Epithelial stem cells of the lung: privileged few or opportunities for many? Development 133: 2455-2465.

18. Rawlins EL, Okubo T, Que J, Xue Y, Clark C, et al. (2008) Epithelial stem/progenitor cells in lung postnatal growth, maintenance, and repair. Cold Spring Harb Symp Quant Biol 73: 291-295.

19. Boers JE, den Brok JL, Koudstaal J, Arends JW, Thunnissen FB (1996) Number and proliferation of neuroendocrine cells in normal human airway epithelium. Am J Respir Crit Care Med 154: 758-763.

Page 72: Identification of Molecular and Functional Heterogeneity

61

20. Jeffery PK (1983) Morphologic features of airway surface epithelial cells and glands. Am Rev Respir Dis 128: S14-20.

21. Thompson AB, Robbins RA, Romberger DJ, Sisson JH, Spurzem JR, et al. (1995) Immunological functions of the pulmonary epithelium. Eur Respir J 8: 127-149.

22. Morrison SJ, Uchida N, Weissman IL (1995) The biology of hematopoietic stem cells. Annu Rev Cell Dev Biol 11: 35-71.

23. Pinto D, Clevers H (2005) Wnt control of stem cells and differentiation in the intestinal epithelium. Exp Cell Res 306: 357-363.

24. Blenkinsopp WK (1967) Proliferation of respiratory tract epithelium in the rat. Exp Cell Res 46: 144-154.

25. Evans MJ, Cabral LJ, Stephens RJ, Freeman G (1975) Transformation of alveolar type 2 cells to type 1 cells following exposure to NO2. Exp Mol Pathol 22: 142-150.

26. Rawlins EL, Okubo T, Xue Y, Brass DM, Auten RL, et al. (2009) The role of Scgb1a1+ Clara cells in the long-term maintenance and repair of lung airway, but not alveolar, epithelium. Cell Stem Cell 4: 525-534.

27. Forbes S, Vig P, Poulsom R, Thomas H, Alison M (2002) Hepatic stem cells. J Pathol 197: 510-518.

28. Kajstura J, Rota M, Hall SR, Hosoda T, D'Amario D, et al. (2011) Evidence for human lung stem cells. N Engl J Med 364: 1795-1806.

29. Smith LJ (1985) Hyperoxic lung injury: biochemical, cellular, and morphologic characterization in the mouse. J Lab Clin Med 106: 269-278.

30. Evans MJ, Cabral-Anderson LJ, Dekker NP, Freeman G (1981) The effects of dietary antioxidants on NO2- induced injury to type 1 alveolar cells. Chest 80: 5-8.

31. Van Winkle LS, Johnson ZA, Nishio SJ, Brown CD, Plopper CG (1999) Early events in naphthalene-induced acute Clara cell toxicity: comparison of membrane permeability and ultrastructure. Am J Respir Cell Mol Biol 21: 44-53.

32. Hong KU, Reynolds SD, Giangreco A, Hurley CM, Stripp BR (2001) Clara cell secretory protein-expressing cells of the airway neuroepithelial body microenvironment include a label-retaining subset and are critical for epithelial renewal after progenitor cell depletion. Am J Respir Cell Mol Biol 24: 671-681.

33. Reynolds SD, Giangreco A, Power JH, Stripp BR (2000) Neuroepithelial bodies of pulmonary airways serve as a reservoir of progenitor cells capable of epithelial regeneration. Am J Pathol 156: 269-278.

34. Kim CF, Jackson EL, Woolfenden AE, Lawrence S, Babar I, et al. (2005) Identification of bronchioalveolar stem cells in normal lung and lung cancer. Cell 121: 823-835.

35. Jackson EL, Willis N, Mercer K, Bronson RT, Crowley D, et al. (2001) Analysis of lung tumor initiation and progression using conditional expression of oncogenic K-ras. Genes Dev 15: 3243-3248.

36. Whitcutt MJ, Adler KB, Wu R (1988) A biphasic chamber system for maintaining polarity of differentiation of cultured respiratory tract epithelial cells. In Vitro Cell Dev Biol 24: 420-428.

37. Engelhardt JF, Yankaskas JR, Wilson JM (1992) In vivo retroviral gene transfer into human bronchial epithelia of xenografts. J Clin Invest 90: 2598-2607.

38. Gray TE, Guzman K, Davis CW, Abdullah LH, Nettesheim P (1996) Mucociliary differentiation of serially passaged normal human tracheobronchial epithelial cells. Am J Respir Cell Mol Biol 14: 104-112.

39. Matsui H, Randell SH, Peretti SW, Davis CW, Boucher RC (1998) Coordinated clearance of periciliary liquid and mucus from airway surfaces. J Clin Invest 102: 1125-1131.

Page 73: Identification of Molecular and Functional Heterogeneity

62

40. Engelhardt JF, Schlossberg H, Yankaskas JR, Dudus L (1995) Progenitor cells of the adult human airway involved in submucosal gland development. Development 121: 2031-2046.

41. Hajj R, Baranek T, Le Naour R, Lesimple P, Puchelle E, et al. (2007) Basal cells of the human adult airway surface epithelium retain transit-amplifying cell properties. Stem Cells 25: 139-148.

42. Kumar PA, Hu Y, Yamamoto Y, Hoe NB, Wei TS, et al. (2011) Distal airway stem cells yield alveoli in vitro and during lung regeneration following H1N1 influenza infection. Cell 147: 525-538.

43. Engelhardt JF, Yankaskas JR, Ernst SA, Yang Y, Marino CR, et al. (1992) Submucosal glands are the predominant site of CFTR expression in the human bronchus. Nat Genet 2: 240-248.

44. Schoch KG, Lori A, Burns KA, Eldred T, Olsen JC, et al. (2004) A subset of mouse tracheal epithelial basal cells generates large colonies in vitro. Am J Physiol Lung Cell Mol Physiol 286: L631-642.

45. Hegab AE, Ha VL, Gilbert JL, Zhang KX, Malkoski SP, et al. (2011) Novel stem/progenitor cell population from murine tracheal submucosal gland ducts with multipotent regenerative potential. Stem Cells 29: 1283-1293.

46. Borthwick DW, Shahbazian M, Krantz QT, Dorin JR, Randell SH (2001) Evidence for stem-cell niches in the tracheal epithelium. Am J Respir Cell Mol Biol 24: 662-670.

47. John F. Lechner ML (1981) A Serum-free Method for Culturing Normal Human Bronchial Epithelial Cells at Clonal Density. Methods in Cell Science 9: 43-48.

48. Shi SR, Chaiwun B, Young L, Cote RJ, Taylor CR (1993) Antigen retrieval technique utilizing citrate buffer or urea solution for immunohistochemical demonstration of androgen receptor in formalin-fixed paraffin sections. J Histochem Cytochem 41: 1599-1604.

49. Shi SR, Liu C, Taylor CR (2007) Standardization of immunohistochemistry for formalin-fixed, paraffin-embedded tissue sections based on the antigen-retrieval technique: from experiments to hypothesis. J Histochem Cytochem 55: 105-109.

50. Hu Y, Smyth GK (2009) ELDA: extreme limiting dilution analysis for comparing depleted and enriched populations in stem cell and other assays. J Immunol Methods 347: 70-78.

51. Yamaya M, Finkbeiner WE, Chun SY, Widdicombe JH (1992) Differentiated structure and function of cultures from human tracheal epithelium. Am J Physiol 262: L713-724.

52. Karp PH, Moninger TO, Weber SP, Nesselhauf TS, Launspach JL, et al. (2002) An in vitro model of differentiated human airway epithelia. Methods for establishing primary cultures. Methods Mol Biol 188: 115-137.

53. Wu R, Zhao YH, Chang MM (1997) Growth and differentiation of conducting airway epithelial cells in culture. Eur Respir J 10: 2398-2403.

54. Renthal R, Schneider BG, Miller MM, Luduena RF (1993) Beta IV is the major beta-tubulin isotype in bovine cilia. Cell Motil Cytoskeleton 25: 19-29.

55. Jensen-Smith HC, Luduena RF, Hallworth R (2003) Requirement for the betaI and betaIV tubulin isotypes in mammalian cilia. Cell Motil Cytoskeleton 55: 213-220.

56. Davies JR, Kirkham S, Svitacheva N, Thornton DJ, Carlstedt I (2007) MUC16 is produced in tracheal surface epithelium and submucosal glands and is present in secretions from normal human airway and cultured bronchial epithelial cells. Int J Biochem Cell Biol 39: 1943-1954.

57. Hackett BP, Brody SL, Liang M, Zeitz ID, Bruns LA, et al. (1995) Primary structure of hepatocyte nuclear factor/forkhead homologue 4 and characterization of gene expression in the developing respiratory and reproductive epithelium. Proc Natl Acad Sci U S A 92: 4249-4253.

Page 74: Identification of Molecular and Functional Heterogeneity

63

58. Dumontet C, Duran GE, Steger KA, Murphy GL, Sussman HH, et al. (1996) Differential expression of tubulin isotypes during the cell cycle. Cell Motil Cytoskeleton 35: 49-58.

59. Yu X, Ng CP, Habacher H, Roy S (2008) Foxj1 transcription factors are master regulators of the motile ciliogenic program. Nat Genet 40: 1445-1453.

60. Forbes SJ, Poulsom R, Wright NA (2002) Hepatic and renal differentiation from blood-borne stem cells. Gene Ther 9: 625-630.

61. You Y, Richer EJ, Huang T, Brody SL (2002) Growth and differentiation of mouse tracheal epithelial cells: selection of a proliferative population. Am J Physiol Lung Cell Mol Physiol 283: L1315-1321.

62. LeSimple P, van Seuningen I, Buisine MP, Copin MC, Hinz M, et al. (2007) Trefoil factor family 3 peptide promotes human airway epithelial ciliated cell differentiation. Am J Respir Cell Mol Biol 36: 296-303.

63. Prytherch Z, Job C, Marshall H, Oreffo V, Foster M, et al. (2011) Tissue-Specific stem cell differentiation in an in vitro airway model. Macromol Biosci 11: 1467-1477.

64. Atsuta J, Sterbinsky SA, Plitt J, Schwiebert LM, Bochner BS, et al. (1997) Phenotyping and cytokine regulation of the BEAS-2B human bronchial epithelial cell: demonstration of inducible expression of the adhesion molecules VCAM-1 and ICAM-1. Am J Respir Cell Mol Biol 17: 571-582.

65. Bonnet D, Dick JE (1997) Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat Med 3: 730-737.

66. Tirino V, Camerlingo R, Franco R, Malanga D, La Rocca A, et al. (2009) The role of CD133 in the identification and characterisation of tumour-initiating cells in non-small-cell lung cancer. European Journal of Cardio-Thoracic Surgery 36: 446-453.

67. Zhang WC, Shyh-Chang N, Yang H, Rai A, Umashankar S, et al. (2012) Glycine decarboxylase activity drives non-small cell lung cancer tumor-initiating cells and tumorigenesis. Cell 148: 259-272.

68. Shackleton M, Vaillant F, Simpson KJ, Stingl J, Smyth GK, et al. (2006) Generation of a functional mammary gland from a single stem cell. Nature 439: 84-88.

69. Stingl J, Eirew P, Ricketson I, Shackleton M, Vaillant F, et al. (2006) Purification and unique properties of mammary epithelial stem cells. Nature 439: 993-997.

70. Li A, Simmons PJ, Kaur P (1998) Identification and isolation of candidate human keratinocyte stem cells based on cell surface phenotype. Proc Natl Acad Sci U S A 95: 3902-3907.

71. Ciprandi G, Pronzato C, Ricca V, Passalacqua G, Bagnasco M, et al. (1994) Allergen-specific challenge induces intercellular adhesion molecule 1 (ICAM-1 or CD54) on nasal epithelial cells in allergic subjects. Relationships with early and late inflammatory phenomena. Am J Respir Crit Care Med 150: 1653-1659.

72. Striz I, Mio T, Adachi Y, Heires P, Robbins RA, et al. (1999) IL-4 induces ICAM-1 expression in human bronchial epithelial cells and potentiates TNF-alpha. Am J Physiol 277: L58-64.

Page 75: Identification of Molecular and Functional Heterogeneity

64

Appendices

Appendix A

Antibody Str. 38 Str. 23

Single

tube

CD50 0.0367 2.55E-03 0

CD123 0.18 0.139 0.054

CD180 0.182 0.0448 0

CD195 0.283 1.36 0

CD249 0.332 0.829 0

CD33 0.399 0.64 0.017

CD22 0.441 0.548 0.055

CD122 0.502 0.231 0.46

CD62L 0.511 1.11 0

CD312 0.519 1.25 0

CD181 0.533 0.555 0.48

CDW93 0.567 0.241 0

CD62P 0.609 0.4 0

CD275 0.808 0.173 0.49

CD116 0.948 0.618 0.89

CD266 0.978 0.991 0.071

CD213a2 1.05 0.589 0

CD159c 1.07 1.5 0

CD182 1.07 0.312 0.12

VBTA8 1.07 1.49 0.88

CD32 1.09 0.364 0

CA9 1.11 1.64 0.036

CD16 1.13 0.461 0.085

CD82 1.22 1.35 0.045

CD6 1.26 1.31 0

CD132 1.42 0.383 0.87

CD7 1.65 1.15 0.11

HLA DM 1.69 0.953 0

CD193 1.83 1.95 0.37

CD247 0.207 2.22 0

CD138 0.697 5.36 0

CD85g 1.41 2.35 0

CD56 1.71 2.32 0

CD164 2.01 1.34 0

CD158e2 2.07 1.28 0.65

CD99 2.09 3.36 0

CD227 2.09 2.43 1.48

CD38 2.61 5.97 0

CD117 2.66 0.338 6.35

CD2 2.68 3.38 0

CD87 3.09 0.757 0

Page 76: Identification of Molecular and Functional Heterogeneity

65

CD97 3.09 3.37 0

SSEA-4 3.11 0.632 0.58

CD175s 3.17 8.88 0

FOXP3 3.9 0.0561 0

VDTA2 4.16 2.38 1.31

CD267 4.42 1.17 1.01

CD125 4.52 1 0

CD157 4.89 13 0.015

CD268 4.91 1.31 1.96

CD13 5.37 9.3 31.6

CD90 5.54 1.93 4.42

CD49D 5.55 8.4 1.85

CD51 5.76 1.34 0

GMA DTA 5.86 0.805 0.76

CD36L1 6.18 5.53 1.22

CD91 7.46 6.05 0

CD127 8.14 0.707 0.61

CD61 3.17 17.7 0

CD170 4.57 18.8 0

CD295 6 13.9 0.43

CD314 14.8 2.55 1.86

CD337 17.8 4.26 0.72

CDH3 18.1 49.3 0

CD158A 19.2 0.821 0.015

CD253 19.3 2.37 0.057

CDW218a 20 24.9 0.48

SSEA-3 20.6 3.16 0.017

CD245 21 46.2 22.8

CD252 23.2 2.16 0.31

CD10 27.7 9.02 2.79

CD102 29.3 21.9 3.18

CD263 30.8 2.81 0.63

CD49A 38.7 37.4 0

6D12 39.9 20.7 30.5

CD66C 45 17.5 17.5

CD271 21.9 87.7 2.28

CD205 41.2 61.3 0

CD201 32.8 55.7 17.8

CD148 46.3 56.4 1.51

CD151 58 1.82 7.28

CD130 61.5 14.2 1.41

CD54 67 43.4 31.2

CD108 72.4 33.3 41.9

CD105 76.9 68 0

CD71 82.9 28.7 0.91

CD264 85.8 36.9 42.4

Page 77: Identification of Molecular and Functional Heterogeneity

66

CD65s 86.9 76.1 15.9

CD63 87 77.8 6.84

Podoplanin 89.7 71.3 58.6

CD261 82 92.1 0.36

CD262 82.1 90.3 21.7

CD223 57.4 94.1 44.6

CD44 66.4 94.2 N/A

EGF-R 91.3 96.6 5.42

CD66 95.7 76.3 39

CD9 96.9 96.7 73.1

CD340 97 99.1 75.6

CD221 98.2 99 1.45

CD142 98.2 96.2 95.1

CD47 98.5 99.9 3.49

CD326 98.5 98.7 65.7

CD81 98.5 93.9 75.9

CD24 98.7 74.9 62.3

CD172a 98.8 50.8 47.4

CD29 98.8 99.8 100

CD119 99.1 97.7 1.34

CD58 99.1 99.5 99.42

CD276 99.4 99.7 100

CD49F 99.5 99.9 99.1

CD147 99.7 99.5 100

TNFR RLP 85.1 99.3 1.17

Page 78: Identification of Molecular and Functional Heterogeneity

67

Appendix B

Sequence of primers

Gene Forward Primer Reverse Primer MUC16

Np63 TAp63 FOXJ1 TBP

TGC GGT GTC CTG GTG ACC ACC CGC GGA AAC AAT GCC CAG ACT CAA TGT ATC CGC ATG CAG GAC T CAC CTG AGC CGA GCC GGG ACT TAG CGG TGT GCA CAG GAG CCA AGA GT

CAC CGG CAA GTT CCA GTC ATT GC TGC GCG TGG TCT GTG TTA CTG TGT TAT AGG GAC TGG TGG AC CTC CCG TTA CAC GGC CTC CCG ATT TTC TTG CTG CCA GTC TGG

Page 79: Identification of Molecular and Functional Heterogeneity

68

Appendix C Experimental data from LDA experiments

Str: 29945 Str: 38 Str: 23 Str: 78297 Str: 32 Str: 18 Str: 79168 Str: 39

Cell No: 2000 6000 6000 1500 31000 31000 100 6000

Confluence 6/ 6 6 /6 6/ 6 6/ 6 2/ 2 4/4 4/6 8/ 8

Barrier 6 6 6 6 2 4 0 8

Cilia 6 6 6 6 2 4 0 8

Cell No: 500 3500 3000 1000 6000 6000 50 500

Confluence 4/6 6/6 6/6 6/6 2/ 2 4/4 2/6 8/8

Barrier 2 6 6 6 2 4 0 8

Cilia 0 6 6 6 2 4 0 8

Cell No: 250 2500 2000 500 1000 1000 10 100

Confluence 2/6 6/6 6/6 6/6 4/ 4 4/4 1/ 6 8/8

Barrier 0 6 6 6 4 4 0 5

Cilia 0 6 6 6 0 4 0 0

Cell No: 50 1500 1000 250 500 500 1

Confluence 0/6 4/6 6/6 5/6 4/4 4/4 0/ 6

Barrier 0 4 6 5 0 4 0

Cilia 0 4 6 5 0 4 0

Cell No: 12000 250 250

10000, 5000, 2500, 1500, & 500

Confluence 2/ 2 2/ 4 4/4 1/1

Barrier 2 0 4 1

Cilia 2 0 4 1

Cell No: 500

50 50

Confluence 0/2

2/ 4 4/4

Barrier 0

0 4

Cilia 0

0 4

Cell No:

10 10

Confluence

0 of 4 4

Barrier

0 4

Cilia 0 4

Page 80: Identification of Molecular and Functional Heterogeneity

69

Str: 31 Str: 27 Str: 55 Str: 94214

Cell No: 6000 6000 6000 10 000

Confluence 7/ 7 5 / 5 8/ 8 16/16

Barrier 7 5 8 16

Cilia 7 0 8 16

Cell No: 100 500 500 100

Confluence 6/ 7 5/5 2/ 6 16/ 16

Barrier 2 5 0 16

Cilia 0 0 0 16

Cell No: 100 100

Confluence 3/ 5 0/ 6

Barrier 1 0

Cilia 0 0