144
Characterization of Mesenchymal Stromal Cells Derived from Human Umbilical Cord Tissue By Vanessa Noemi Raileanu A thesis submitted in conformity with the requirements for the degree of Master of Science Graduate Department of Physiology University of Toronto © Copyright by Vanessa Noemi Raileanu 2015

By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

  • Upload
    others

  • View
    7

  • Download
    0

Embed Size (px)

Citation preview

Page 1: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

Characterization of Mesenchymal Stromal Cells Derived from

Human Umbilical Cord Tissue

By

Vanessa Noemi Raileanu

A thesis submitted in conformity with the requirements

for the degree of Master of Science

Graduate Department of Physiology

University of Toronto

© Copyright by Vanessa Noemi Raileanu 2015

Page 2: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

II

Characterization of mesenchymal stromal cells derived from human

umbilical cord tissue

Vanessa Noemi Raileanu

Master of Science

Department of Physiology

University of Toronto

2015

Abstract

Mesenchymal stromal cells (MSCs) have emerged as candidates with therapeutic

potential to treat different pathologies. MSCs isolated from the bone marrow are most

commonly used, however, umbilical cord (UC) tissue presents a source that has not been

as extensively studied, yet can be obtained with more ease. Here, we characterize UC-

MSCs obtained from 40 patient samples and conclude that different cell populations have

the same phenotypic profile. TSG-6 has recently been suggested as a biomarker that can

be used in order to predict the in vivo efficacy of different MSC populations, and we

show that within a subset of UC-MSC samples, there are variations in the expression

levels of TSG-6, however this does not correlate with the cytokine secretion profiles or

the wound healing capacity of the cells in a db/db male mouse excisional wound splinting

model.

Page 3: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

III

Acknowledgements

I would like to express my gratitude to my supervisor, Dr. Ian Rogers, for giving

me the opportunity to work on this project, for his constant support, knowledge and for

being my mentor. This was an amazing learning experience and it developed and

moulded my scientific mind in many ways. I would like to thank my committee members,

Dr. Armand Keating and Dr. Robert Casper for their support and ideas, helping to shape

my project and encouraging my work. Thank you to Annie and Michael for training me

and answering my numerous flow cytometry related questions and to Jenn and Theresa

for helping with the animal studies. I am grateful to Dr. Brown’s lab, specifically Alex

and Prem, for teaching and guiding me through real-time RT-PCR. Thank you to

Insception LifeBank for providing the umbilical cord samples and to Dr. Sue Mueller for

allocating your time and effort to this task. Everyone at Insception has been incredibly

welcoming, friendly and I am grateful for having had the opportunity to be exposed to the

industry aspect of science. Finally, thank you to my friends who have many times been

the source of laughter throughout the years.

To Eddie, you have always stood by my side and made this experience memorable, I

couldn’t have imagined it without you.

My family has been the roots and source of love, support, understanding, and guidance.

Dad, your worldly advice has been invaluable. Mom, your love of science and vast

amount of knowledge that you garner has been my fuel for learning.

Page 4: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

IV

Table of Contents

ACKNOWLEDGEMENTS ....................................................................................................................... III

TABLE OF CONTENTS ........................................................................................................................... IV

LIST OF ABBREVIATIONS .................................................................................................................... VI

LIST OF TABLES ................................................................................................................................... VIII

LIST OF FIGURES .................................................................................................................................... IX

CHAPTER ONE: INTRODUCTION ......................................................................................................... 1

1. INTRODUCTION .................................................................................................................................... 2

1.1 MSCS: STEM OR STROMAL? .................................................................................................................. 2 1.1.1 Pluripotent Stem Cells ................................................................................................................. 2 1.1.2 Stem Cell Nomenclature ............................................................................................................... 8 1.1.3 Tissue Specific Stem Cells ............................................................................................................ 9 1.1.4 Mesenchymal Stem Cells .............................................................................................................10 1.1.5 MSC Immunophenotype ..............................................................................................................12

1.2 IN VIVO IDENTITY................................................................................................................................13 1.2.1 Pericytes ......................................................................................................................................13 1.2.2 Neural Crest Cells .......................................................................................................................15

1.3 SOURCES OF HUMAN MESENCHYMAL STROMAL CELLS ......................................................................16 1.3.1 Embryonic Tissue ........................................................................................................................16 1.3.2 Adult Tissue .................................................................................................................................17 1.3.3 Birth Associated Tissues .............................................................................................................20

1.4 IMMUNOMODULATION .........................................................................................................................25 1.4.1 Innate Immunity ..........................................................................................................................25 1.4.2 Adaptive Immunity ......................................................................................................................28

1.5 WOUND HEALING ................................................................................................................................30 1.5.1 Tissue Repair ..............................................................................................................................30 1.5.2 Diabetic Wound Healing .............................................................................................................32 1.5.3 TSG-6 ..........................................................................................................................................39 1.5.4 Animal Models ............................................................................................................................44 1.5.5 Diabetic Mouse Models of Wound Healing ................................................................................45

1.6 RATIONALE, HYPOTHESIS AND OBJECTIVES .........................................................................50

1.6.1 Rationale .....................................................................................................................................50 1.6.2 Hypothesis and Objectives ..........................................................................................................50

CHAPTER TWO: EXPERIMENTAL METHODS AND MATERIALS ..............................................52

2. EXPERIMENTAL METHODS AND MATERIALS...........................................................................53

2.1 UMBILICAL CORD COLLECTION AND PREPARATION ............................................................................53 2.2 MSC ISOLATION ..................................................................................................................................53 2.3 CELL CULTURE ....................................................................................................................................56 2.4 CRYOPRESERVATION ...........................................................................................................................56 2.5 FLOW CYTOMETRY ANALYSIS .............................................................................................................56 2.6 RNA EXTRACTION AND REAL-TIME PCR ...........................................................................................59 2.7 CYTOKINE ARRAY ...............................................................................................................................61

2.7.1 Array Procedure .........................................................................................................................61 2.7.2 Data Analysis ..............................................................................................................................65

2.8 EXCISIONAL MURINE MODEL ..............................................................................................................65 2.8.1 Surgical Procedure .....................................................................................................................65 2.8.2 Wound Analysis ...........................................................................................................................66 2.8.3 Tissue Collection and Fixation ...................................................................................................68

Page 5: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

V

2.8.4 Immunohistochemistry ................................................................................................................68 2.9 STATISTICAL ANALYSIS .......................................................................................................................69

CHAPTER THREE: RESULTS ................................................................................................................70

3. RESULTS .................................................................................................................................................71

3.1 MSC ISOLATION EFFICIENCY AND EXPLANT CULTURE .......................................................................71 3.2 MSC IMMUNOPHENOTYPE ...................................................................................................................75 3.3 MSC WOUND HEALING EFFICIENCY ...................................................................................................83

3.4 TSG-6 Expression ..........................................................................................................................85 3.5 Cytokine Secretion Analysis ...........................................................................................................88

3.6 MURINE EXCISIONAL WOUND HEALING ..............................................................................................91

CHAPTER FOUR: DISCUSSION ...........................................................................................................104

4. DISCUSSION .........................................................................................................................................105

4.1 CHANGES IN MSC PROFILE WITH CULTURE .......................................................................................105 4.2 PARACRINE SIGNALLING ...................................................................................................................107 4.3 DIABETES COMPLICATIONS ...............................................................................................................111 4.4 TSG-6 AND WOUND HEALING ...........................................................................................................112

5. CONCLUSION AND FUTURE STUDIES .........................................................................................121

6. REFERENCES ......................................................................................................................................123

Page 6: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

VI

List of Abbreviations

Alpha-MEM Alpha minimum essential medium

AP-1 Activator protein 1

APC Antigen presenting cells

APC Allophycoerythrin

BM-MSC Bone marrow mesenchymal stromal cells

CCL5 Chemokine (C-C motif) ligand 5

CD Cluster of differentiation

CFU-F Colony forming until-fibroblastoid

CT Cord tissue

CXCL Chemokine (C-X-C motif) ligand

DC Dendritic cells

DMEM Dulbecco's modified eagle's medium

EGF Epidermal growth factor

ES cell Embryonic stem cell

FGF-basic Fibroblast growth factor-basic

FGF-4 Fibroblast growth factor-4

FITC Fluorescein isothiocyanate

FS Forward scatter

FS TOF Forward scatter time of flight

GAGs Glycosylaminoglycans

GM-CSF Granulocyte-macrophage colony-stimulating factor

GvHD Graft-versus-host

HA Hyaluronic acid

HGF Hepatocyte growth factor

HSCs Hematopoietic stem cell

HUCPVCs Human umbilical cord perivascular cells

ICAM-1 Intercellular adhesion molecule 1

ICM Inner cell mass

IDO Indolemine 2,3-dioxygenase

IFN-γ Interferon gamma

IGF-1 Insulin-like growth factor 1

IL Interleukin

ISCT International Society for Cellular Therapy

LIF Leukemia inhibitory factor

LPA Lipoaspirate

MHC Major histocompatibility complex

MIF Migration inhibitory factor

MIP Macrophage inflammatory protein

MMPs Matrix metalloproteinases

MPSC Multi-potential stem cell

MSCs Mesenchymal stromal cells

NF-IL6 Nuclear factor IL-6

NF-κB Nuclear factor-κB

NK Natural killer

Page 7: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

VII

PE Phycoerythrin

PerCP-Cy5.5 Peridin chlorophyll protein-cyanine 5.5

PGE-2 Prostaglandin E2

PBLs Peripheral blood leukocytes

PBS Phosphate buffered saline

RIN RNA integrity number

SCF Stem cell factor

SS Side scatter

SSEA-3 Stage specific embryonic antigen-3

SSEA-4 Stage specific embryonic antigen-4

SS TOF Side scatter time of flight

STZ Streptozoicin

TGF-β Transforming growth factor-β

TLR Toll-like receptor

TNF-α Tumour necrosis factor alpha

TSG-6 Tumor necrosis factor-stimulated gene 6

UCT Umbilical cord tissue

UCB Umbilical cord blood

VCAM-1 Vascular cell adhesion molecule 1

VEGF Vascular endothelial growth factor

vWF von Willebrand factor

Page 8: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

VIII

List of Tables

Table 1: Antibodies and fluorochromes used to analyze UCT-MSCs. ............................. 58

Table 2: Real time RT-PCR Primer Sequences. ............................................................... 60

Table 3: Cytokines detected in the array kit. .................................................................... 63

Table 4: Personal data collected for each cord sample. .................................................... 73

Table 5: Growth characteristics of individual cord samples. ............................................ 74

Table 6: RNA integrity analysis for a subset of UCT-MSCs analyzed for TSG-6 mRNA

expression levels. .............................................................................................................. 86

Page 9: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

IX

List of Figures

Figure 1: Developmental Potential. .................................................................................... 7

Figure 2: Stages of wound healing.................................................................................... 37

Figure 3: MSC anti-inflammatory effects are mediated through TSG-6. ......................... 43

Figure 4: Explant culture of human umbilical cord samples. ........................................... 55

Figure 5: Wound healing calculations. ............................................................................. 67

Figure 6. Time to cell outgrowth for 40 UCT-MSC samples. .......................................... 75

Figure 7. Cell number obtained at first passage for 9 UCT-MSC samples. ..................... 76

Figure 8. Cell number obtained at each passage for three MSC populations. .................. 77

Figure 9: Colour density plots of CT16, CT24, and CT15 at early, mid and late analysis.

........................................................................................................................................... 80

Figure 10: Percent of cells illustrating expression of hematopoietic and stromal markers

at early, mid, and late analysis for 20 samples analyzed at each passage. ........................ 82

Figure 11: Percent of cells illustrating expression of hematopoietic and stromal markers

at early, mid, and late analysis for 40 samples analyzed at early passage and 20 cord

tissues analyzed at mid and late passages. ........................................................................ 83

Figure 12: Variable TSG-6 mRNA expression among a subset of UCT-MSC samples. . 86

Figure 13: TSG-6 mRNA correlation with maternal age and newborn weight. ............... 87

Figure 14: Cytokine secretion profiles of CT15 expressing low TSG-6 mRNA, CT16

expressing high TSG-6 mRNA, and CT24 illustrating no TSG-6 mRNA expression. .... 89

Figure 15: Wound closure analysis. .................................................................................. 94

Figure 16: Wound bed histology for CT16-treated mice. ................................................. 96

Figure 17: Wound bed histology for CT15-treated mice. ................................................. 98

Figure 18: Wound bed histology for control mice. ......................................................... 100

Figure 19: Wound bed immunohistochemistry for CT-16 treated mice. ........................ 102

Figure 20: Wound bed immunohistochemistry for CT-15 treated mice. ........................ 103

Page 10: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

X

Figure 21: Wound closure analysis illustrating different interpretations........................ 119

Figure 22: CT15-treated mouse wound healing calculations excluded for days 7, 10, and

14..................................................................................................................................... 120

Page 11: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

Chapter One: Introduction

Page 12: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

2

1. Introduction

1.1 MSCs: stem or stromal?

1.1.1 Pluripotent Stem Cells

The defining characteristics of pluripotent stem cells include the ability to differentiate

into any cell type after unlimited cell renewal in the stem cell state. This is best illustrated

by their ability to contribute to all tissues of the mouse when pluripotent stem cells are

incorporated into aggregation chimeras (Puri & Nagy, 2012). Some of the first studies

done on pluripotent stem cells were on cells derived from mouse teratocarcinomas in

which the tumour was seen to contain stem cells, known as embryonal carcinoma cells (G.

R. Martin & Evans, 1975). These cells were observed to exhibit pluripotency,

differentiating to form the endoderm, mesoderm, as well as the ectoderm (G. R. Martin &

Evans, 1975). After in vitro culturing under a defined set of medium conditions, the

embryonal carcinoma cells were seen to form keratinizing epithelium, endodermal cysts,

fibroblasts, cartilage, adipose tissue, beating muscles, pigmented cells, and neural cells

(G. R. Martin & Evans, 1975). As such, pluripotent cells can be stimulated under

different culture conditions to differentiate into various cell types. Since that time,

pluripotent stem cells have classically been obtained from the foetus. Mammalian

development begins when an oocyte is fertilized by a sperm, forming a single cell embryo,

the zygote. The zygote is totipotent, as it can give rise to an embryo with all the cells

needed to form an organism, as well as the placenta which is vital for fetal development

(Mitalipov & Wolf, 2009). As a result, each cell that is considered totipotent can give rise

to a whole organism, and this is said to be true until the four cell stage embryo in humans

(Figure 1) (Mitalipov & Wolf, 2009). Mammalian embryogenesis begins with a set of

Page 13: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

3

cleavage divisions to generate a population of blastomeres, which eventually undergo

cellular differentiation followed by the segregation of the different developmental

lineages, compaction and formation of the blastocyst (Nichols et al., 1998). The

trophectoderm, the outer layer of cells of the blastocyst, forms the trophoblast and

components of the placenta, whereas the inner cell mass (ICM) of the blastocyst gives

rise to non-trophoblast extraembryonic tissues and of all fetal cell types, including germ

cells (Nichols et al., 1998). The ICM becomes more differentiated eventually giving rise

to all three germ layers (endoderm, mesoderm, and ectoderm). This cell population is

considered to be pluripotent, because it can form cells of the three germ layers, having the

capacity to give rise to any fetal and adult cell type (Mitalipov & Wolf, 2009). The

pluripotent cell state of the ICM does not exist for a prolonged period of time, however,

the pluripotent cells found in the ICM can be isolated as embryonic stem cells (ES) from

the blastocyst, obtained from the pre-implantation embryo (Boroviak, Loos, Bertone,

Smith, & Nichols, 2014; Takahashi & Yamanaka, 2006). As a result, human pluripotent

stem cells include human embryonic stem cells and induced pluripotent stem cells. ES

cells were first isolated from the ICM of the 129 SvE strain mouse by Evans and

Kaufman in 1981, and also by Martin in that same year from early blastocysts obtained

by mating random bred ICR female mice with SWR/J males (Evans & Kaufman, 1981; G.

R. Martin, 1981). The derived ES cell lines exhibited the ability to proliferate, divide

indefinitely, differentiate into cells of the three germ layers (Mitalipov & Wolf, 2009). In

the mouse, ES cells are obtained from ICM of the late blastocyst at 4 days post coitum,

and are often maintained in media with the addition of leukemia inhibitory factor (LIF),

fetal calf serum, and feeder layers, and hence can be passaged indefinitely without

Page 14: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

4

differentiation (Tang et al., 2010). However, different mouse strains have a different

efficiency in establishing ES cell lines. The 129/Sv strain is most often used. ES cell lines

that have been used in the past include CCE, D3, and E14, which have been derived from

the 129/Sv strain (Kawase et al., 1994). The 129/Sv strain exhibits a high incidence of

spontaneous testicular teratomas as well as teratocarcinomas, and as a result, this strain

has been used as a source of embryonic carcinoma cell lines as well (Kawase et al., 1994).

Many ES cell lines have also been obtatined from the C57BL/6 strain as well as the

BALB/c strain, however BALB/c strain shows a lower efficiency in establishing ES cell

lines (Kawase et al., 1994). It has been shown that live offspring can be obtained from

mice derived completely from ES cells. Nagy et al. (1993) established ES cell lines from

crossing chinchilla 129 Sv females with agouti 129/Sv-CP males. One cell line, called R1,

was used to create an ES cell tetrapolid aggregate and was able to produce offspring that

were entirely ES cell derived (Nagy, Rossant, Nagy, Abramow-Newerly, & Roder, 1993).

This was validated by coat colour, which showed only agouti contribution and no

tetroploid cells, from albino mice, were found in the blood of the mice derived entirely

from ES cells (Nagy et al., 1993). Earlier passages of the R1 cell line (up to passage 14)

yielded more robust results. Even with permissive strains, only about 30% of the embryos

gave rise to stable mouse ES cell lines (Czechanski et al., 2014). Strains that are known to

be non-permissive include CBA, NOD, and DBA, however, protocols have started to

become available describing the derivation of mouse ES cell lines even from non-

permissive strains (Czechanski et al., 2014).

Human embryonic stem cells are obtained from the ICM by removing the outer

trophectoderm layer (Reubinoff, Pera, Fong, Trounson, & Bongso, 2000). Human

Page 15: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

5

embryonic stem cell lines were first obtained in 1998 by Thomson et al. from human

embryos, produced by in vitro fertilization, from the ICM of the embryos cultured until

the blastocyst stage (Thomson et al., 1998). The cells were able to differentiate in vitro

without the addition of mouse embryonic fibroblast feeder layers, both in the presence

and absence of human leukemia inhibitory factor (Thomson et al., 1998). The established

cell lines expressed surface markers stage-specific embryonic antigen (SSEA)-3 and

SSEA-4, which are not present on mouse ES cells, which express SSEA-1 (Thomson et

al., 1998). Other markers found on human ES cell lines include TRA-1-60, TRA-1-81,

and alkaline phosphatase (Thomson et al., 1998). The ES cell lines derived by Thomson

et al. were maintained in culture for 4 to 5 months (passages 14 to 16) and injected into

severe combined immunodeficient (SCID)-beige mice. Teratoma formation was noted in

these mice and differentiated cells from all three embryonic germ layers (endoderm,

mesoderm, and ectoderm) could be identified within the tumors (Thomson et al., 1998).

Differentiated tissues in the teratomas included cartilage, squamous epithelium, primitive

neuroectoderm, anaglionic structures, muscle, bone, and glandular epithelium (Reubinoff

et al., 2000). Stem cell lines from human blastocysts have been shown to be similar to

mouse ES cells derived from post-implantation mouse epiblast cells, referred to as EpiSC.

These mouse epiblast cells have been shown to be propagated using conditions used for

human ES cell culture (Tesar et al., 2007). Human ES cells are larger in size and grow as

a monolayer, and EpiSCs grow in a similar fashion, rather than exhibiting growth typical

of mouse ES cells; small, compact, and form domed colonies (Tesar et al., 2007).

Additionally, human ES cells lack a response to leukemia inhibitory factor and

differentiation of human ES cells is seen to occur rapidly, regardless if the cells are

Page 16: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

6

deprived of a feeder layer, even in the presence of LIF (Reubinoff et al., 2000). On the

other hand, for mouse ES cells, it has been shown that LIF is required for maintenance of

the cells in an undifferentiated state.

Page 17: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

7

Figure 1. Developmental potential of cells within an organism. The zygote, 2-cell stage,

and 4-cell stage are considered totipotent, whereas the blastocyst is considered

pluripotent and the embryo, fetus, infant, adult and elderly contain multipotent and

unipotent cells. (Mitalipov, S., & Wolf, D. (2009)).

Figure 1: Developmental Potential.

Page 18: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

8

1.1.2 Stem Cell Nomenclature

The use of the term stem cell dates back to the 19th century, where it was used

specifically in the context of the embryo (Maehle, 2011). Although the term stem cell is

widely applied in the literature today, there is a disparity in the scientific community as to

the definition and cell type used to characterize a true stem cell. Classically, the term is

used to define a cell which can divide and proliferate indefinitely without differentiation,

can form all three embryonic germ layers under appropriate stimulation and can also

repopulate a tissue in vivo. However, this nomenclature has not been vigorously applied.

For example, the term stem cell has been used for many decades when describing the

process of hematopoiesis, referring to a cell that can sustain the development of blood

cells, namely hematopoietic stem cells (HSCs) (Dykstra et al., 2007; Ramalho-Santos &

Willenbring, 2007). However, even though this small subpopulation of cells are termed

'stem cells', they are multi-potent, having the ability to differentiate into cells of the blood

system, rather than being pluripotent (Seita & Weissman, 2010). HSCs hence possess the

potential for both multi-potency as well as self-renewal. Consequently, they may also be

termed non-pluripotent stem cells, or progenitor cells, as the term progenitor cells is

assigned to blood stem cells which have started to differentiate into a lymphoid and

myeloid progenitor cell (Young et al., 2001). As a result, there is a difference between

lineage-committed progenitor stem cells and lineage-uncommitted pluripotent stem cells

(Young et al., 2001). Animals that have been lethally irradiated suffer from bone marrow

failure, however, injections of non-irradiated bone marrow cells are capable of

reconstituting the whole immune system, hence saving the lives of these mice (Spangrude,

Heimfeld, & Weissman, 1988). Overall, the term 'stem cell' has evolved and expanded

Page 19: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

9

from its original rigid definition, and now encompasses different stem cell populations,

such as totipotent stem cells termed "zygote", pluripotent stem cells such as induced

pluripotent stem cells and ES cells, and unipotent macrophage progenitors (Seita &

Weissman, 2010).

1.1.3 Tissue Specific Stem Cells

Most tissues are mainly composed of mature cell types which are terminally

differentiated, however also present is a small subpopulation of stem cells specific to

each tissue, such as haematopoietic, neural, gastrointestinal, epidermal, hepatic, and

mesenchymal stem cells (Jiang et al., 2002). The role of these tissue specific stem cells is

to act as a reservoir of replenishing cells that maintain the tissue. When compared with

ES cells, tissue specific stem cells have less self-renewal ability and pluripotency is not

exhibited, even though they differentiate into multiple lineages (Jiang et al., 2002). The

prototypical tissue specific stem cell, and also the best studied due to its early isolation, is

the hematopoietic stem cell mentioned earlier (Bryder, Rossi, & Weissman, 2006). Other

examples include mesenchymal stem cells, however, very few studies have shown that

isolated mesenchymal stem cell populations are true stem cells. Some have reported that

purification of cell populations from the bone marrow (BM) yields mesenchymal stem

cells as well as a subset of more immature cell types, exhibiting the capacity to

differentiate into cells of mesenchymal origin, visceral mesoderm, neuroectoderm and

endoderm (Jiang et al., 2002). Some have shown that there are clonal populations of stem

cells in the connective tissues of post-natal animals, where the clones consist of

pluripotent mesenchymal stem cells (Young et al., 2001). Moreover, others have reported

that when mice have been bone marrow ablated, donor mesenchymal stem cells first

Page 20: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

10

replace a portion of the mesenchymal stem cells in the bone marrow of the recipient mice

(Prockop, 1997). The results from this study suggest that the progeny of mesenchymal

stem cells acquire the phenotypes of different target tissues before they leave the marrow

or after they have entered the microenvironment of specific tissues. Overall, there seems

to be only a very limited number of studies that have been done to date which have

shown and proven the fact that there are stem cells present within isolated mesenchymal

stem cell populations. The method that is used for this task is limiting dilution techniques

for clonal analysis.

1.1.4 Mesenchymal Stem Cells

The concept of mesenchymal stem cells dates back to the 19th century, where it was

shown that ectopic bone, marrow, and fibrous tissue formed when bone marrow was

placed in mice, in a different tissue from that of origin (Bianco, Robey, & Simmons,

2008). The population of cells had osteogenic potential and hence were termed

osteogenic stem cells. In the 1960's and 1970's, Freidenstein and coworkers conducted a

set of experiments, which identified these BM stromal and osteogenic stem cells by

isolating fibroblast colonies from the BM and spleen of guinea pigs. When these cells

were placed in diffusion chambers in vitro, at the right density, bone formation was

observed (Friedenstein, Chailakhjan, & Lalykina, 1970). The fibroblasts were seen to

form discrete colonies derived from a single cell, and hence were called colony forming

unit-fibroblastoid cells, or CFU-F (Friedenstein et al., 1970). These experiments

established and solidified that there existed a non-hematopoietic cell population in the

BM, which supported the process of hematopoiesis, were able to differentiate to bone and

also form colonies derived from single cells in tissue culture (Friedenstein et al., 1970). In

Page 21: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

11

the 1990's, Arnold Caplan derived the term mesenchymal stem cells, and this

nomenclature is frequently used and engrained in the scientific literature today.

Nevertheless, data to support the identity of mesenchymal cells as true stem cells are

sparse and controversial. Clonal self-renewal and multi-lineage differential potential are

two classifications that encompass the definition of a "stem cell." In tissues, there are a

limited number of mesenchymal cells that are specifically stem cells, as a result, clonal

self-renewal has been difficult to prove with robust and reproducible experiments

(Sarugaser, Hanoun, Keating, Stanford, & Davies, 2009). Moreover, clonal populations

of cells have not been shown to be derived from a single cell. Another criterion

encompassing the term stem cell is the ability to differentiate to multiple lineages.

Mesenchymal stem cells have been shown to differentiate into the classical tri-lineage

pathway, however, differentiation into other cell types, such as skeletal muscle,

myocardium, and tendon has not yet been proven with robust evidence (Bianco et al.,

2008). Although some studies have illustrated the ability of mesenchymal stem cells to

differentiate into cells other than osteoblasts, chondrocytes and adipocytes, the capacity

of the cells to do this are not widely accepted by everyone in the scientific community

and are cited to be merely artefacts of culture conditions not exhibiting any functional

capacity. Lastly, due to the fact that bone and muscle are derived from different

progenitors in the developing foetus, there is uncertainty as to whether there is a common

post-natal mesenchymal progenitor cell (Nombela-Arrieta, Ritz, & Silberstein, 2011). As

a result, caution should be taken in regards to the nomenclature of these cells, more

specifically, when using the term stem cell to describe an isolated mesenchymal cell

population, as this may lead to misconceptions about their "stemness." The International

Page 22: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

12

Society for Cellular Therapy (ISCT) has suggested that even though the acronym MSCs

is used, the term stromal, rather than stem, should be applied (Keating, 2012). As a result,

the term "stromal" rather than "stem", will be used for this project, as a

heterogeneous population of cells were isolated and the true stem cell identity of this

population was not a focus of the study, and hence was not proven.

1.1.5 MSC Immunophenotype

Amongst confusion in the scientific community about which cells constitute

mesenchymal stromal cells (MSCs) and a lack of standardized criteria to define MSCs in

vitro, the ISCT has proposed a set of minimal criteria by which to phenotypically identify

mesenchymal stromal cells: (1) cells must adhere to plastic under standard culture

conditions, (2) must express CD105, CD73, and CD90, and lack expression of CD45,

CD34, CD14 or CD11b, CD79a or CD19, and HLA-DR, and (3) differentiate into

osteocytes, adipocytes and chondrocytes in vitro (Karp & Leng Teo, 2009; Lv, Tuan,

Cheung, & Leung, 2014). Unfortunately there is no single antigen which is MSC specific,

and which can be used to select for MSCs. The markers provided by the ISCT are

expressed on a variety of other cell types, especially blood cells. Surface antigen

characterization of expanded MSC cultures has shown the expression of CD44, CD71,

CD51, CD106, and STRO-1 (Chamberlain, Fox, Ashton, & Middleton, 2007; Phinney &

Prockop, 2007). However, different groups use various isolation methods and culture

conditions for MSCs, and as a result, the cells usually represent a heterogeneous

population, expressing different variations as well as levels of these markers. MSCs

isolated from different tissues and species do not all express the same molecules and may

have slightly different properties (Chamberlain et al., 2007). This may cause some

Page 23: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

13

variation in the immunophenotype stated by the ISCT. For example, MSCs isolated from

human adipose tissue initially express CD34, and this marker is also expressed on murine

MSCs (Meirelles Lda, Fontes, Covas, & Caplan, 2009). Discrepancies in marker

expression have also been shown in MSCs isolated from different compartments of the

bone marrow; MSCs isolated from endosteal or stromal niche in the bone marrow have

been shown to express Oct-4 and Nanog, which are both nuclear markers, and SSEA-4,

which is a surface marker widely used to identify embryonic stem cells (Bara, Richards,

Alini, & Stoddart, 2014). However, these markers have not been shown to be expressed

on MSCs obtained from perivascular locations (Bara et al., 2014). Overall, even though

there is a consensus regarding the cell surface identity of MSCs, cells have been shown to

be phenotypically heterogeneous and marker profiles of each isolated MSC population

should be studied individually.

1.2 In Vivo Identity

1.2.1 Pericytes

The in vivo identity of MSCs has not yet been fully elucidated and is still an area of

question and debate. In tissues, MSCs are a rare cell population present in low numbers,

thus making their study and identification difficult (Nombela-Arrieta et al., 2011). The

exact numbers and frequencies of MSCs in tissues are difficult to determine due to

various isolation methods and culture techniques, however, in the bone marrow it has

been estimated that MSCs are found at a frequency of 0.001%-0.01% of the total

nucleated cells, and hence are found 10-fold less than HSCs (Bernardo, Locatelli, &

Fibbe, 2009). It has also been shown that especially for the BM, the frequency of MSCs

declines with age, from 1/10,000 nucleated marrow cells in a newborn to about

Page 24: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

14

1/1,000,000 nucleated marrow cells in an 80-year-old-person (Bernardo et al., 2009).

This has also been shown for adipose tissue derived mesenchymal stem cells (Alt et al.,

2012). However, there have been several hypotheses as to the presumed in vivo identity

of MSCs. One of these theories postulates that MSCs in vivo may be derived from

pericytes (Lv et al., 2014). There are many reports describing a perivascular niche for

MSCs, where the cells have been proposed to lie on the basement membrane opposed to

endothelial cells (da Silva Meirelles, Caplan, & Nardi, 2008; Lv et al., 2014). This

observation is supported by the fact that MSCs are usually isolated from arterial or

venous walls (da Silva Meirelles et al., 2008). However, MSCs have also been obtained

from capillaries, post-capillary venules, arteries, and veins, whereas "pericyte" refers only

to cells in capillaries or post-capillary venules (Nombela-Arrieta et al., 2011).

Nevertheless, pericytes have been shown to display features similar to those exhibited by

MSCs, expressing the same surface antigens and having multi-lineage differentiation

potential into cells of mesenchymal origin, specifically osteocytes, chondrocytes and

adipocytes. Other hypotheses that have arisen are that pericytes represent an ancestor cell

of MSCs or that they represent a distinct MSC cell subset (Bara et al., 2014). It should be

noted that even though several cell characteristics can be obtained from in vitro cultures,

the results obtained should not be extrapolated to the in vivo identity of the cells, as the

properties of MSCs may be altered by various culture conditions. Isolation of cells from

the same source has yielded varying results in regards to phenotype, proliferation, and

differentiation capabilities. Hence in vitro characteristics of MSCs may only be induced

by specific culture conditions rather than being a direct reflection of their true in vivo

identity.

Page 25: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

15

1.2.2 Neural Crest Cells

Another developmental origin of MSCs has been suggested to be the neural crest. In the

embryonic lineage, neural crest derived from the ectoderm gives rise to an astonishing

array of cells and tissues, including vascular smooth muscle cells in the central nervous

system as well as the thymus, whereas mural cells found in the coleomic organs,

specifically the gut, lung, and liver, are all derived from the mesoderm and mesothelium

(Armulik, Genove, & Betsholtz, 2011). This indicates a process whereby mesothelial

cells undergo an epithelial-to-mesenchymal-transition, delaminate, and migrate into the

organs to produce mesenchymal components, including fibroblasts, vascular smooth

muscle cells, and pericytes (Armulik et al., 2011). The hypothesis that MSCs arise from

the neural crest originates from the observation that MSCs have been shown to

differentiate to neuronal and glial cells. In the developing embryo, the neural crest arises

as a multipotent stem cell population, which forms at the interface between the

neuroepithelium and the future epidermis of the developing embryo (Mayor & Theveneau,

2013). Neural crest cells are derived from the ectoderm, and it has been shown that the

cells are able to form and contribute to many different cell types, which can be sub-

divided into four main categories (Gilbert, 2000). The cranial neural crest cells

differentiate into cartilage, bone, glia, and connective tissue of the face, the trunk neural

crest cells differentiate into either pigment synthesizing melanocytes, or sympathetic

ganglia, the adrenal medulla, and the nerves of the aorta (Gilbert, 2000). The vagal and

sacral neural crest cells form the parasympathetic ganglia of the gut, and finally, the

cardiac neural crest gives rise to the melanocytes, neurons, cartilage, and connective

tissues, as well as the entire musculoconnective tissue wall of the heart (Gilbert, 2000).

Markers that are used to uniquely identify neural crest cells, such as Twist, p75NTR,

Page 26: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

16

Snail1, Snail2, Sox9, and Mpz, were also found on isolated murine MSCs (Morikawa et

al., 2009). This suggests that MSCs may have a neural crest origin. Another marker,

called nestin, has been used to group MSCs into two cell populations, each exhibiting

different developmental fates. MSCs lacking nestin expression, found within the

developing bone marrow of long bones, contribute to the developing bones of the fetus,

and soon after lose MSC identity (Isern et al., 2014). Nestin positive MSCs, identified by

GFP expression under the control of the nestin promoter, help maintain the hematopoietic

niche of the perinatal bone marrow and do not participate in osteochondral development

in the fetus. These cells have also been shown to originate from the neural crest (Isern et

al., 2014).

1.3 Sources of Human Mesenchymal Stromal Cells

1.3.1 Embryonic Tissue

It has been proposed that MSCs can be isolated from fetal and most postnatal tissues.

MSCs have classically been obtained from the BM, however, this procedure is invasive,

can lead to infections and incur pain, hence alternative sources of MSCs have been

investigated, such as fetal tissues or the umbilical cord (UC). Embryonic MSCs have

been shown to originate mainly from the neuroepithelium as well as the neural crest,

appearing by mid-gestation (Hu et al., 2003; Uccelli, Moretta, & Pistoia, 2008). First

trimester fetal MSCs have been derived from liver, blood, as well as the bone marrow and

it has been shown that these cells exhibit similar characteristics to adult MSCs

(Campagnoli et al., 2001). The morphologic, immunophenotypic, and functional

characteristics of fetal derived MSCs are similar to those found in adult tissues

(Campagnoli et al., 2001). However, others have suggested that the differentiation

Page 27: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

17

capacity of fetal derived MSCs differs based on the tissue source. MSCs isolated from 20

week foetuses showed that while bone marrow, liver, lung, and spleen derived MSCs

were able to differentiate into adipocytes, MSCs isolated from the spleen had a lower

potential of differentiation into this cell type, while the bone marrow, lung and liver

showed a significantly higher osteogenic differentiation (in 't Anker et al., 2003). Second

trimester fetal MSCs have also been obtained from the fetal lung (in 't Anker et al., 2003).

In addition, MSCs have been cultured from fetal pancreas, which have been shown to be

able form CFU-F (Hu et al., 2003). MSCs isolated from fetal pancreatic tissue mainly

express CD44, CD29, and CD13, but not HLA-DR or von Willebrand factor (vWF) (Hu

et al., 2003). These cells are able to differentiate into the tri-lineage pathway (Hu et al.,

2003). Single cell suspensions of MSCs derived from the lung, liver, spleen and bone

marrow from week 20 foetuses express CD90, CD105, CD166, and CD73, and are

negative for hematopoietic markers CD45, CD14, and CD31 (in 't Anker et al., 2003).

Overall, fetal derived MSCs have been proven to express the classical MSC marker

profile and show tri-lineage differentiation. Varying results among some studies in

regards to differentiation potential suggests differences in isolation or culturing methods

from these sources.

1.3.2 Adult Tissue

Bone Marrow

Adult sources of MSCs are mostly commonly isolated from the bone marrow, which

were initially identified by Freidenstein and are the most common studied cell source. In

the bone marrow, MSCs are found in the stromal compartment, where they support

haematopoiesis. Another function of BM-MSCs has been suggested to be related to the

Page 28: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

18

development, stabilization, and maintenance of the sinusoidal network in the BM (Bianco

et al., 2008). BM-MSCs have also been shown to be committed, but not differentiated,

osteogenic progenitors (Bianco et al., 2008). MSCs are isolated usually by density

gradient centrifugation, to remove unwanted cell types such as hematopoietic cells, from

bone marrow aspirates obtained from the iliac crest. MSCs have been shown to have a

characteristic fibroblast morphology when cultured, expressing alpha-smooth actin,

CD105 (SH2), CD73 (SH3), and SH4, CD106, CD120a, VCAM-1, vWF, cytokeratins,

and extracellular matrix proteins such as fibronectin, vimentin, collegen I and collagen IV,

and are negative for CD1a, CD14, CD31, CD45, and CD56 (Conget & Minguell, 1999;

Pittenger et al., 1999). BM-MSCs are able to differentiate into chondrogenic, adipogenic

and osteogenic lineages under specific culture conditions (Pittenger et al., 1999).

However, cells isolated from the bone marrow have a low cell yield, and the older age of

volunteers are not ideal (Choudhery et al., 2012). Although the effects of advanced age

on MSCs have been controversial, previous work suggests that cell populations undergo

changes when obtained from donors of advanced age, as the expression of surface makers,

CD44, CD90, CD105, and Stro-1 were found to undergo age-related changes (Stolzing,

Jones, McGonagle, & Scutt, 2008). Overall it has been shown that there are various age

related changes to MSCs when obtained from the bone marrow. Furthermore, there is

only a small frequency of MSCs present within the bone marrow, with reports suggesting

that only about 0.001 to 0.01% of cells isolated are MSCs (Pittenger et al., 1999).

Adipose Tissue

Another source of MSCs has been adipose tissue, which has been suggested to be

superior to bone marrow, due to the fact that it is a more convenient source of cells, as the

Page 29: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

19

tissue can be harvested in large amounts, with a less invasive procedure (Choudhery et al.,

2012; Fraser, Wulur, Alfonso, & Hedrick, 2006). More specifically, three types of

surgical procedures are used for adipose tissue harvesting; surgical resection, tumescent,

or conventional liposuction and ultrasound assisted liposuction (Oedayrajsingh-Varma et

al., 2006). Cells are usually obtained from the tissue by enzymatic digestion with

collagenase (Hass, Kasper, Bohm, & Jacobs, 2011). It has been reported that all

procedures result in little patient discomfort and low donor site morbidity

(Oedayrajsingh-Varma et al., 2006). The most common source for adipose tissue is the

abdomen or thigh regions, due to the abundance of subcutaneous adipose tissue and high

yield of cells (Schreml et al., 2009). Some studies have suggested that there is a higher

yield of MSCs from the abdomen than from the hip and thigh regions, whereas others

have stated approximately equal cell yield from the lower abdomen and inner thigh

(Schreml et al., 2009). Another harvesting site which can be used, although not as

common, is the infrapatellor Hoffa's fat pad (Schreml et al., 2009). The MSCs isolated

from adipose tissue, most often referred to as processed lipoaspirate (LPA) cells or

adipose derived stem cells, are similar to those obtained from bone marrow, however

some differences exist between the two cell populations. BM-MSCs as well as those

obtained from adipose tissue both exhibit homogeneity in cell morphology, size, as well

as granularity (Choudhery et al., 2012; De Ugarte et al., 2003). It has been shown that

LPA cells express surface markers CD13, CD29, CD44, CD58, CD90, CD105, and

CD166, and do not demonstrate expression of the epitopes CD14, CD19, CD31, and

CD45 (De Ugarte et al., 2003). Several studies have suggested that CD106 (VCAM-1) is

found to be expressed on BM-MSCs but not on LPA cells (De Ugarte et al., 2003; Hass et

Page 30: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

20

al., 2011). Differences in expression of other markers, such as CD49d have also been

noted (De Ugarte et al., 2003). Several studies have demonstrated the ability of the cells

to undergo adipogenic, chondrogenic, osteogenic, as well as myogenic differentiation

(Choudhery et al., 2012; Fraser et al., 2006; Rodriguez, Elabd, Amri, Ailhaud, & Dani,

2005). In comparison to other cell sources, MSCs obtained from adipose tissue have been

shown to differentiate into more mature and larger adipocytes (Choudhery et al., 2012).

LPA cells have been shown to have a higher population doubling time as well as exhibit a

lower percentage of cells undergoing senescence during earlier passages when compared

with BM-MSCs (Kern, Eichler, Stoeve, Kluter, & Bieback, 2006). Many studies indicate

that adipose tissue can be used as an alternative and valuable source of MSCs.

Other sources

Other sources of adult derived MSCs include peripheral blood, dental pulp, synovium,

skin and skeletal muscle.

1.3.3 Birth Associated Tissues

Umbilical Cord Blood

Umbilical cord blood (UCB) is a proven source of hematopoietic stem cells, and has been

shown to be a possible source of MSCs; however data regarding the isolation of MSCs

has been controversial. This is mainly due to difficulties in isolating MSCs from UCB

due to their low frequency. Nevertheless, UCB is an attractive source of MSCs due to the

fact that the isolation procedure is painless and there is no harm to the mother or infant.

UCB-MSCs are also a more immature cell type than those obtained from the bone

marrow. Cells are usually obtained by venous puncture of the umbilical vein. UCB-MSCs

Page 31: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

21

show a similar surface phenotype to BM-MSCs, being positive for CD29, CD49b, CD44,

CD58, and CD105 and negative for cell surface epitopes of the hematopoietic lineage

CD31, CD45, CD19, CD34, and HLA-DR (O. K. Lee et al., 2004). However, a study

done by Lee et al. (2004) found that the cells were negative for CD90, which has been

shown to be expressed by cells from the BM as well as adipose tissue. Consistent with

bone marrow and adipose tissue derived MSCs, some studies have shown UCB-MSCs to

be able to differentiate into osteoblasts, adipocytes and chondrocytes (W. Wagner et al.,

2005). However, others have proven UCB-MSCs to differentiate into osteoblasts,

adipocytes, and cells of the neural lineage, but not chondrocytes (Goodwin et al., 2001).

Others have suggested differentiation only into cells of the chondrogenic and neurogenic

lineages (Bieback, Kern, Kluter, & Eichler, 2004) Nevertheless, MSC isolation from

UCB is a laborious process, time consuming, and results in low cell yield. In comparison

to bone marrow and adipose tissue, which illustrate an isolation efficiency of 100%, UCB

has an isolation efficiency around 30%, as well as slower growth rate (Rebelatto et al.,

2008). Several studies have examined procedures that can be used to obtain a greater cell

yield, such as ensuring a storage time of less than 15 hours, a net volume of more than 33

ml of blood, and a mononuclear cell count greater than 1 x 108 of mononuclear cells, and

no signs of coagulation or hemolysis (Bieback et al., 2004).

Another cell type that has been isolated from umbilical cord blood and exhibits

similarities to MSCs are multi-potential stem cells (MPSCs). Rogers et al. has shown that

upon isolation, these cells are able to adhere to plastic, are CD45 and CD34-positive, and

are a distinct cell type from mesenchymal cells as well as hematopoietic cells (Rogers et

al., 2007). Additionally, unlike other cell types, MPSCs can be derived from 100% of

Page 32: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

22

cord blood samples (Rogers et al., 2007). These cells possess mesenchymal properties,

expressing CD90, CD105, and CD73 on the cell surface (Rogers et al., 2007). MPSCs are

induced to form after 8 days in culture in media supplemented with fibroblast growth

factor 4 (FGF-4), stem cell factor (SCF) and Flt3 ligand (Rogers et al., 2007). Positive

therapeutic potential of MPSCs have been illustrated in a mouse hind limb ischemia

model, whereby the cells were documented to release many paracrine factors and

differentiate in vivo into endothelial cells, smooth muscle cells, as well as striated muscle

cells (Whiteley et al., 2014). Other models, such as a rat spinal cord injury model, has

demonstrated that the therapeutic properties of the cells can be attributed to the secretion

of cytokines and chemokines that possess anti-inflammatory, neuroprotective and

angiogeneic properties, hence facilitating the endogenous repair processes (Chua et al.,

2010). The cells were also seen to be present one week after transplantation (Chua et al.,

2010). Overall, it has been shown that MPSCs are a stable and reproducible cell

population that has shown to have therapeutic potential and seem to resemble MSCs,

however, unlike MSCs are a result of cell culture (Whiteley et al., 2014).

Umbilical Cord Tissue

The umbilical cord is a vehicle for the transport of blood from the placenta to the foetus

and vice versa. It consists of two arteries and one vein that are all surrounded by the

Wharton's jelly and a layer of amnion. Umbilical cord tissue is another source that

harbours MSCs. This source is attractive due to the fact that its collection is non-invasive

and the tissue is usually discarded as medical waste. UC-MSCs have a low

immunogenicity and do not have as many bioethical concerns encompassing their use

(Han et al., 2013). This is due to the fact that MSCs express low levels of major

histocompatibility complex (MHC) class I, and are negative for MHC class II (Ankrum,

Page 33: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

23

Ong, & Karp, 2014). However, some studies have also shown that under certain culture

conditions, MSCs are immunogenic and can stimulate the humoral and cellular immune

response. These MSCs demonstrate increased expression of MHC class I and MHC class

II when exposed to interferon gamma (IFN-γ) (Ankrum et al., 2014). The umbilical cord

stroma, known as the Wharton's jelly, is a connective tissue composed of mainly

glycosylaminoglycans (GAGs), and it is most often the source used to isolate cells (Secco

et al., 2008). Additionally, MSCs can also be isolated from the perivascular region of the

umbilical cord. For this isolation method, the epithelium is removed to expose the matrix

underneath, after which the vessels, with the intact extracellular matrix, are removed and

the cells are harvested using enzymatic digestion with collagenase (Sarugaser et al.,

2009). These cells as referred to as human umbilical cord perivascular cells (HUCPVCs).

MSCs can be extracted using the explant or enzymatic digestion techniques, although the

latter is the conventional method. Vessels are stripped out of the cord and cut into small

pieces (around 2 cm). The ends are tied together to form a circle with the outside of the

vessel exposed. Everything is digested with 0.1% collagenase IV or collagenase II, at

37 °C for 16–18 h (Han et al., 2013). The cells are then harvested at 37°C at 5 % CO2.

This method prevents the endothelial cells from being released during the digestion

process.

In comparison to the enzymatic digestion method, the explant isolation method does not

involve the use of collagenase and hence is more cost effective. This method also allows

for easier clinical approval. This is due to the fact that collagenase, used in the enzymatic

digestion protocol, must be made for clinical use and must also undergo extensive testing.

The explant method involves draining the cord of blood, washing it and cutting the cord

Page 34: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

24

into 4-6 mm thick sections. These are further divided into 3-4 smaller pieces and plated

onto culture dishes after which medium, such as Dulbecco's modified eagle's medium

(DMEM) or alpha-MEM (alpha minimum essential medium), supplemented with 10%

fetal bovine or calf serum and antibiotics, is added and cultured under 5 % CO2 at 37 °C

(Han et al., 2013). After a varying period of time, cells become attached to the culture

dish. Viable cells have been obtained using both methods, however some differences

between the two isolation techniques have been noticed. When compared with enzymatic

digestion, explant culture has shown to have a lower MSC yield per tissue gram and has

shown to require more days in primary culture until the first cell harvest (Gittel et al.,

2013). For example, equine UC-MSCs have illustrated 10 days in primary culture by

digestion method as compared to 18 days in explant culture (Gittel et al., 2013). However,

the results from our study has shown that cells can be seen in as little as five days after

explant. The tissue explant method also has been cited to have a longer culture cycle and

yield a lower number of primary cells per centimetre of umbilical cord (Han et al., 2013).

However, the explant culture method has several advantages over enzymatic digestion, as

it prevents cellular damage, it is more economical, and cells isolated using this method

have been suggested to release higher amounts of certain growth factors, such as basic

fibroblast growth factor (FGF basic) (Yoon et al., 2013). Additionally, adherence and

proliferation of cells after sub-culturing has also been shown to be more efficient (Han et

al., 2013).

MSCs are present in higher frequencies in the umbilical cord tissue when compared with

other sources such as the bone marrow or peripheral blood. Additionally, UC-MSCs have

a higher proliferation as well as expansion potential when compared with MSCs derived

Page 35: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

25

from adult tissues, perhaps due to the fact that they are a more immature cell type

(Trivanovic et al., 2013; H. S. Wang et al., 2004). MSCs obtained from the umbilical

cord express CD44, CD105, CD105 (SH2), CD73 (SH3), but not markers indicative of

blood cells, such as CD45, CD34, CD19, CD11b, and CD14. When cultured with specific

induction medium, they have the potential to differentiate into adipocytes, chondrocytes,

as well as osteoblasts (Huang et al., 2013). It has been suggested that MSCs from donors

of various ages differ in regards to their proliferation as well as clonogenicity,

specifically that cells from younger mothers have a higher proliferative potential as well

as a greater osteogenic differentiation (Huang et al., 2013).

Other Sources

MSC isolation has also been reported from various other birth associated tissues, such as

the placenta and amniotic fluid, however these sources are not as common as the

umbilical cord blood or the umbilical cord tissue itself.

1.4 Immunomodulation

1.4.1 Innate Immunity

The innate immunity is a first-line of defence response against microorganisms and

infections. MSCs are able to modulate the immune system, and the nature of this

modulation is dependent on the cellular and inflammatory environment (Le Blanc &

Davies, 2015). During the early phases of infection, MSCs exhibit predominantly pro-

inflammatory effects due to exposure to Toll-like receptor (TLR) 2 and TLR4. Activated

MSCs migrate to the site of injury and secret various chemokines, such as the chemokine

(C-X-C motif) ligand (CXCL) 9, CXCL10, macrophage inflammatory protein (MIP)-1α

Page 36: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

26

and MIP-1β (Le Blanc & Davies, 2015). The expression of Toll-like receptors can prime

MSCs; TLR4 has been shown to cause secretion of pro-inflammatory cytokines by MSCs

and TLR3 primed MSCs are able to exert immune suppressive functions (Waterman,

Tomchuck, Henkle, & Betancourt, 2010). MSCs can also suppress immune activation.

This occurs when the cells are exposed to pro-inflammatory cytokines such as IFN-γ and

tumour necrosis factor alpha (TNF-α) (Le Blanc & Davies, 2015). They do not express

MHC class II on their surface and have low or intermediate expression of MHC class I,

hence they are considered to be immune privileged cells and can be infused into

autologous or allogeneic hosts. The mechanism of action has been cited to be

multifactorial, dependent on both soluble factor secretion, as well as cell-to-cell contact

(Waterman et al., 2010). In vitro, MSCs can suppress lymphocyte alloreactivity in mixed

lymphocyte cultures (Le Blanc & Ringden, 2007). MSCs are able to inhibit the growth of

monocytes towards dendritic cells (DC), which are known to accumulate in inflamed

tissues and are considered antigen presenting cells (APCs) (Moretta, 2002). When DCs

are cultured with BM-MSCs, the immune cells are not able to stimulate CD4+ T cell

proliferation, and MSCs can also alter the cytokine secretion profile of DCs (English,

French, & Wood, 2010). MSCs are able to suppress TNF-α secretion by DCs, hence

attenuating immune responses (Aggarwal & Pittenger, 2005). Additionally, the

expression of several DC surface receptors, which are responsible for natural killer (NK)

cell activation and cell killing, can be down regulated by MSCs, such as NKp30 and

natural-killer group 2, member D (Uccelli et al., 2008). MSCs have also been shown to

impact other effector cells of the innate immune system, such as NK cells. These cells

play a role in controlling the spread of some tumours and also microbial infections by the

Page 37: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

27

production of various pro-inflammatory cytokines (Vivier, Tomasello, Baratin, Walzer, &

Ugolini, 2008). Similar to their action on DCs, MSCs can abolish the secretion of IFN-γ

by NK cells that have been stimulated by IL-2 (Le Blanc & Ringden, 2007). MSCs can

prevent the proliferation of resting NK cells, however only a partial inhibition of

proliferation was seen when MSCs were cultured with activated NK cells (which have

been incubated with IL-2 for more than 7 days). This may be due to MSC surface

expression of ligands recognized by different activating NK cell receptors, such as

DNAM-1, NKG2D, and well as low levels of HLA class I (Le Blanc & Ringden, 2007).

Under normal conditions, the expression of HLA class I molecules on the surface of

autologous cells prevents NK cell activation due to interaction with a specific set of

inhibitory receptors present on the surface of NK cells (Spaggiari, Capobianco, Becchetti,

Mingari, & Moretta, 2006). Low levels of MHC class I on MSCs can induce autologous

and allogenic NK cell mediated cytotoxicity lysis of MSCs, under inflammatory

conditions where NK cells are exposed to IL-2 (Spaggiari et al., 2006). Hence, this

suggests that inhibitory interactions as a result of HLA class I on MSCs are not sufficient

to protect MSCs from lysis (Spaggiari et al., 2006). However, this is countered by IFN-γ,

a cytokine released by NK cells, which has been shown to be able to up-regulate HLA

class I molecules on MSCs, thus rendering the cells resistant to NK-mediated lysis

(Spaggiari et al., 2006). The up-regulation of HLA class I molecules was also seen to

decrease cytokine production by NK cells. Since this study illustrated that NK cells are

able to lyse MSCs, this raised the question of why MSCs are not killed by NK cells in

vivo. It has been suggested that in vivo, there may be MSCs localized in specific tissue

niches, expressing higher levels of MHC class I, or that NK cells might not reach an

Page 38: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

28

activation state sufficient for MSC lysis (Spaggiari et al., 2006). This data could be useful

for therapies using MSCs, such as suppression of graft-versus-host (GvHD) in BM

transplants.

1.4.2 Adaptive Immunity

The adaptive immunity is a second line of defence against pathogens and is activated

once the innate immunity has already taken effect. MSCs are able to regulate the adaptive

immune system through direct cell-to-cell interaction as well as through soluble factor

secretion, however, it is not know which method is more prevalent. MSCs are able to

attenuate the activation of the immune system by inhibiting T lymphocyte cell

proliferation. In vitro, the addition of autologous BM-MSCs are able to inhibit T-cell

proliferation, even after stimulation with IL-2 (Di Nicola et al., 2002). This reaction was

dose dependent; T lymphocytes and irradiated allogenic dendritic cells were cultured at a

ratio of 1:1, and increasing amounts of irradiated BM-MSCs were added. T lymphocyte

proliferation was seen to be optimal at a ratio of 1:5 (Di Nicola et al., 2002). This

inhibition of T lymphocyte proliferation by BM-MSCs was reversible. Stimulated T cells

cultured with irradiated BM-MSCs for 7 days resulted in a reduced T lymphocyte

proliferation, however when the T lymphocytes were re-timulated (by at addition of

dendritic cells or IL-2) for two days after this time period of inhibition, this restimulation

lead to a proliferation of the T lymphocytes that was comparable with control cultures

without BM-MSCs (Di Nicola et al., 2002). MSCs can also decrease the amount of

cytokines produced by T-cells, namely IFN-γ, IL-2, IL-4 and TNF-α, which are

associated with inflammatory processes (Ghannam, Bouffi, Djouad, Jorgensen, & Noel,

2010). Proliferation of CD4+ and CD8+ T-cells stimulated by peripheral blood

Page 39: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

29

leukocytes (PBLs) or DCs is inhibited by MSCs (Di Nicola et al., 2002). It has been

suggested that MSCs are able to inhibit T-cell receptor dependent and independent

proliferation, along with suppression of IFN-γ and TNF-α production, thus inducing

peripheral T-cell tolerance (Zappia et al., 2005). This is hypothesized to occur through

the regulation of the NF-kB signalling pathway and also by stopping the T-cell cycle in

the G0/G1 phase (Keating, 2012). Many factors have been suggested to be involved in T-

cell suppression by MSCs, namely transforming growth factor-β (TGF-β), hepatocyte

growth factor (HGF), indoleamine 2,3-dioxygenase (IDO), and prostaglandin E2 (PGE-2)

(Sato et al., 2007). However, there are species differences in the immune modulation

activity of MSCs, as IDO, a tryptophan catabolizing enzyme, acts in humans as well as

Rhesus monkeys, whereas nitric oxide is mainly involved in mice (Keating, 2012).

For humans, the immune suppression activity of MSCs has been compared to the

mechanism used by professional antigen-presenting cells in inhibiting T-cell responses to

autoantigens and fetal alloantigens (such as preventing the rejection of the fetus during

pregnancy) in vivo (Terness et al., 2002). IDO is strongly induced at the level of

transcription by IFN-γ along with other proinflammtory cytokines, thus causing the

conversion of tryptophan to kynurenine by the enzyme (Meisel et al., 2004). Tryptophan

is an essential amino acid, needed for the biosynthesis of important proteins (Terness et

al., 2002). T-cells are known to be sensitive to tryptophan depletion, and at low

tryptophan concentrations, cell cycle is arrested at the G1 cell cycle point, as T cells

possess a specific cell cycle regulatory checkpoint that has been shown to be sensitive to

tryptophan concentrations in tissue microenvironments (Mellor & Munn, 1999; Terness

et al., 2002). MSCs do not constitutively express IDO, however, the protein can be

Page 40: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

30

detected by Western blot analysis after stimulation with INF-γ (Meisel et al., 2004).

MSCs and mixed lymphocyte reaction co-cultures suggest that MSCs are the primary

source of IDO activity. This is further supported by the fact the addition of tryptophan is

able to restore T cell proliferation (Meisel et al., 2004).

1.5 Wound Healing

1.5.1 Tissue Repair

MSCs hold great promise for clinical applications and in regenerative medicine due to

their therapeutic abilities. In vivo, MSCs are involved in many processes, such as cellular

homeostasis, aging, tissue damage as well as inflammatory diseases (Ma et al., 2014).

There have been clinical trials done to assess the ability of MSCs in various pathological

conditions mostly using BM-MSCs. The first clinical trial in which the cells were tested

for their therapeutic effects was for children who had undergone allogenic BM

transplantation for severe osteogenesis imperfecta, characterized by defective type I

collagen production affecting tissues such as bone and ligament, where a significant

improvement in bone structure and function was seen in these patients after the

administration of MSCs (Horwitz et al., 1999). Since this time, there have been more than

300 patients that have received systemically infused MSCs for various indications

(Horwitz & Dominici, 2008). Moreover, many clinical trials are currently being done to

investigate the efficacy of MSC therapy for hematological pathologies such as aplastic

anemia, cardiovascular diseases such as heart disease and vascular disease, and

neurological and inherited disorders such as Hurler syndrome (Giordano, Galderisi, &

Marino, 2007). MSCs have also been used for cutaneous wound healing. The main

mechanisms by which MSCs exert their therapeutic properties is by the secretion of

Page 41: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

31

various growth factors and cytokines, such as IL-6, IL-7, and granulocyte-macrophage

colony-stimulating factor (GM-CSF) (Deans & Moseley, 2000). There are four published

clinical studies which use MSCs in cutaneous wound healing (Isakson, de Blacam,

Whelan, McArdle, & Clover, 2015). Both autologous as well as allogenic cells have been

used in these studies (Isakson et al., 2015). For example, Badiavas et al. used BM cells

obtained from the iliac crest and applied the aspirate directly to the wound as well as

injected a small amount into the edges of the wound. Among the three patients studied, a

reduction in wound size was noticed, with improved thickness, vascularity, and integrity

of the dermis, greater generation of granulation tissue and epithelization, and after two

years, complete wound closure (Badiavas & Falanga, 2003). Falanga et al. used a fibrin

spray to deliver BM-MSCs to acute surgical wounds. Histological analysis of the wounds

suggested that some MSCs were able to migrate into the upper layers of the wound bed

and differentiate into fibroblastic cells (Falanga et al., 2007). This same study

administered BM-MSCs to diabetic wounds and noticed a decrease in the size of the

wound at 16 weeks after three topical applications of MSCs (Isakson et al., 2015). Others

have applied autologous BM-MSCs to diabetic foot ulcer patients. Dash et al.

administered one million MSCs per cm of wound area with a syringe into the ischemic

limb and along the edges of the wound (Dash, Dash, Routray, Mohapatra, & Mohapatra,

2009). When compared with controls, which received only standard wound care regimens,

patients who also concurrently received the cells had a reduced ulcer size (71% when

compared to only 23% in the control group), experienced an increase in pain-free walking

distance (7.5 fold increase when compared to 2.2 fold increase during the 12 week follow

up) and an increase in the cellularity of the wound (Dash et al., 2009). Biopsy analysis

Page 42: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

32

revealed reticulin fibers, indicating that BM-MSCs were able to contribute to dermal

rebuilding and closure of the non-healing chronic wound (Dash et al., 2009). Overall,

these clinical trials have shown that MSCs are able to accelerate wound closure for

chronic non-healing wounds where conventional treatments have failed.

1.5.2 Diabetic Wound Healing

Epidermal wound healing is classically subdivided into four steps which usually follow a

sequential progression of overlapping events in healthy individuals; hemostasis,

inflammation, proliferation and migration, and resolution and remodelling (Figure 2)

(Sonnemann & Bement, 2011). Various types of immune cells are recruited and

implicated in the wound healing process, namely platelets, which are responsible for the

formation of a platelet plug, neutrophils followed by macrophages, which both contribute

to scar formation, keratinocytes, which migrate over the injured dermis, fibroblasts and

finally myofibroblasts, which interact and produce extracellular matrix, mainly in the

form of collagen, which helps form the mature scar (Gurtner, Werner, Barrandon, &

Longaker, 2008). Hemostasis is the process where further blood loss is prevented due to

constriction of the damaged blood vessels and platelet clot formation (Sonnemann &

Bement, 2011). Also, the formation of a network of fibrin fibrils is initiated as fibrinogen

cleaves thrombin (Sonnemann & Bement, 2011). This allows growth factors to bind and

promote the inflammatory stage of wound healing (Sonnemann & Bement, 2011).

Inflammation is characterized by the recruitment of leukocytes to the wound. The first

leukocytes that arrive are neutrophils, which engulf bacteria, and then monocytes, which

differentiate into macrophages and remove debris as well as neutrophils which have died

(Sonnemann & Bement, 2011). This stage is also characterized by angiogenesis, whereby

Page 43: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

33

new vessels are formed, thus supplying oxygen and nutrients to the wound lesion

(Sonnemann & Bement, 2011). The next stage, proliferation and migration, is

characterized by epidermal cells and fibroblasts (Sonnemann & Bement, 2011).

Epidermal cells migrate over the wound site by assuming a more flattened, elongated

morphology, and also recruit fibroblasts, which form granulation tissue (Sonnemann &

Bement, 2011). The new epidermis moves over the wound area as a coherent sheet, due

to cell to cell contact that is maintained between the epidermal cells (Sonnemann &

Bement, 2011). Fibroblast cells are able to differentiate into myofibroblasts, due to

growth factors in the wound environment. These cells help bring the edges of the wound

closer together. Both fibroblasts and myofibroblasts secrete a large amount of collagen

along with other extracellular matrix proteins, hence forming the basis of new granulation

tissue (Sonnemann & Bement, 2011). Myofibroblasts also align the collagen fibrils that

make up the extracellular matrix by bringing the edges of the wound closer together

(Sonnemann & Bement, 2011). Lastly, the final phase of wound healing is resolution and

remodeling, whereby the following events take place: the edges of the migrating sheet of

new skin make contact with each other and epidermal proliferation as well as migration

stops, leukocytes in the wound leave or undergo programmed cell death, the extracellular

matrix is remodelled, and granulation tissue is removed (Sonnemann & Bement, 2011).

At the end of this phase, only the scar tissue remains, which is composed of the aligned

extracellular matrix filaments (Sonnemann & Bement, 2011). However, there can be

interruptions of this orderly sequence of events, thus leading to delayed wound healing or

even wounds that do not heal at all. Chronic wounds do not follow this orderly sequence

of events, and are often characterized by a prolonged state of inflammation (Guo &

Page 44: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

34

Dipietro, 2010). Most chronic wounds are ulcers that are associated with ischemia,

diabetes mellitus, or pressure (Guo & Dipietro, 2010). Diabetes associated wounds

represent a serious problem as they are a major cause of hospitalizations and may lead to

amputations. There are 150 million people with diabetes world-wide, and 15% of those

suffer from foot ulcerations, which often become non-healing chronic wounds (Wu,

Driver, Wrobel, & Armstrong, 2007). The mechanisms of wound healing are altered in

those with diabetes and such impairments are further magnified by insults such as

bacterial infections, tissue ischemia, trauma and poor management, all which can

aggravate the wound and cause diabetic foot ulcers to heal slowly, hence transforming

into chronic wounds (Jeffcoate & Harding, 2003). Epidermis from chronic ulcers have

shown to be very thick, hyperproliferative, and containing mitotically active cells in

suprabasal layers, whereas mitosis occurs only in the basal layers of the skin from normal

volunteers (Stojadinovic et al., 2005). The pathogenic triad are the three main factors

responsible for chronic wound formation in those with diabetes; namely neuropathy,

ischemia, and trauma (Falanga, 2005). Cells and cytokines that are essential for wound

healing to take place are altered in people with diabetes, for example it has been shown

that there is a diminished response of keratinocytes. At the non-healing edge of diabetic

foot ulcers, keratinocytes show very little migration, hyperproliferation, as well as

incomplete differentiation (Brem & Tomic-Canic, 2007). There is also a reduced capacity

of the endothelial cells to form new blood vessels, alterations in macrophage function,

collagen accumulation, bone healing, angiogenic response, and the quantity as well as

quantity of granulation tissue is also affected in this pathology (Brem & Tomic-Canic,

2007; Prosdocimi & Bevilacqua, 2012). Chronic diabetic ulcers persist due to a disrupted

Page 45: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

35

formation of granulation tissue and deep tissue necrosis (Galkowska, Wojewodzka, &

Olszewski, 2006). Fibroblasts obtained from diabetic ulcers have shown to differ in their

proliferative capacity when compared to non-diabetic ulcer fibroblasts and age-matched

fibroblasts (Loot et al., 2002). Macrovascular disease, such as a reduction in capillary size

and thickening of the basement membrane, are responsible for circulatory problems

associated with diabetes, which also contributes to the pathogenesis of impaired wound

healing (Falanga, 2005). There is also impaired growth factor production; previous

studies have shown that in diabetic foot ulcers, there is a lack of up-regulation of bFGF

and insulin-like growth factor 1 (IGF-1) expression and reduced expression of TGF-β1

and IL-15 in the vascular endothelium (Galkowska et al., 2006). These growth factors

play roles in angiogenesis, hence this may be responsible for the delayed granulation

tissue formation and retarded healing in those with diabetes (Galkowska et al., 2006).

Although there are various treatment options available, the primary and most important

prevention strategy is maintaining normal blood glucose levels, followed by appropriate

management of the affected wound area, including proper debridement, protective

footwear, systemic antibiotics, pressure off-loading, and moist dressings (Vuorisalo,

Venermo, & Lepantalo, 2009). Specifically, debridement refers to the removal of all

necrotic tissue, callus present around the wound and also foreign material (Wu et al.,

2007). This is important in order to decrease the risk of infection, which can prevent

wound contraction and healing. After debridement, the wound is washed and cleansed

and a moist wound environment should be maintained (Wu et al., 2007). Proper

offloading, one of the biggest challenges in those with diabetic foot ulcers but the primary

mode of healing, includes having patients use a wheel chair, crutches, or even bed rest to

Page 46: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

36

prevent pressure on the affected area (Wu et al., 2007). Total contact casts or removable

cast walkers have also been used (Wu et al., 2007). Current treatment options focus on

modifying controllable causative factors, such as treatment of the wound by oral

antibiotics, such as cephalexin (Isakson et al., 2015). Antimicrobial creams, applied

directly on the ulcer, have also been used. Other interventions include surgical

debridement and negative pressure wound therapy, however these are associated with

long healing times (Isakson et al., 2015). Current treatment options can be ineffective and

limited in their reparative capacities, and there is a need for more innovative therapeutic

alternatives. As a result, MSCs have been studied for their wound healing properties in

many animal models and have shown promise in accelerating healing in diabetic wounds.

Due to the many aberrations present in those with diabetes, such as growth factor

abnormalities, the application of cells may be able to correct and help improve some of

these deficiencies. Particularly, MSCs have shown to enhance wound healing mainly

through the release of cytokines and growth factors that have shown to be absent in the

diabetic wound healing process. The application of cells as therapeutic agents ultimately

results in increased angiogenesis, reepithelialisation, and granulation tissue formation

(Isakson et al., 2015).

Page 47: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

37

Figure 2: Stages of wound healing.

Page 48: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

38

Figure 2. (a) Image depicts intact skin, prior to wound. (b) Immediately after a wound

occurs, a platelet plug forms in order to attain hemostasis. Platelets also have a role in

releasing various cytokines that attract inflammatory cells, such as neutrophils and

macrophages, to the wound site. (c) Neutrophils are amongst the first cells to be recruited,

within an hour after the wound has occurred, in order to clear debris. They also have a

role in releasing additional cytokines. Monocytes are recruited to the wound after 24-48

hours, mature into macrophages, and continue the process of cleaning the wound.

Macrophages secret cytokines in order to stimulate angiogenesis and attract fibroblasts

(d) Proliferation and migration occurs when fibroblasts arrive at the wound and secrete a

collagen matrix, thus forming the bulk of scar tissue. Fibroblasts also differentiate into

myofibroblasts, which help bring the edges of the wound closer together. Epidermal cells

at the wound margin proliferate and migrate into the wound, thus forming a new

epithelial layer. (e) Remodelling occurs when fibroblasts and other inflammatory cells

secrete matrix metalloproteinases (MMPs) to assist in the cross-linking of the collagen

matrix. (Sonnemann, K. J., & Bement, W. M. (2011)).

Page 49: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

39

1.5.3 TSG-6

The gene TSG-6, tumor necrosis factor-stimulated gene 6, was originally identified by

screening a cDNA library from foreskin fibroblasts treated with TNF-α (Milner & Day,

2003). It is a 35 KDa protein, which is not normally expressed in healthy tissues and

cells, but rather secreted in response to inflammation. Hence, it is associated with

inflammatory diseases and its expression is induced by various inflammatory cytokines,

such as TNF-α and IL-10 (Milner & Day, 2003). More specifically, the sera of patients

with arthritis have TSG-6 protein expression, whereas it is not detected in the sera

collected from patients with no history of articular joint disease (Wisniewski et al., 1993).

It has been shown that the expression of the secretory protein TSG-6 is tightly regulated

at the level of transcription by the pro-inflammatory cytokines IL-1, TNF-α, and bacterial

lipopolysaccharide (Klampfer, Chen-Kiang, & Vilcek, 1995). However, studies have

shown that the regulation of the TSG-6 promoter by cytokines is complex (Klampfer et

al., 1995). The region between -163 and -58 in the TSG-6 promoter allows for gene

induction by TNF-α and IL-1. This region contains a CCAAT box as well as binding sites

for the transcription factor activator protein 1 (AP-1) and for the nuclear factor IL-6 (NF-

IL6) family of transcription factors (Klampfer, Lee, Hsu, Vilcek, & Chen-Kiang, 1994).

The NF-IL6 transcription factor (-106 to -114) is needed for the activation of TSG-6 by

the cytokines TNF-α and IL-1, which are likely to be found in an inflammatory region

(Klampfer et al., 1995). Two binding sites for NF-IL6 are needed within the TSG-6 5′

promoter and enhancer region. NF-IL6 can function as an activator or inhibitor of TSG-6

transcription, depending on the activator to inhibitor ratio. Stimulation by cytokines leads

to an increased level of the activator forms of NF-IL6 (Klampfer et al., 1994).

Page 50: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

40

Additionally, the AP-1 regions within the TSG-6 promoter have also been found to be

important for TSG-6 expression by TNF-α and IL-1 (Klampfer et al., 1994). Specifically,

the AP-1 site has been shown to cooperate with the NF-IL6 site in the activation of the

TSG-6 promoter (Klampfer et al., 1994).

TSG-6 has a high degree of sequence homology to the NH2-terminal of the CD44

protein, part of the hyaluronan family of binding proteins (T. H. Lee, Wisniewski, &

Vilcek, 1992). CD44 is a major cell surface receptor for HA found to be expressed on the

surface of a variety of cell types, such as hematopoietic cells, B and T cells (Lesley et al.,

2004). As a result, this indicates that it may also function as a regulator of cell-cell and

cell-matrix interactions during inflammation and tumorigenesis (T. H. Lee et al., 1992).

TSG-6 has also been shown to be able to interact with hyaluronic acid (HA), a

glycosylaminoglycan found in the extracellular component of most tissues, and

abundantly in cartilage as well as synovial fluid (T. H. Lee et al., 1992). Binding of CD44

to HA has been implicated in the adhesion of lymphocytes to endothelium at

inflammatory lesions. HA can form a complex with TSG-6 in an inflammatory milieu,

exhibiting the ability to bind to cells expressing the CD44 receptor (Lesley et al., 2004).

This has been suggested to be a mechanism by which leukocytes exhibit enhanced

adhesion during inflammation, as it has been shown that HA and TSG-6 are upregulated

during an inflammatory response (Lesley et al., 2004).

Recently, TSG-6 protein has been suggested as a biomarker that may be used to predict

the in vivo efficacy of MSCs at reducing sterile inflammation in various animal models.

A study done by Lee et al., showed that there was a wide variation in the efficacy of

human MSCs obtained from bone marrow aspirates of healthy individuals at reducing

Page 51: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

41

inflammation in a chemical injury of the cornea mouse model. To find a possible

biomarker that predicated the efficacy of various cell populations, different characteristics

that have previously been associated with MSC anti-inflammation and immune

suppressive effects were looked at. These included the differential potential of the cells,

as well as the mRNA expression levels of different genes (R. H. Lee et al., 2014). Genes

that were studied included TSG-6, heme-oxygenase 1, cyclooxygenase 2, PGE-2, IL-1

receptor antagonist, TGF-β1, and IDO1, however, only TSG-6 was shown to have a

significant positive correlation with the ability of the cells to suppress inflammation in

vivo (R. H. Lee et al., 2014). In this study, the role of TSG-6 was confirmed by

overexpressing the gene in cells, which initially showed very little mRNA expression.

TSG-6 overexpression resulted in an increased efficacy of the cells in the cornea model

(R. H. Lee et al., 2014). TSG-6 was also seen to be higher in MSCs obtained from female

donors, and a negative correlation was noted with height, weight, osteogenic

differentiation, as well as date of collection, but no correlation with age of the donors was

seen. Many studies to date have shown the correlation of TSG-6 with the therapeutic

potential of the cells. A rat model of intra-cerebral hemorrhage illustrated that MSCs had

a protective effective on the blood brain barrier, and this was thought to be due to the

secretion of TSG-6 by MSCs, which has been shown to inhibit the NF-κB pathway in

macrophages (Figure 3) (Choi, Lee, Bazhanov, Oh, & Prockop, 2011). TSG-6 expression

and secretion has been suggested to be a major factor responsible for MSC induced

inhibition of maturation and function of bone marrow-derived dendritic cells (Liu et al.,

2014), improvement of myocardial infarctions in mice (R. H. Lee et al., 2009),

Page 52: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

42

accelerated wound healing in murine full-thickness skin wounds (Qi et al., 2014), as well

as reduced renal tubular inflammation and fibrosis (Wu et al., 2014).

Page 53: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

43

Figure 3: MSC anti-inflammatory effects are mediated through TSG-6.

Figure 3. The anti-inflammatory effects of MSCs have been proposed to be due to TSG-6,

a secreted protein, acting on macrophage cells present near a wound site. When

inflammation is induced by the administration of zymosan, activated NF-κB signalling

increases the production of pro-inflammatory cytokines, such as TNF-α, which in turn

prime MSCs to secrete TSG-6. TSG-6 deceases the production of pro-inflammatory

cytokines through interaction with CD44 alone or in a complex with hyaluronin. Overall,

this reduces the recruitment of neutrophils to the wound site, hence reducing excessive

inflammation. (Choi, H., Lee, R. H., Bazhanov, N., Oh, J. Y., & Prockop, D. J. (2011)).

Page 54: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

44

1.5.4 Animal Models

MSCs have been used and tested in numerous animal models for their wound healing

capabilities. In vivo, MSCs have an ability to migrate to injured areas, release trophic

factors that aid in tissue repair and regeneration, enhance angiogenesis, act as

chemoattractants for other cell types and inhibit fibrosis and apoptosis (Joyce et al., 2010).

They have been investigated in different preclinical studies for various genetic diseases,

such as Duchenne muscular dystrophy. In such studies, UC-MSCs showed positive

effects, reaching the musculature, however, the cells did not differentiate in skeletal and

muscle cells (Zucconi et al., 2011). MSCs have also been used as therapeutic strategies

for models of lung injury. Using a bleomycin induced lung injury mouse model, MSCs

have shown to hasten repair, decrease fibrosis, and attenuate inflammation in injured

lungs (Rojas et al., 2005). Other studies have illustrated that infused MSCs are able to

home to the lung and adopt an epithelium-like phenotype, reducing collagen deposition in

the lung tissue of mice, thus making these cells candidates for the treatment of lung

disease (Ortiz et al., 2003). Moreover, MSCs have also been shown to be effective in

animal models of myocardial infarction, such as swine and murine myocardial infarction

models, where beneficial effects were seen due to a decreased inflammatory response,

reduced infarct size, and improved cardiac function. Although studies have shown that IV

infused MSCs become entrapped in the lung, others have reported that the cells are able

to implant in the injured myocardium, where they maintain wall thickness and improve

contractile dysfunction (Iso et al., 2007; Kraitchman et al., 2003; R. H. Lee et al., 2009;

Shake et al., 2002). In all the cases, the majority of the cells lodge in the lung, but some

are also able to make it through to the injured tissue. It is possible that the cells trapped in

Page 55: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

45

the lung are secreting factors into the blood stream (Iso et al., 2007). MSCs are also

effective treatments for limb ischemia, diabetic wound healing mouse models, as well as

neurodegenerative diseases, mainly by the secretion of cytokines and chemotactic factors

(Rosova, Dao, Capoccia, Link, & Nolta, 2008).

1.5.5 Diabetic Mouse Models of Wound Healing

Since wound healing is impaired in patients with diabetes, the most accurate wound

healing murine models will incorporate the diabetes phenotype. There are two main type

of diabetes, type I and type II diabetes, each having different root causes. Type I diabetes

is due to autoimmune destruction of the beta cells in the pancreas, which produce the

hormone insulin, and type II diabetes is caused by insulin resistance coupled with beta

cell dysfunction, where there is a lack of beta cell compensation (King, 2012). There are

a plethora of animal models that have been developed to study both type I as well as type

II diabetes, however, the db/db, Akita, and streptozoicin (STZ)-induced C57BL/6J are the

most commonly used in research (Michaels et al., 2007). The db/db mice, representing a

model of type II diabetes, are homozygous for a mutation in the leptin receptor, a major

mediator of satiety, which exerts effects within the arcuate nucleus of the hypothalamus.

This model represents the clinical features of type II diabetes, as the mice become obese

at approximately three to four weeks of age and show an elevation in blood glucose levels

at four to eight weeks (The Jackson Laboratory, 2014). The Akita mice have a

heterozygous mutation in the insulin II gene, while STZ injection causes toxicity and

destruction of the pancreatic islet beta cells, with both these models representing type I

diabetes (Michaels et al., 2007).

Page 56: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

46

The genetic background of the mouse model being used must also be considered. Most

animal models on wound healing use the C57BL/6 db/db mice to study impairments in

wound healing due to type II diabetes (Harris, Mitchell, Yan, Simpson, & Redmann,

2001). Furthermore, strain differences in mice dictate the severity of the diabetes

phenotype and the effect of high-fat diets on the overall health of the mice (Harris et al.,

2001). The db/db genotype in C57BL/6 mice behaves in a similar fashion to the ob/ob

mutation on the same strain. In comparison to the C57BL/KsJ background, C57BL/6

mice merely become insulin resistant, while the C57BL/KsJ develop overt diabetes

(Hummel, Coleman, & Lane, 1972). Although the two strains are alike in the early stages

of disease, illustrating hyperglycemia, rapid weight gain, and hyperinsulinemia, disease

progression differs at later stages. Specifically, C57BL/KsJ strain illustrates severe

diabetes, as seen by hyperphagia, obesity, permanent hyperglycemia starting at 2-3

months, temporarily elevated plasma insulin concentrations (which seem to be halted

prematurely) and degenerative changes in the islets of Langerhans (Hummel et al., 1972).

Diabetes is less severe on the C57BL/6 background. Like C57BL/KsJ mice, hyperphagia

and obesity are present, however, these mice display mild diabetes characterized by

transitory hyperglycemia and markedly elevated plasma insulin concentrations, as well as

hypertrophy of the islet cells and increased proliferative capacity of the beta cells

(Hummel et al., 1972). Other mice with the db/db phenotype include BALB/c mice.

When compared with BALB/c mice, C57BL/6 mice have a higher susceptibility to diet-

induced obesity, type 2 diabetes, and atherosclerosis (The Jackson Laboratory, 2014).

Type II diabetes is a pathology which represents a major health concern worldwide, due

to the prevalence of a sedentary lifestyle and diets rich in calories (Wild, Roglic, Green,

Page 57: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

47

Sicree, & King, 2004). There are many different models of rodent wound healing that

have been described and used, including incisional, excisional, burn, as well as

granulation tissue models (Galiano, Michaels, Dobryansky, Levine, & Gurtner, 2004).

While excisional wounds are most commonly used in the literature, there are numerous

variations in the size of the wound generated, the number of wounds, how the wound is

generated, as well as occlusive dressings, splints, or non-occlusive bandages that have

been used (Ansell, Campbell, Thomason, Brass, & Hardman, 2014). Incisional wounds

usually exhibit more similarity among studies; 10-15 mm in length, full thickness, and

scalpel induced, where most commonly a suture is used to close the wound margins

(Ansell et al., 2014). Wound healing in both models (incisional and excisional) fill with a

matrix composed of fibrin, fibronectin and plasma-derived components (Davidson, 2001).

Burn models can also be used, induced chemically, thermally, or by radiation. Thermal

burns have been shown to create a zone of coagulative necrosis, in which the denaturation

of plasma and cellular protein leads to the obstruction of blood vessels (Davidson, 2001).

Hence, tissue is devoid of many essential nutrients (Davidson, 2001). The epithelial layer

of the skin is more intact in first and second degree burns, however healing does not

progress as rapidly because the necrotic tissue must first be eliminated (Davidson, 2001).

Granulation tissue wound healing models, also known as dead space models, are

designed to measure the amount of newly deposited connective tissue, hydroxyproline

formation, collagen formation and fibrils, as well as identification of different cell types

(Davidson, 2001). Subcutaneous implants lead to the formation of a fibrin clot, and

granulation tissue formation (Davidson, 2001). These models selectively study

connective tissue formation during wound healing by placing polyvinyl alcohol sponges,

Page 58: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

48

consisting of a small piece of perforated silicone tubing filled with silicone strips of

polyvinyl alcohol sponge, in the subcutaneous space of the skin, and removed after a

certain number of days (Davidson, 2001). Histological and biochemical analysis is then

done to examine properties of healing (Diegelmann, Lindblad, & Cohen, 1986). PVC

tubes can also be implanted, consisting of a PVC tube with polyester-polyurethane

sponges (Paulini, Korner, Beneke, & Endres, 1974). Alternatively, vicose cellulose

sponges of different designs have also been used (Pallin, Ahonen, Rank, & Zederfeldt,

1975). Other materials that can be used include porous Teflon tubing and nylon mesh

(Davidson, 2001).

Challenges to translating mouse wound healing studies to humans arise due to differences

between mouse and human wound healing. Mice have a subcutaneous muscle layer,

called the panniculus carnosus, which heals wounds mainly by contraction, rather than by

granulation tissue formation and fibrosis as seen in humans (Perez & Davis, 2008). This

is the primary method of wound healing in rodents. However, the excisional wound splint

method offers a murine model that closely resembles wound healing in humans, because

the splint keeps the wound open. Healing occurs from the wound margins, and the

process of epithelization, granulation tissue formation, scar formation, contraction, and

angiogenesis takes place, as the splint prevents contraction of the skin (Galiano et al.,

2004). In full thickness wounds occurring in mice as well as humans, there is a loss of the

basement membrane, hence the wound must heal by re-epithelization by keratinocytes as

well as by extracellular matrix production (Stroncek JD, 2008). MSCs have been used as

a cellular therapy and have shown many therapeutic effects in cutaneous wound repair.

For instance, the application of MSCs on excisional wounds in db/db mice has shown to

Page 59: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

49

exhibit a decreased epithelial gap, increased granulation tissue formation, acceleration of

wound closure by promoting cell migration to the wound site, as well as increased

neovascularization, hence supplying a greater amount of oxygen and nutrients to the

wound site (Arno et al., 2014; Javazon et al., 2007). MSCs promote wound repair not by

engrafting into the injury site or by differentiation into other cell types, but rather by the

secretion of various cytokines, such as vascular endothelial growth factor (VEGF),

epidermal growth factor (EGF), and erythropoietin (Chen, Wong, & Gurtner, 2012).

These factors have various anti-inflammatory, angiogenic, and chemotactic properties,

stimulating the recruitment of keratinocytes, macrophages and other cell types (Chen et

al., 2012). Overall, MSCs have been shown to accelerate the healing of chronic cutaneous

wounds, thus making them ideal therapeutic candidates in clinical settings.

Page 60: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

50

1.6 Rationale, Hypothesis and Objectives

1.6.1 Rationale

Previous work on MSCs has largely focused on isolation of the cells from BM. UC-

MSCs have been investigated in some studies for their therapeutic potential in various

models, mainly using enzymatic digestion of the tissue to isolate cells. However, isolation

of UC-MSCs using an explant culture of the whole umbilical cord tissue has not been

previously studied in detail for large sample sizes. Preceding studies in our lab have

proven that MSCs isolated from the umbilical cord are able to enhance wound healing in

a db/db excisional wound mouse model. However, the patient to patient differences in

MSC samples has not been looked at, and the passage variation in immunophenotype has

also not been investigated. Literature suggests that different MSC populations exhibit

variations in their therapeutic potential in murine models, and that the gene TSG-6 may

be used as an informative biomarker to predict the in vivo efficacy of cells. This work has

been done with BM-MSCs, however whether the same trend is seen with UC-MSCs

applied in a murine diabetic excisional wound model, and the impact this may have on

cytokine secretion profiles of MSC populations, remains largely unknown.

1.6.2 Hypothesis and Objectives

I propose the following hypothesis: MSCs isolated from different cord tissue units may

exhibit variations in their immunophenotype with increasing passage number and show

varying TSG-6 mRNA expression profiles, thus this may impact the cytokine secretion

profile and wound healing capabilities of different cell populations.

Page 61: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

51

To test this hypothesis, I propose the following four aims:

1. Determine the isolation efficiency from 40 different umbilical cord samples.

2. Study the phenotypic profile of 20 MSC samples with passage number and between

patient samples.

3. Investigate the cytokine secretion profile of UCT-MSCs.

4. Assess TSG-6 as a potential candidate marker to evaluate the wound healing efficacy

of two UCT-MSCs populations, expressing high and low levels of TSG-6 mRNA, in a

db/db excisional wound mouse model.

Page 62: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

52

Chapter Two: Experimental Methods and Materials

Page 63: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

53

2. Experimental Methods and Materials

2.1 Umbilical Cord Collection and Preparation

Cord tissue (n=40) was obtained from full-term, vaginal and caesarean deliveries from

across Canada. Cord tissue was sprayed with 70% ethanol, and alcohol gauze was used to

wipe down the entire length of the cord until the exterior surface was clean. The cord

tissue was transferred to a petri dish and a scalpel was used to cut off the end portions of

the tissue, which were discarded. Five ringlets, each approximately 4 mm in length, were

cut from each tissue, and washed twice with 10 ml of phosphate buffered saline (PBS -/-).

The arteries as well as the vessels were flushed with PBS (-/-) to remove any blood clots.

All four pieces were transferred to a 2 ml cryovial and 1.8 ml of cold CryoStor (BioLife

Solutions, Lot# 15003) was added to each, ensuring that all sections were free-floating in

the freezing solution. Each cryovial was chilled for 10 minutes at 4°C and then

transferred into a pre-chilled freezing device, and placed in the -80°C freezer overnight.

Cyrovials were then transferred into liquid nitrogen the next day for long term storage.

Sterility testing was done for each cord tissue sample, with BacT/ALERT Standard

Aeorbic and BacT/ALERT Standard Anaerobic bottles (bioMerieux Inc., USA), using 4

ml of PBS (-/-) obtained from washing each cord tissue unit.

2.2 MSC Isolation

MSCs were isolated using an explant culture protocol. Pieces of umbilical cord were

thawed in a 37°C water bath for 2 minutes. Tissues were then submerged in 10 ml of

medium (complete alpha-MEM, 10% Fetal Bovine Serum, 1x Antibiotic-Antimycotic;

Life Technologies Lot #1631429). Tissue pieces were evenly plated in a single well of a

6-well polystyrene dish (Falcon) in 1 ml of complete alpha-MEM, labelled as "P0I".

Page 64: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

54

Medium change was done 24 hours post thaw and every 2 days afterwards. MSCs started

to migrate from the cord tissue explants at various time points, ranging from 5-38 days

(Figure 4). When 80% confluency was reached, the cells were passaged using 0.25%

trypsin-EDTA solution and the whole contents of the plate were transferred on 10-cm

plates (Falcon), labelled as P1. At this point, tissue pieces were moved to a new well in a

6-well polystyrene dish, labelled as "P0II." The cells from P0II were passaged when

confluency was reached.

Page 65: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

55

Figure 4. Images depict primary culture of human UCT-MSCs, illustrating the umbilical

cord surrounded by MSCs which have migrated from the tissue and adhered to the plastic

dish. Cell isolation can be seen at 5 days after initial plating of the tissue (Figure A). Cells

become more confluent at 8 days (Figure B) and at 14 days (Figure C). Magnification, 4X.

Figure 4: Explant culture of human umbilical cord samples.

Page 66: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

56

2.3 Cell Culture

At each passage, cells were plated at a density of ~230,000 cells/10 cm plate (Falcon),

with 10 ml/plate of complete alpha-MEM. Cultures of cells were maintained in an

incubator at 37°C with 5% CO2. Complete medium change was done every 2 days. Once

the cell cultures were 80-90% confluent, cells were replated 1:4 in 10 cm tissue culture

plates with 10 ml of complete alpha-MEM for expansion.

2.4 Cryopreservation

Cells at specific passages were cryopreserved using 80% serum (complete alpha-MEM,

10% Fetal Bovine Serum, 1x Antibiotic-Antimycotic) and 20% dimethyl sulfoxide

(Sigma Life Science, Lot# RNBD5041), in either 1 ml or 1.5 ml cryovials. The vials were

placed in an isopropanyl buffered container (Mr. Frosty) for step freezing at a rate of -

1ºC/minute to -80ºC overnight and then transferred to liquid nitrogen for long term

storage.

2.5 Flow Cytometry Analysis

Cell surface antigen expression was analyzed by flow cytometry. Cells at passage 1

(n=40), p5 (n=20), and p10 (n=20) were harvested by treatment with 0.25% trypsin-

EDTA, washed using 10 ml of PBS (-/-), and re-suspended in stain buffer (PBS -/- with

1% fetal bovine serum) at a concentration of 1x107 cells/ml. When cell number was a

limiting factor, cells were re-suspended at a concentration of 5x106 cells/ml, or lower.

100 μl of prepared cell suspension was aliquoted into a total of nine tubes. Cells were

incubated with 2 μl of IgG from mouse serum (Sigma, Canada) in the dark for 10 minutes.

Cells were then stained using the Human MSC Analysis Kit (BD Biosciences, Canada)

Page 67: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

57

with the appropriate antibody diluted 1:10 or 1:30, as listed in Table 1. After incubation

for 30 minutes on ice in the dark, cells were washed twice with 1 ml of stain buffer and

centrifuged at 400xg for 2 minutes. Afterwards, cells were re-suspended in 300 μl of stain

buffer and 0.5 μl of DAPI was added to each tube. Antibody binding was analyzed using

a Beckman Coulter flow cytometer. Multicolour fluorescent beads (Flow-Set Pro

Fluorospheres, Beckman Coulter Ireland, Inc., Lot#3125121) were used for instrument

standardization and to ensure reproducibility of each experiment, where the fluorescence

of the beads in each channel was adjusted to match the fluorescence values from previous

experiments. All plots were generated using Kaluza Flow Analysis Software (Beckman

Coulter). Debris and auto-fluorescence were removed by using forward scatter (FS) and

side scatter (SS). Two light scatter parameters, FS and SS, were used to ensure a stringent

gating of single cells. A dot plot depicting side scatter time of flight (SS TOF) vs. SS

peak was first used to gate single cells. Aggregates which escaped the single cell gate

could be seen as the few events which were high in forward scatter time of flight (FS

TOF) signal in the second dot plot. The 488 nm blue laser detected both scatter

parameters. DAPI was then used to discriminate between live, apoptotic, and dead cell

populations. A gate was used to select DAPI negative (live cells) on a FL9: DAPI vs.

forward scatter peak dot plot. The maximum number of events that was used for each

analysis was 15,000 cells. Isotype controls were included in the analysis to identify

positive and negative cell populations, ensuring that the observed staining is due to

antibody binding to surface epitopes, rather than artefact, and that non-specific binding to

Fc receptors is excluded (AbD Serotec, 2015). Compensation controls were done for each

fluorochrome and this was used to subtract the spectral overlap of specific fluorochromes.

Page 68: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

58

Table 1: Antibodies and fluorochromes used to analyze UCT-MSCs.

Tube Test

1 FITC Mouse Anti-Human CD90 (1:30 dilution)

2 PE Mouse Anti-Human CD44 (1:30 dilution)

3 PerCP-Cy5.5 Mouse Anti-Human CD105 (1:30 dilution)

4 APC Mouse Anti-Human CD73 (1:30 dilution)

5 Nothing

6 hMSC Positive Isotype Control Cocktail (1:10 dilution)

PE hMSC Negative Isotype Control Cocktail (1:10 dilution)

7 hMSC Positive Cocktail (1:10 dilution)

PE hMSC Negative Cocktail (1:10 dilution)

8 hMSC Positive Isotype Control Cocktail (1:10 dilution)

PE Mouse IgG2b,k hMSC Positive Isotype Control Cocktail (1:10 dilution)

9 hMSC Positive Cocktail (1:10 dilution)

PE Mouse Anti-Human CD44 (1:30 dilution)

Table 1. Antibody markers used to stain each sample, PE, phycoerythrin; FITC,

fluorescein isothiocyanate; PerCP-Cy5.5, peridin chlorophyll protein-cyanine 5.5; APC,

allophycoerythrin.

Page 69: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

59

2.6 RNA Extraction and Real-Time PCR

Total RNA was extracted (RNeasy Mini kit; Quigen) from 16 randomly chosen MSC

umbilical cord samples (passage 4), from cryopreserved cells. Cryovials were obtained

from liquid nitrogen, and immediately incubated in a 37°C water bath for 2 minutes. The

cells were then transferred, drop-wise, to a 50 ml Falcon tube containing 10 ml of alpha-

MEM medium. Cells were pelleted at 290xg for 5 minutes, after which the supernatant

was removed and RNA was extracted from each sample. RNA concentration was

quantified using a Nanodrop Spectrophotometer and for a subset of samples, RNA

integrity was looked at using the Agilent RNA ScreenTape Assay (Agilent Technologies,

Canada). 0.38 µl/µg of total RNA per sample was used for cDNA synthesis using reverse

transcriptase (Superscript III Reverse Transcriptase; Invitrogen). cDNA concentrations

were quantified using a Nanodrop Spectrophotometer. Real time RT-PCR analysis was

completed in triplicate for human GAPDH and human TSG-6 for all samples (Table 2). A

primer optimization protocol was done to determine the ideal concentration for each

primer pair. The optimal concentration for TSG-6 was found to be 300 nM of forward

primer and 900 nM of reverse primer. The primer concentrations for GAPDH used was

300 nM of forward primer and 300 nM of reverse primer. SYBR Green PCR MasterMix

(Invitrogen, Canada) was used for gene amplification and analyzed using the Applied

Biosystems 7900HT PCR machine. Reaction profiles consisted of an incubation at 95°C

for 10 seconds, followed by 40 cycles at 95°C for 15 seconds and 60°C for 1 minute, and

lastly 95°C for 15 seconds, 60°C for 15 seconds, and 95°C for 15 seconds. Data was

analyzed with the Sequence Detection Software 2.1 (Life Technologies), using the

standard curve method.

Page 70: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

60

Table 2: Real time RT-PCR Primer Sequences.

Gene Forward and Reverse Primer Sequence (5′ to 3′) Amplicon Size (bp)

TSG-6

F: AGCACGGTCTGGCAAATACA

129

R: GCAGCACAGACATGAAATCCAAT

GAPDH

F: CGAGCCACATCGCTCAGA

95

R: AGTTAAAAGCAGCCCTGGTGA

Table 2. List of the forward and reverse primer sequences for human TSG-6 and human

GAPDH and the amplicon size. The melting temperature of the primers was 60°C.

GAPDH= Glyceraldehyde 3-phosphate dehydrogenase, TSG-6= TNF-alpha stimulated

gene 6.

Page 71: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

61

2.7 Cytokine Array

2.7.1 Array Procedure

The cytokine and growth factor expression profile of three umbilical cord samples were

compared using a proteome assay (R&D Systems, Minneapolis, USA). Passage 5 were

obtained from liquid nitrogen, and thawed in a 37°C water bath for 2 minutes. The cells

were then transferred, drop-wise, to a 50 ml Falcon tube containing 10 ml of alpha-MEM

medium. Cells were pelleted at 290xg for 5 minutes. The supernatant was then removed

and cells were re-suspended in 1 ml of complete alpha-MEM, after which 0.5 ml of the

suspension was plated onto one 10 cm tissue culture dish. 10 ml of complete alpha-MEM

was added to each dish afterwards and cells were incubated at 37°C with 5% CO2. A

complete medium change was done 24 hours post-thaw and every 2 days afterwards.

Cells were passaged 1:4 onto 10 cm culture dishes when confluency was reached. At

passage 7, cells were plated at 100,000 cells/well in a 24 well dish. Duplicate wells were

done for each cord sample. Cells were maintained in 2 ml/well of complete alpha-MEM

and medium change was done every two days. The cells were allowed to reach 80%

confluency, at which point 1 ml of complete alpha-MEM was added to each well. The

supernatant was conditioned for 48 hours prior to the assay. All reagents and the protocol

outline were conducted according to the manufacturer's instructions using the Human

Cytokine Array C1000: AAH-CYT-1000 (R&D Systems, Minneapolis, USA). This

antibody array kit analyzed 55 different cytokines and chemokines in duplicate on each

membrane (Table 3). The conditioned medium was collected (1 ml for each sample) and

centrifuged at 290xg for 5 minutes. The array membranes were incubated for 1 hour in

blocking buffer. Membranes were then incubated with 1 ml of prepared sample,

Page 72: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

62

containing cell supernatant with a detection antibody cocktail, overnight in a cold room

(2-4°C) on a rocking platform. Membranes were then washed for 10 minutes on a rocking

platform. This was repeated three times, after which diluted streptavidin-HRP was added.

The membranes were incubated for 0.5 h at room temperature on a rocking platform.

Afterwards, membranes were washed three times for 10 minutes each followed by

incubation for 1 minute with a Chemi Reagent Mix. Membranes were exposed to X-ray

film (Denville Scientific Inc., Metuchen NJ) for 2, 5, and 10 minutes. Pixel density was

measured using the ImageJ Software (National Institutes of Health).

Page 73: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

63

Table 3: Cytokines detected in the array kit.

Systematic Name Abbreviation Alternative

Nomenclature

Reference Spot

Activin A

ADAMTS-1

Angiogenin ANG

Angiopoietin-1 Ang-1

Angiopoitein-2 Ang-2

Angiostatin/Plasminogen

Amphiregulin AR

Artemim

Reference Spot

Coagulation Factor III TF

Chemokine (C-X-C motif) ligand 16 CXCL16

Dipeptidyl Peptidase IV DPPIV CD26

Epidermal Growth Factor EGF

Endocrine Gland-Derived Endothelial

Growth Factor

EG-VEGF PK1

Endoglin CD105

Endostatin/Collagen XVIII

Endothelin-1 ET-1

Fibroblast Growth Factor acidic FGF acidic FGF-1

Fibroblast Growth Factor basic FGF basic FGF-2

Fibroblast Growth Factor-4 FGF-4

Fibroblast Growth Factor-7 FGF-7 KGF

Glial Cell-Derived Neurotrophic Factor GDNF

Granulocyte-Macrophage Colony-

Stimulating Factor

GM-CSF

Heparin-binding EGF-like Growth Factor HB-EGF

Hepatocyte Growth Factor HGF

Insulin-like Growth Factor-Binding Protein 1 IGFBP-1

Insulin-like Growth Factor-Binding Protein 2 IGFBP-2

Insulin-like Growth Factor-Binding Protein 3 IGFBP-3

Interleukin-1β IL-1β IL-1F2

Interleukin-8 IL-8 CXCL8

LAP (TGF-β1)

Leptin

Monocyte Chemoattractant Protein-1 MCP-1 CCL2

Macrophage Inflammatory Protein-1α MIP-1α CCL3

Matrix Metalloproteinase-8 MMP8

Matrix Metalloproteinase-9 MMP9

NRG1- β1 HRG- β1

Pentraxin 3 PTX3 TSG-14

Page 74: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

64

Platelet-Derived Endothelial Cell Growth

Factor

PD-ECGF

Platelet-Derived Growth Factor-AA PDGF-AA

Platelet-Derived Growth Factor-AB/ Platelet-

Derived Growth Factor-BB

PDGF-AB/PDGF-

BB

Persephin

Platelet Factor 4 PF4 CXCL4

Placental Growth Factor PIGF

Prolactin

Serpin B5 Maspin

Serpin E1 PAI-1

Serpin F1 PEDF

Tissue Inhibitor of Metalloproteinases-1 TIMP-1

Tissue Inhibitor of Metalloproteinases-4 TIMP-4

Thrombospondin-1 TSP-1

Thrombospondin-2 TSP-2

Urokinase-type Plasminogen Activator uPA

Vasohibin

Vascular Endothelial Growth Factor VEGF

Vascular Endothelial Growth Factor-C VEGF-C

Reference Spots

Negative Control

Table 3. A list of the 55 cytokines and chemokines detected in duplicate on nitrocellulose

membranes, showing the systematic names and alternative nomenclature.

Page 75: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

65

2.7.2 Data Analysis

Pixel density was quantified using ImageJ Software (National Institutes of Health). In

order to obtain the net cytokine value, each image was converted to greyscale and an

inverted value for each cytokine was calculated by subtracting the pixel density obtained

on ImageJ, for each cytokine, from 255. The background pixel density from each

membrane was subtracted from the net cytokine value. Afterwards, each cytokine value

was normalized to the loading control. The average of four density values was then

calculated to obtain the final pixel density for each respective cytokine.

2.8 Excisional Murine Model

2.8.1 Surgical Procedure

Ten-week old db/db C57BL/6 mice were purchased from The Jackson Laboratories

(Jackson Laboratories, Stock #000642). Twelve-week old db/db C57BL/6 mice were

used for their impaired wound healing capabilities. Mice were individually housed for

two weeks prior to surgeries in an animal facility with a 12-hour light/dark schedule.

Each mouse was provided ad libitum access to food and water. On the day of the

surgeries, the back of each mouse was shaved with an electric clipper followed by the

application of Nair, to ensure that all hair was removed. The surgery site was wiped clean

with an alcohol swab. An isoflurane gas chamber was used to induce anaesthesia in each

mouse, at 1-4% isoflurane. Each mouse was maintained at a 1-2% isoflurane level during

the surgery. Mice were individually anesthetised using a subcutaneous injection of

buprenorphine Temgesic (0.15 mg/kg). A silicone splint was attached to the back of each

mouse using an adhesive (Krazy Glue, Elmer's Inc., Columbus, OH) and four nylon

sutures were used to ensure that the splint was fixed to the back of each mouse. A sterile

Page 76: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

66

6-mm biopsy punch was used to induce a full thickness wound extending through the

panniculus carnosus, centered in the middle of the splint, on the back of each mouse.

CT15P8 and CT16P8 MSCs were obtained from liquid nitrogen and thawed, after which

3x106 cells were resuspended in 40 μl of PBS (-/-) and applied topically on each wound,

followed by 40 μl of a fibrin sealant. For control mice, 40 μl of alpha-MEM media was

applied to the wound bed, followed by 40 μl of a fibrin sealant. A clear polyurethane

dressing (Tagaderm) was placed over the entire wound area. Animals were transferred to

a clean cage, and placed under a warming lamp for 10 minutes to recover from the

procedure. The mice were house in the animal facility at LTRI.

2.8.2 Wound Analysis

Digital pictures of the wounds were taken with a Nikon camera at day 0, day 3, day 7,

day 10 and day 14 post surgery. The wound area was analyzed by measuring the area of

splint (the inner diameter only) and the area of the wound (by delineating the margins of

the wound) on each respective day, using ImageJ Software (National Institutes of Health)

(Figure 5). The ratio of the percent of remaining wound relative to the splint was

calculated. The final wound area was calculated as a percent of the original wound

(denoted as 100%), created on day 0.

Page 77: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

67

Figure 5. Representative images illustrating how wound closure and healing are

calculated. The splint as well as wound area were traced, after which the inner area of the

splint (A) was compared against the area of the wound (B). Final wound closure on each

respective day was calculated as a percent of the original wound size (day 0, denoted as

100%).

Figure 5: Wound healing calculations.

Page 78: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

68

2.8.3 Tissue Collection and Fixation

An isoflurane gas chamber was used to induce anaesthesia in each mouse, at a 1-4%

isoflurane concentration, for approximately 1 minute. Mice were euthanized by cervical

dislocation. Tagaderm dressing was removed, and the wound area, including the splint

was excised, cut in half, and fixed in 10% formalin for 120 minutes at 4°C. Tissues were

washed in 1 x PBS (-/-) and stored in 70% ethanol for 48 hours. The tissues were then

dehydrated in a graded ethanol series (80% for 30 minutes, 95% for 45 minutes, and

twice in 100% ethanol for 60 minutes). The tissues were cleared in toluene two times for

60 minutes, immersed in paraffin at 65°C, and embedded in paraffin blocks. The

embedded tissues were sectioned on a microtome and cut into 5 µm sections, placed on

Fisherbrand Superfrost Plus (Fisher Scientific) microscope slides, and allowed to dry

overnight.

2.8.4 Immunohistochemistry

Immunohistochemistry was done on paraformaldehyde fixed tissue sections. All samples

were embedded in paraffin and underwent routine histological processing. Sections were

deparaffinized in xylene and rehydrated using an ethanol gradient on a Leica staining

machine. TBE buffer or citrate buffer was used for antigen retrieval, performed in a

microwave. Tissue sections were blocked using goat serum. Sections from all mice were

incubated, overnight at 4°C, with the following antibodies: rabbit anti-Ki-67 (Thermo

Fisher Scientific Inc., USA), mouse anti-smooth muscle actin (Dako, CA, USA), rabbit

anti-human collagen IV (Abcam, Crambridge, MA, USA), and mouse anti-cytokeratin 6

(Biolegend, CA, USA). Sections were stained with secondary antibodies conjugated to

Alexa Fluor 488 Goat Anti-Mouse IgG or Alexa Fluor 594 Donkey Anti-Rabbit IgG.

Page 79: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

69

Sections were then incubated in DAPI for five minutes for nucleus visualization. Slides

were mounted using DABCO. Each section was visualized under using a Zeiss LSM 510

confocal microscope. Sections were also stained with haematoxylin and eosin. A wound

was considered entirely healed if the wound bed was completely filled in with new tissue,

spanning the area of the whole wound.

2.9 Statistical Analysis

A two-tail student’s t-test for paired data was done to assess significance between passage

numbers for immunophenotypic analysis. Wound closure comparisons were performed

by a two-way ANOVA followed by a Bonferroni post hoc test. Statistical analysis was

done using GraphPad Prism Software. Significance was assumed for p<0.05.

Page 80: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

70

Chapter Three: Results

Page 81: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

71

3. Results

3.1 MSC Isolation Efficiency and Explant Culture

Most MSC studies have used cells derived from the BM. As a result, there is a plethora of

information regarding BM-MSCs, specifically in regards to their growth characterises,

immunophenotype, and passage variation. Among the other sources of MSCs being

studied are MSCs derived from the UCT. Although some research groups have looked at

characterizing this cell type, the information is sparse or contradictory. As a result we

aimed to characterize a large sample size of MSCs derived from human umbilical cord

tissue. Characterization includes; isolation efficiency, growth characteristics,

immunophenotype, passage variation and therapeutic potential.

Information pertinent to collection was recorded, such as the mother's age, the type of

birth, gender and baby weight (Table 4). Information that was not collected at the time of

birth is denoted as N/A. Most cord samples were collected from vaginal deliveries, from

a maternal age range of 21-41 years old.

There are different methods to isolate MSCs from UCT, the most common is to first

isolate the Wharton's jelly followed by collagenase treatment to separate out the MSCs.

In this study we opted for the less expensive and less labour intensive method of directly

isolating MSCs from explant cultures. This method relies on a culture medium that

selectively supports the growth of MSCs and not other cells of the cord such as

endothelial cells. Cutting small sections of cord tissue and plating them in selective

medium allows for minimal manipulation and does not require the use of collagenase,

which is both expensive and adds an extra regulatory hurdle when obtaining approval for

clinical use of the UCT-MSCs. Although the different types of cells that can be isolated

Page 82: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

72

from UCT, including MSCs, endothelial cells, muscle, pericytes and blood, all have

proven therapeutic properties, for clinical use the population of cells must be defined. In

this study I set out to determine the characteristics and properties of the cells isolated

using a defined explant culture system.

As described in the materials and methods, freshly collected umbilical cords, drained of

blood were washed, sectioned and frozen. Cell isolation was done on frozen tissue that

was quickly thawed at 37ºC for 2 minutes, rinsed in alpha-MEM/5% FBS. Tissue was

placed onto dry tissue culture plates and medium was added as to not disrupt the tissue.

Cell outgrowth appeared 5-38 days after the initial plating (Table 5). Figure 6 depicts the

results as a bar graph. All cells that appeared from the tissue were heterogeneous in size

however demonstrated a typical MSC spindle shaped appearance. There were also few

cells that resembled endothelial cells. The cells migrated away from the tissue and at first

were scattered and demonstrated no organization. As the cells proliferated and became

more confluent they aligned forming parallel organized sheets of cells. For all samples

plated the cells were not passaged until the 35 mm well was 80% confluent. The contents

of the well was counted and all of it was passaged onto a 10 cm plate (designated as

passage 1). The average number of cells obtained at first passage, from one 35 mm well,

was 75, 000 for a subset of 9 UCT-MSC samples (Figure 7). At this point all samples

exhibited elongated spindle shaped appearance. Cells were passaged 1:4 onto 10 cm

plates once confluency was reached. Cord tissue samples yielded different cell counts

with passaging (Figure 8).

Page 83: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

73

Table 4: Personal data collected for each cord sample.

Table 4. Personal data collected for each cord tissue sample includes maternal age, type

of birth, gender of newborn, as well as weight of newborn (g).

Cord Tissue

Sample

Age of Mother Type of Birth Gender of

Newborn

Weight of Baby

(g)

CT 16 27 Vaginal Male 3164

CT 17 35 Vaginal Female 3840

CT 18 21 Vaginal Female N/A

CT 19 39 Vaginal Female N/A

CT 20 31 Vaginal Female N/A

CT 21 34 Vaginal Male 3101

CT 22 31 Vaginal Male 2795

CT 23 33 Vaginal Female 4015

CT 24 38 Vaginal Female 2380

CT 25 41 N/A N/A N/A

CT 26 34 Vaginal Male 2965

CT 27 34 Caesarian Female 3528

CT 28 30 Caesarian Female 3359

CT 29 30 Vaginal Male 3294

CT 30 31 Vaginal N/A 3355

CT 31 34 Vaginal N/A 2930

CT 32 29 Vaginal Female 6700

CT 33 30 Vaginal Female N/A

CT 34 35 Caesarian Female 3355

CT 35 40 Vaginal Male N/A

Page 84: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

74

Table 5: Growth characteristics of individual cord samples.

Table 5. Time to cell outgrowth ranged from 5-38 days, depending on the cord tissue

sample.

Cord Tissue Sample Time to Cell Outgrowth (Days)

CT 16 15

CT 17 16

CT 18 16

CT 19 16

CT 20 16

CT 21 16

CT 22 16

CT 23 16

CT 24 16

CT 25 14

CT 26 17

CT 27 27

CT 28 14

CT 29 17

CT 30 22

CT 31 14

CT 32 22

CT 33 14

CT 34 14

CT 35 14

CT 36 38

CT 37 38

CT 38 17

CT 39 17

CT 40 17

CT 41 17

CT 42 5

CT 43 5

CT 44 5

CT 45 5

CT 46 5

CT 47 38

CT 48 5

CT 49 5

CT 50 20

CT 51 5

CT 53 5

CT 54 5

CT 55 5

Page 85: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

75

Figure 6. Time to cell outgrowth for 40 UCT-MSC samples.

Figure 6. Time to cell outgrowth ranged from 5-38 days, depending on the cord tissue

sample.

Page 86: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

76

Figure 7. Cell number obtained at first passage for 9 UCT-MSC samples.

Figure 7. The average number of cells obtained at first passage was 75,000 cells/ 35 mm

well dish for a subset of 9 UCT-MSCs.

Page 87: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

77

Figure 8. Cell number obtained at each passage for three MSC populations.

Figure 8. Variable cell numbers were attained for CT17, CT19, and CT22 with passaging.

Cell counts at each passage represent the results obtained for one 10 cm plate. Each plate

was passaged 1:4 when confluency was reached.

Page 88: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

78

3.2 MSC Immunophenotype

Immunophenotypic analysis of MSCs obtained from explant culture was done by flow

cytometry analysis for each cord sample at early (n=40), mid (n=20) and late (n=20)

passages to analyze cell surface marker expression. All UCT-MSC samples were

analyzed using the BD Stem Flow Human MSC Analysis Kit. Cells at each passage were

collected, and stained with a positive cocktail of markers, consisting of a set of markers

showing specificity for stromal cells, a negative cocktail, containing a set of markers used

to identify hematopoietic cells, compensation controls, as well as respective isotope

controls. All samples analyzed illustrated similar phenotypic cell surface expression

profiles. Figure 9 shows three cord samples stained for the positive and negative human

monoclonal antibodies at early, mid, and late passages.

When analyzing the same 20 samples at early, mid, and late passages, we observed that

the greatest number of cells expressing makers indicative of hematopoietic cells were

found at the earliest passage analyzed (p2), indicating that the isolation procedure results

in a heterogeneous cell population. There is a significant decrease in the number of cells

expressing hematopoietic markers from p2 to p10 (P<0.01). The percent of cells showing

CD45 expression was significantly reduced at mid and late passages, thus suggesting that

the media selects for MSCs specifically (Figure 10). No difference in the detection

profiles of the expression levels of CD44 was noticed at low (99.7% ± 0.059), mid

(99.5% ± 0.374), or high (99.2% ± 0.372) passage. The same trend was observed for

CD90 at low (99.7% ± 0.0787), mid (99.8% ± 0.05029) and high (99.0% ±0.56848)

passage, as well as with CD73 at low (99.7% ± 0.0579) mid (99.5% ± 0.0146) and high

(99.2% ± 0.3415) passage. It was noticed that CD105 expression was lower when

Page 89: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

79

compared to the other positive markers, and its expression levels also decreased with

increasing passage numbers, illustrating the lowest expression at passage 10 (84.7% ±

3.525). There was a significant decrease in cell surface expression of CD105 from p5 to

p10 (P<0.05).

Because I started with 40 independent samples but only carried 20 samples all the way

through the ten passages, a graph with the complete data is presented in Figure 11. This

includes the additional 20 UCT-MSC samples for passage 2 only. A significant decrease

of CD105 expression was noticed from p2 to p10, and from p5 to p10.

Page 90: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

80

P2

FS Peak

FS Peak

CD

90

FIT

C

CD

73

AP

C

CD

44

PE

Figure 9: Colour density plots of CT16, CT24, and CT15 at early, mid and late

passages.

A.

B.

C.

.

CD

34

/11

b/1

9/4

5/H

LA

-DR

PE

C

D3

4/1

1b

/19

/45

/HL

A-D

R P

E

P5

P10

P2

P5

P10

CD

10

5 P

er-C

P-C

Y5

.5

CD

90

FIT

C

CD

34

/11

b/1

9/4

5/

HL

A-D

R P

E

CD

73

AP

C

CD

44

PE

CD

10

5 P

er-C

P-C

Y5

.5

CD

90

FIT

C

CD

73

AP

C

CD

44

PE

CD

10

5 P

er-C

P-C

Y5

.5

FS Peak

Page 91: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

81

Figure 9. Flow cytometry analysis of cord samples of CT16 at p2, p5, and p10 (Figure

9A), CT24 at p2, p5, and p10 (Figure 9B) and CT15P7 (Figure 9C) showing cell surface

expression of hematopoietic makers and the stromal makers CD44, CD73, CD90, and

CD105 at each time point.

Page 92: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

82

Figure 10. Flow cytometry analysis of cord samples (n=20) at early, mid and late

passages illustrates decreasing expression of hematopoietic markers with passaging while

exhibiting constant expression of CD44, CD90, and CD73. Hematopoietic markers

significantly decrease from p2 to p10 and CD105 expression is significantly lower from

p5 to p10. Values are ± SE. *P<0.05, **P<0.01.

Figure 10: Percent of cells illustrating expression of hematopoietic and stromal

markers at early, mid, and late analysis for 20 samples analyzed at each passage.

Page 93: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

83

Figure 11: Percent of cells illustrating expression of hematopoietic and stromal

markers at early, mid, and late analysis for 40 samples analyzed at early passage

and 20 cord tissues analyzed at mid and late passages.

Figure 11. Flow cytometry analysis of cord samples at early (n=40), mid (n=20) and late

passages (n=20) illustrates decreasing expression of hematopoietic markers with

passaging while exhibiting constant expression of CD44, CD90, and CD73. CD105

expression is significantly decreased when comparing p2 to p10 and p5 to p10. Values

are ± SE. *P<0.05.

Page 94: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

84

3.3 MSC Wound Healing Efficiency

Cultures of MSCs from different donors are being used in animal models as well as many

clinical trials, however, the cells have shown to be heterogeneous in nature, exhibiting

variations in their therapeutic efficacy. The therapeutic capacity of MSCs have been

proposed to be due to the secretion of various cytokines and chemokines. These trophic

factors have been shown to affect migration, proliferation, and survival of the cells

surrounding the wound (Maxson, Lopez, Yoo, Danilkovitch-Miagkova, & Leroux, 2012).

Others have suggested that the expression levels of a subset of key genes are responsible

for the immune suppressive and inflammation modulating capabilities of the cells, such

as hemeoxygenase 1, cyclooxygenase 2, IL1 receptor antagonist, and TGF-β1. However

it has recently been shown that in BM-MSCs, these markers are not significantly

correlated with the ability of the cells to attenuate the inflammatory response. Several

recent studies have suggested that a strong significant correlation can be found with the

gene TSG-6. BM-MSCs that express high mRNA levels of TSG-6 show better efficacy in

various animal models. The potent anti-inflammatory effects of MSCs have been

suggested to be due to TSG-6 protein secretion, and various mechanisms of action have

been proposed. TSG-6 has been suggested to bind to hyaluronic acid as well as inter-

alpha-inhibitor, interacting through the CD44 receptor found on macrophages, ultimately

decreasing the nuclear translocation of NF-kB through TLR-2, hence reducing the

inflammatory response (Choi et al., 2011). Respectively, TSG-6 was suggested to be a

correlative gene that may be used as a marker of the in vivo efficacy of MSCs. To our

knowledge, there is limited information regarding the correlation between TSG-6 and the

cytokine secretion profile of UCT-MSCs. Consequently, we aimed to test the levels of

Page 95: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

85

TSG-6 mRNA in a subset of UCT-MSC samples and examine whether this is closely

correlated with the cytokine secretion profiles as well as wound healing capabilities of

UCT-MSCs.

3.4 TSG-6 Expression

Total RNA was isolated from each sample and RNA integrity was analyzed from a subset

of cord samples (P4) (n=15) using Agilent RNA ScreenTape Assay (Agilent

Technologies, Canada) (Table 6). All of the samples analyzed had a RNA integrity

number (RIN) greater than 9.3, indicating high integrity RNA. We noticed that the

expression levels of TSG-6 mRNA varied widely among the samples, where some UCT-

MSCs expressed high levels of TSG-6 mRNA, and others showed a markedly decreased

expression (Figure 12). Not all of the samples analyzed for TSG-6 mRNA expression

levels contained data pertaining to maternal age and newborn weight. However, for the

cord tissue samples for which this information was available, no correlation between

TSG-6 mRNA expression and maternal age (Figure 13A) or the weight of the newborn

(Figure 13B) was found.

Page 96: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

86

Table 6: RNA integrity analysis for a subset of UCT-MSCs analyzed for TSG-6

mRNA expression levels.

Table 6. Each MSC population that was analysed by real time RT-PCR was tested for the

integrity of the RNA samples extracted and the ratio of 28S:18S ribosomal RNA.

Figure 12. Real-time RT-PCR assays for human-specific mRNA for TSG-6. MSCs from

different cord samples at passage 4 express varying levels of TSG-6. Values are

normalized by standard curve for hGAPDH and for CT34P4. Values are ± SE.

Sample RIN

CT16P4 9.7

CT17P4 9.4

CT20P4 9.6

CT21P4 9.7

CT26P4 9.3

CT30P4 9.4

CT33P4 9.3

CT29P4 9.6

CT34P4 9.9

CT35P4 9.7

CT15P7 9.3

Figure 12: Variable TSG-6 mRNA expression among a subset of UCT-MSC

samples.

Page 97: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

87

Figure 13: TSG-6 mRNA correlation with maternal age and newborn weight.

A.

B.

Figure 13. Maternal age (Figure 13A) and newborn weight (Figure 13B) are not

correlated with TSG-6 mRNA levels.

y = -0.0477x + 1.9153

R² = 0.281

-0.5

0

0.5

1

1.5

2

0 5 10 15 20 25 30 35 40 45

Maternal Age

Rel

ativ

e TS

G-6

mR

NA

Exp

ress

ion

Lev

el

y = -0.0001x + 0.9088

R² = 0.0091

0

0.2

0.4

0.6

0.8

1

1.2

1.4

1.6

1.8

2

0 500 1000 1500 2000 2500 3000 3500 4000 4500

Newborn Weight (g)

Rel

ativ

e TS

G-6

mR

NA

Exp

ress

ion

Lev

el

Page 98: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

88

3.5 Cytokine Secretion Analysis

Previous studies on MSCs from various sources has yielded insight into the secreted

cytokines and has suggested that the trophic factors released by the cells depends on the

tissue of isolation, but does not vary widely from donor to donor among identical

isolation sources (Park et al., 2009). To our knowledge, limited insight is available on the

trophic factors secreted by UCT-MSCs. In our study, a comparison was done between

three cord samples, CT15P8 showing low TSG-6 mRNA expression, CT24P8 illustrating

no TSG-6 mRNA expression, and CT16P8 illustrating a high TSG-6 mRNA expression.

A cytokine antibody array kit was used to analyse the expression of 55 proteins in the

conditioned medium of three respective samples. Each array was done in duplicate, and

the results of those experiments are plotted separately (Figure 14). A threshold of 0.3 was

chosen to discriminate between cytokines secreted at high and low amounts. The results

obtained illustrate that even though there were slight variations in the cytokines secreted

for each MSC sample, the cytokines secreted at high levels were the same among the

three samples. Activin A, ANG, IGFBP-3, IL-8, PTX-3, TIMP-1, thromobosondin-1, and

urokinase-type uPA were highly expressed in all three samples, while DPPIV and

PDGFAA was high only in CT15 (Figure 14A), and angiopoietin 1 and HGF was highly

expressed only in CT16 (Figure 14B). GM-CSF and serpin F1 were found to be highly

expressed only for CT15 and CT16. No cytokines were found to be uniquely expressed

for CT24 only (Figure 14C).

Page 99: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

89

Figure 14: Cytokine secretion profiles of CT15 expressing low TSG-6 mRNA, CT16

expressing high TSG-6 mRNA, and CT24 illustrating no TSG-6 mRNA expression.

A.

B.

Page 100: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

90

C.

D.

Figure 14. Analysis of CT15P8 having low TSG-6 mRNA expression (Figure 14A),

CT16P8 illustrating high TSG-6 mRNA expression (Figure 14B), and CT24P8 showing

no TSG-6 mRNA expression (Figure 14C) indicates similar cytokine secretion profiles of

cord units expressing low vs. high TSG-6 mRNA levels. Figure 14D shows media only.

Values are ± SE.

Page 101: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

91

3.6 Murine Excisional Wound Healing

Diabetes is a systemic disease that affects the whole body, mainly characterized by an

inability of beta cells to sufficiently supply the body with insulin and hence adjust for

increased glucose concentrations (Kahn, Cooper, & Del Prato, 2014). Type II diabetes is

characterized by insulin resistance, declining insulin production, and eventual pancreatic

beta-cell failure (Kahn et al., 2014). One of the complications arising from diabetes

includes chronic non-healing wounds. There have been many animal models that have

been used to replicate human physiology in diabetes. However, many rodent models do

not effectively replicate human wound healing, which is characterized by re-

epithelization and granulation tissue formation. One of the factors responsible for this

discrepancy is the significant difference between murine and human wound healing. Mice

heal mainly by contraction, due to a subcutaneous panniculus carnosus muscle layer,

whereas humans heal by granulation tissue formation and re-epithelization (Wong, Sorkin,

Glotzbach, Longaker, & Gurtner, 2011). Our lab has developed a murine excisional splint

wound healing model, which closely mirrors human wound healing. This is done by

creating a full thickness wound, which effectively removes the panniculus carnosus layer,

on the back of mice. Splints are then centered and fixed on the skin using an adhesive and

four sutures. The main goal of our study was to compare the wound healing efficiencies

of UCT-MSCs isolated from CT15P8, showing low levels of TSG-6 mRNA expression,

to cells isolated from CT16P8, expressing high levels of TSG-6 mRNA. Mice

administered only media were used as controls. Digital pictures were taken on day 0, 3, 7,

10, and day 14 (Figure 15A). Upon analysis of digital pictures, no significant differences

were noticed between mice receiving CT15 or CT16. However, there was a significant

Page 102: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

92

difference between wound closures for CT15 and CT16 treated mice versus controls at

most of the time points studied. CT-treated mice showed a progressive closure of the

wound lesion, with 72% ±2.64 of the original wound by day 3, 53% ±2.122 by day 7,

39% ±5.54 by day 10, and 28% ±3.35 by day 14. A similar trend was seen for CT16-

treated mice, with 72% ±5.63 of the wound closed by day 3, 61% ± 8.93 closed by day 7,

47% ±6.23 closed by day 10, and 32% ±2.15 closed by day 14 (Figure 15B). Histological

analysis of CT16 (Figure 16) and CT15 (Figure 17) illustrated complete wound closure,

whereas analysis of digital photos only showed a 28% ±3.96 and 32% ±2.15 wound

closure by day 14, respectively. Excisional wounds treated with CT15 and CT16 showed

a more robust and faster rate of healing when compared with control mice. Some control

mice (2 out of 4 mice studied) did not show any healing of the wound bed (Figure 18A),

while two mice illustrated complete re-epithelisation of the wound bed with small blood

vessels visible within the wound (Figure 18 B,C). Additionally, some fat cells could be

seen within the newly formed tissue of a control mouse. However, a thick distinct keratin

layer was seen for control mice that had healed. As a result, even though the wound was

fully closed, this suggests that there may be more scarring in the control mice when

compared with CT15 and CT16 treated mice. All mice for both cord tissues illustrated

complete wound closure. Histological examination of the wounds from CT16, CT15 and

control mice disclosed variations in the thickness of granulation formation, presence and

number of newly formed blood vessels, as well as fat within the new tissue. For all the

animals studied, the wound bed was not developed enough for there to be striations of the

skin, specifically fully stratified squamous epithelium. Many chronic inflammatory cells,

such as lymphocytes as well as neutrophils, could be seen, as detected by cell

Page 103: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

93

morphology. Fibroblasts could be seen at the top of the wounds. Immunohistochemistry

illustrated that CT16-treated mice showed a trend towards a greater number of cells

positive for Ki67 than for CT15-treated wounds (Figure 19B). A greater amount of

collagen IV, appearing in the epidermis and dermis, as well as β-catenin positive sections

were noticed for CT16 (Figure 19 B,C) rather than CT15 treated wounds (Figure 20).

Collagen IV, cytokeratin 6, and β-catenin staining was seen around the around the wound

margins for CT15-treated mice (Figure 20). Even though some muscle could be seen in

the histology sections for CT16 and CT15- treated mice, this was most likely not newly

formed muscle.

Page 104: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

94

Figure 15: Wound closure analysis.

A. Control CT15P8 CT16P8

Day 0

Day 3

Day 7

Day 10

Day 14

B.

Page 105: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

95

Figure 15. Digital pictures (Figure 15A) illustrate wound closure of CT15 (having low

TSG-6 mRNA expression) and CT16 (having high TSG-6 mRNA expression) when

compared with control mice at days 0, 3, 7, 10, and 14. Wound area analysis (Figure 15B),

done by comparing the % of the original wound on day 0 with each respective day

analyzed, illustrates that CT15 and CT16 had a similar rate of healing and show a greater

wound closure when compared with control mice. aa, P<0.01 for CT15 versus control;

aaa, P<0.001 for CT15 versus control, b, P<0.05 for CT16 versus control, bb, P<0.01 for

CT16 versus control.

Page 106: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

96

Figure 16: Wound bed histology for CT16-treated mice.

Page 107: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

97

Figure 16. H&E staining of wounds treated with CT16. (A) All mice (n=5) illustrated

complete wound closure by day 14. Arrows in Figure 16A indicates original wound edges

(H&E, x1.25). (B) Mice illustrated different thickness of granulation tissue formation, as

well as blood vessels and some fat cells that have formed within the wound lesion. The

wound beds were infiltrated with chronic inflammatory cells (H&E, x5, inset, x20). (C)

Fibroblast cells can be noticed at the top of the wound. Arrows indicate fibroblast cells.

(H&E, x20). Ke= keratin, AT= adipose tissue, V=blood vessels, GT=granulation tissue.

Images show one representative animal.

Page 108: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

98

Figure 17: Wound bed histology for CT15-treated mice.

Page 109: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

99

Figure 17. H&E staining of wounds treated with CT15. (A) All mice (n=5) illustrated

complete wound closure by day 14. Arrows in Figure 17A indicates original wound edges

(H&E, x1.25). (B) Mice illustrated different thickness of granulation tissue formation, as

well as blood vessels and some fat cells that have formed within the wound lesion. The

wound beds were infiltrated with many inflammatory cells (H&E, x5, inset, x20). (C)

Fibroblast cells can be noticed at the top of the wound. Arrows indicate fibroblast cells

(H&E, x20). Ke= keratin, AT= adipose tissue, V=blood vessels, GT=granulation tissue.

Images show one representative animal.

Page 110: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

100

Figure 18: Wound bed histology for control mice.

Page 111: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

101

Figure 18. H&E staining of control mice. (A) Some mice (n=2) did not show wound

closure after 14 days. Arrows in Figure 18A indicates original wound edges (H&E,

x1.25). (B) A subset of mice (n=2) showed complete wound closure. Arrows in Figure

18B indicates original wound edges (H&E, x1.25). (C) Mice illustrated granulation tissue

formation, with blood vessels and some fat cells present within the wound lesion. The

wounds bed were infiltrated with many chronic inflammatory cells, such as lymphocytes.

Fibroblast cells can be seen at the top of the wound (H&E, x5, inset, x40). Ke= keratin,

AT= adipose tissue, V=blood vessels, GT=granulation tissue. Images show two

representative animals.

Page 112: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

102

Figure 19. (A) Confocal microscopy illustrated that there was greater expression of

collagen IV around the wound edge for CT16-treated mice with compared with CT15-

treated mice. (B) A trend toward a greater number of Ki67 positive cells was also noticed,

based on observations of the sections. Arrows indicate cells stained positive for Ki67. (C)

β-catenin staining was also seen at the edge of the wound. Arrow indicates delineation

between wound bed and wound edge. Scale bar= 50 µm.

Figure 19: Wound bed immunohistochemistry for CT-16 treated mice.

Page 113: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

103

Figure 20. (A) Confocal microscopy illustrated the expression of collagen IV, noticed

around the wound edge for CT15 treated mice. Arrow indicates boundary between wound

bed and wound edge. (B) Cytokeratin 6 staining was also seen around the wound edge.

(C) β-catenin staining was seen near the wound lesions for CT15-treated mice. Scale bar=

50 µm.

Figure 20: Wound bed immunohistochemistry for CT-15 treated mice.

Page 114: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

104

Chapter Four: Discussion

Page 115: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

105

4. Discussion

4.1 Changes in MSC profile with culture

MSCs have emerged in recent years as cells that can be used as ideal therapeutic

candidates for tissue repair, various diseases and pathologies. Traditionally, MSCs were

isolated from hematopoietic sources, such as bone marrow, peripheral blood, and

umbilical cord blood. They have also been isolated from parenchymal non-hematopoietic

tissues such as muscle, fat, or liver (Tolar, Le Blanc, Keating, & Blazar, 2010). However,

isolation from some sources, such as the bone marrow, can be painful and lead to possible

infection. Other sources, such as peripheral blood, contain only a very small number of

MSCs. As a result, our lab has used an explant method of MSC isolation from umbilical

cord tissue. This method is cost effective, simple, and yields a large number of expanded

cells over a few passages. Moreover, it does not impact the donor negatively or incur any

pain or risk of infection, as cord tissue is usually discarded as medical waste. In our study,

the cord samples were collected from across Canada, from both vaginal and caesarean

delivery, from a maternal age range of 21-41 years old.

It has been suggested that all cultures of MSCs are homogenous, while others have shown

differences in immunophenotype and wound healing capabilities between patients. MSCs

which are isolated and expanded under different culture conditions differ in their

properties and therapeutic potential (Prockop, 2009). MSCs expanded from various

sources have been used in many animal models as well as some clinical trials. Earlier

passages have been documented to be ideal, as some reports suggest that ex vivo

prolonged culture may be correlated with genomic alterations, changes in cell

morphology as well as surface marker profile expression (Otte, Bucan, Reimers, & Hass,

Page 116: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

106

2013; Y. Wang et al., 2013). Phenotypic characterization studies have shown that UCT-

MSCs express CD73, CD90 and CD105 in 99% of the cells in P1, however, this

decreases slightly during passaging until P11 (Otte et al., 2013). Others have suggested

that passaging does not have an effect on the immunophenotypic profile of UCT-MSCs,

exhibiting the ability to maintain a stable expression of stromal markers even at higher

passages (Shi, Zhao, Qiu, He, & Detamore, 2015). UCT-MSCs have been shown to be

negative for markers indicative of hematopoietic cells (Shi et al., 2015). As a result, we

sought to address the question of patient to patient differences and also passage variation

for surface epitopes in MSC populations isolated from 40 umbilical cord tissue samples

analyzed at p2, and 20 analyzed at p5 and p10, using the protocol developed in our lab.

The data obtained demonstrates that by using our explant isolation procedure as well as

culture conditions, a heterogeneous population of cells are isolated at early passages,

including both stromal as well as hematopoietic cells. However, with passaging, the

hematopoietic cells are lost, suggesting that the media conditions are specific for MSCs.

We have also demonstrated that CD105 expression is lower when compared with CD44,

CD73, and CD90, and that there is a significant decrease in CD105 expression with

passaging. The reduction in surface expression of CD105 has also been shown with cord

blood derived MSCs (Gaebel et al., 2011). CD105, also known as Endoglin, is a type I

membrane glycoprotein belonging to the TGF-β complex. It has important functions in

angiogenesis, proliferation, and adhesion (Mark et al., 2013). Additionally, TGF-β and

Endoglin have been suggested to play important roles in wound healing, regulating

different cellular functions, such as recruitment of stem cells to the wound (Valluru,

Staton, Reed, & Brown, 2011). Endoglin is also decreased on MSCs with an increased

Page 117: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

107

differentiation potential (Valluru et al., 2011). Cord blood derived MSCs selected for

high CD105 surface expression were seen to enhance heart performance and reduce scar

formation (Gaebel et al., 2011). This illustrates that MSC populations that show a higher

expression of CD105 may be more effective in treating cardiac ischemia (Gaebel et al.,

2011). Also, the percentage of MSCs expressing Endoglin have been shown to be

increased during brain injury (Valluru et al., 2011). Taken together, this could suggest

that MSCs from earlier passages, expressing higher levels of CD105, may have better

wound healing properties.

Although MSCs have been shown to have many positive effects in various animal models

of injury, such as stroke, type I and type II diabetes, and GvHD, the engraftment of MSCs

and the differentiation into different cell types at the wound site is usually low. As a

result, several studies have looked at various optimization strategies to increase the

efficacy of MSC based therapies. This includes transgenic approaches in which MSCs

over-express proteins of interest, preconditioning of MSCs by in vitro priming (for

example culture in hypoxic conditions), and attempting to shift MSC distribution in the

body by altering cell surface receptors (J. Wagner, Kean, Young, Dennis, & Caplan,

2009). However, despite these various optimization approaches, MSCs alone have been

shown to have significant wound healing capabilities. This has mainly been attributed to

paracrine and trophic factors released by the cells, having pro-regenerative roles in anti-

inflammation, angiogenesis, immunomodulation, and antifibrosis (J. Wagner et al., 2009).

4.2 Paracrine Signalling

MSCs rarely engraft into tissues or generate newly differentiated cells, thus attributing

their mode of action on surrounding cells and tissues to a multitude of secreted trophic

Page 118: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

108

factors (Caplan & Dennis, 2006). Key cytokines that have been identified as being

secreted by MSCs include VEGF-A, MCP-1, FGF-1, MMP-8, and MMP-9 (Briquet et al.,

2010). Other cytokines, adhesion molecules, and metalloproteinases that have been

detected in human BM-MSC conditioned medium include IL-6, IL-8, vascular cell

adhesion protein 1 (VCAM-1), intercellular adhesion molecule 1 (ICAM-1), MMP-2,

MMP-3, MMP-13 and TIMP-4 (Briquet et al., 2010). Various other studies have also

looked at the secretome of different donor derived BM-MSCs, applying a similar assay

that was used in our study, and found that the key cytokines constitutively expressed

included IL-6, IL-8, MCP-1, Chemokine (C-C motif) ligand 5 (CCL5), chemokine (C-X-

C motif) ligand 1 (CXCL1), INF-γ, IL1-α, TGF-β, GM-CSF, and angiogenin (Hwang et

al., 2009). The same study did not find constitutive expression of MIP-1α, IL-2, IL-4, IL-

10, IL-12, and IL-13, however showed that BM-MSCs were very similar to CB-MSCs in

regards to the cytokines secreted (Hwang et al., 2009). The findings in our study are in

accordance with these sources, as highly expressed cytokines from three different cord

tissue samples included activin A, MCP-1, IL-8 and others such as TIMP-1, angiopoietin,

and GM-CSF. In the context of wound healing, VEGF is a crucial promoter of cell

proliferation, migration and chemotaxis, and angiogenesis (Guo & Dipietro, 2010). Along

with VEGF, angiopoietins also have effects on the vascular endothelium, however unlike

VEGF, angiopoietins do not regulate endothelial cell proliferation (Werner & Grose,

2003). In diabetic mice, wounds exhibit an imbalance of VEGF-A and angiopoietins,

specifically showing higher levels of angipoietin-2 (causing vessel destabilization and

remodeling) and lower levels of VEGF-A (Werner & Grose, 2003). Conversely,

angiopoietin-1 has been shown to stabilize blood vessels in healing wounds (Werner &

Page 119: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

109

Grose, 2003). MCP-1, a chemokine known as macrophage chemoattractant protein-1, is a

major chemoattractant for monocytes/macrophages and influences the gene expression of

murine macrophages (Werner & Grose, 2003). It has also been shown to attract T cells

and mast cells to the wound site and is a major bioactive chemoattractant for neutrophils

(Werner & Grose, 2003). High amounts of IL-8 has been found to stimulate inflammation

and inhibit wound contraction (Werner & Grose, 2003). In vitro, wound repair was

impaired by IL-8 due to inhibition of keratinocyte proliferation and collagen lattice

contraction by fibroblasts, however others have found stimulatory effects of IL-8 on

keratinocyte proliferation (Werner & Grose, 2003). GM-CSF, granulocyte-macrophage

colony stimulating factor, was another highly expressed cytokine found in our study. It is

mitogenic for keratinocytes and stimulates migration and proliferation of endothelial cells,

thus enhancing neovascularization and granulation tissue formation (Werner & Grose,

2003). TIMPs (tissue inhibitors of matrix metalloproteinases), also found to be highly

expressed in our study, are capable of inhibiting all of the MMPs, endopeptidases which

have diverse roles in wound healing, such as degradation of the extracellular matrix,

facilitating migration of cells to the centre of the wound, eliminating damaged protein

(Muller et al., 2008). However, their expression in chronic wounds is reduced (Muller et

al., 2008). Additionally, the levels of MMPs usually decrease in normal wound healing,

but chronic wounds have been shown to have an increased production of pro-

inflammatory cytokines and proteases such as MMPs (Muller et al., 2008). Chronic

wounds are thus characterized by a reduced expression of TIMPs, usually produced by

fibroblasts, and this results in an imbalance of the ratio of TIMPs and MMPs in wounds,

ensuing in poor healing by the breakdown of too many components of the extracellular

Page 120: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

110

matrix and by the inhibition of growth factors that are essential for tissue synthesis

(Muller et al., 2008). Thus, the presence of TIMP-1 in the conditioned medium of MSCs

could restore this imbalance. Activin A, a member of the TGF-β superfamily, regulates

various aspects of cell growth and differentiation, such as inducing the expression of

growth factors in dermal fibroblasts, which can thus stimulate keratinocyte proliferation

in a paracrine manner (Werner & Grose, 2003). The important role of Activin A in the

timely formation of granulation tissue and scar has been shown with transgenic mice

overexpressing soluble activin antagonist in the epidermis (Werner & Grose, 2003).

Among the three cord tissue samples studied, we found that the cytokine secretion profile

was similar and comparable, thus suggesting that MSCs isolated from any cord tissue can

be used as a potential therapeutic agent, without major differences in the therapeutic

capacity of the cells. Our finding is consistent with other reports which have

demonstrated that MSCs derived from different sources, such as human placenta, cord

blood and bone marrow have a common cytokine expression pattern, including

expression of macrophage migration inhibitory factor (MIF), IL-8, serpin E1, Gro-α, and

IL-6 (Hwang et al., 2009). It should be noted that contradictory to these findings, a subset

of studies have also noticed differences between the secretome profiles of MSCs from

various anatomical locations. For example, MSCs derived from the bone marrow, adipose

tissue, and dermal tissue showed similar expression of VEGF-A, angiogenin, FGF basic,

and nerve growth factor, however, MSCs derived from adipose tissue expressed

significantly higher levels of IGF-1, VEGF-D, and IL-8 (Hsieh et al., 2013). Our study

suggests that the MSCs from cord tissue are similar, and that we are isolating comparable

cell populations from different patient samples. However, it should be taken into account

Page 121: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

111

that there have been reported differences between cytokine secretion analysis using a

proteome profiler, like we used in our study, and PCR analysis (Hwang et al., 2009). This

suggests that future studies may look to address if there are differences in cytokine

expression between different cord samples by also using PCR analysis. Other techniques

that could be used to verify the results obtained using the cytokine array could be by

Western blot analysis.

4.3 Diabetes Complications

MSCs have been used as potential therapeutic agents in many animal models. Particularly,

it has been shown that growth factor deficiencies have been accounted for as an important

factor contributing to diabetic wound healing impairment. Many complications can arise

from diabetes, including obesity, various metabolic abnormalities such as impaired

function of many cells types, inflammation, alterations in adipokines, cardiovascular

disease characterized by atherogenic dyslipidemia and increased levels of free fatty acids,

changes in thrombosis and fibrinolysis, as well as non-healing wounds (Pandey, Chawla,

& Guchhait, 2015). Healing requires an orderly, but overlapping, progression of three key

events, specifically inflammation, proliferation and angiogenesis and remodelling.

However, in those with diabetes, these events are often prolonged, lacking, or faulty. This

is mainly due to an impairment of cytokine production by local cells, such as fibroblasts

and endothelial cells, and also reduced angiogenesis (Wu et al., 2007). Additionally,

aging has shown to cause a delay in wound healing, and every stage of the wound healing

process has shown be affected in advanced age (Guo & Dipietro, 2010). This includes

enhanced platelet aggregation, increased secretion of inflammatory mediators, delayed

infiltration of macrophages and lymphocytes, impaired macrophage function, decreased

Page 122: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

112

secretion of growth factors, delayed re-epithelization, delayed angiogenesis and collagen

deposition, reduced collagen turnover and remodelling, and decreased wound strength

(Guo & Dipietro, 2010). Effective treatment options are still lacking, as currently

available options are able to achieve only a 50% healing rate (Wu et al., 2007). UCT-

MSCs have been proposed to be the optimal therapeutic cells for enhancing tissue repair

due to ease of isolation as well as the multiple paracrine factors that they release. In order

to study the effects of the cells on cutaneous wounds, our lab has developed a db/db

excisional wound mouse model. We use a splint model because it prevents healing of

murine skin by contraction, which is the main mechanism of wound healing in mice, and

allows the wound to heal by re-epithelization and granulation tissue formation. Full-

thickness wounds also ensure removal of the panniculus carnosus layer, which is present

in loose skinned species, such as mice, and is attached to the base of the dermis

(Davidson, 2001). Wounds in splinted mice have shown to have an increased time to

complete wound closure, as well as increased granulation tissue formation when

compared to non-splinted mice (Galiano et al., 2004).

4.4 TSG-6 and Wound Healing

Even though numerous clinical trials have been done with MSCs, there is a large

variability in the quality of MSCs isolated due to heterogeneity of cell cultures (R. H. Lee

et al., 2014). Recent data has suggested that TSG-6 could be used as an informative

biomarker to predict the in vivo efficacy of the cells. However, MSCs derived from BM,

rather than UC, were used in these studies. As a result, we were interested in knowing if

there are variations in wound healing capacities among cord tissue samples dependent on

their TSG-6 mRNA expression levels of TSG-6. Ultimately, we wanted to know if TSG-6

Page 123: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

113

gene expression could be used as a marker in our model to reliably predict the wound

healing capacity of the cells in vivo. MSCs from two different cord tissues, CT16

expressing high levels of TSG-6 mRNA, and CT15, expressing low levels of TSG-6

mRNA, were topically applied to excisional wounds in mice and wound lesions were

monitored over a period of 14 days by taking digital pictures. We found that between the

two cord samples, there was no significant difference in wound closure although both

resulted in a statistically significant faster wound closure compared to controls.

Histological analysis illustrated that the new tissue extended across the wound and that

blood vessels, both large and small (depending on the mouse), could be noticed within

the wound bed for both CT16 and CT15-treated wounds. There were variations in the

number and size of blood vessels for each mouse but no difference attributed to the cord

tissue used were noted. In some mice, fat cells could also be noticed within the newly

formed tissue. The granulation tissue is characterized by small capillaries and fibroblasts.

Many late stage inflammatory cells could be noticed within the wound lesion for both

animals, such as lymphocytes. Fibroblasts could be noticed within the wound bed, but

also at the top of the wounds, most likely trying to make collagen to replace lost tissue.

Since the basement membrane between the epidermis and dermis is damaged in full

thickness skin wounds, the wound cannot healing by re-epithelization alone and

fibroblasts are needed to make new ECM (Stroncek JD, 2008). Some dark pieces of

tissue could be noticed next to the wound, and sometimes even extending into the wound.

This is probably scab that has formed, containing dead cells. However, complete healing

of the skin, specifically, formation of fully stratified squamous epithelium, was not seen.

Unwounded skin has three main layers, the epidermis which is composed of a squamous

Page 124: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

114

epithelium which proliferates and produces a layer of keratin, which is constantly shed

(Barbara Young, 2014). The layer below is the dermis, which is tightly bound to the

epidermis by a basement membrane (Barbara Young, 2014). It supports the epidermis

and is composed of fibrous and fibroadipose tissue (Barbara Young, 2014). Residing in

this layer are blood vessels, sensory receptors, and nerves (Barbara Young, 2014). The

layer beneath the dermis is called the hypodermis, and it contains larger vessels which

support and drain the blood vasculature (Barbara Young, 2014). This layer extends to the

underlying connective tissue. Additionally, in later stages of the healing process,

myofibroblasts undergo apoptosis, the granulation tissue, which is initially rich is cells,

becomes scar tissue characterized by an excess of ECM and limited cell numbers

(Stroncek JD, 2008). Capillary density also is reduced, and the wound becomes paler,

rather than the pink colour that is characteristic of the earlier phases of wound healing

(Stroncek JD, 2008). However, these structures were not noticed to have formed for the

time frame used in our study.

Some control mice did not heal at all (2 out of 4). A thin collagen section was observed,

as a result, there was no scaffold for epithelial cells to migrate across the wound.

However, two out of four control mice did illustrate healing and new tissue formation,

however the quantity as well as quality of granulation tissue differed from MSC treated

wounds. Small blood vessels as well as fat could be noticed within the wound bed and

next to the wound lesion of these mice, many inflammatory cells could be noticed, which

have probably migrated from the surrounding vessels. Additionally, neovascularisation

was reduced in control mice used in this study. This may be due to the fact that MSCs

secrete many cytokines that play a role in angiogenesis. Such as VEGF, as well as FGF2,

Page 125: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

115

both promoting angiogenesis in wounds (P. Martin, 1997) In genetically diabetic mice, it

has been shown that VEGF is not expressed at wound lesions, hence impairing healing

and neovascularisation (P. Martin, 1997). Cytokine secretion analysis using conditioned

medium from three cord tissue units illustrated that the highly secreted cytokines are

similar between the three samples. Generally, MSCs secrete high levels of growth factors

from the chemkoine family and those associated with angiogenesis (Potian et al., 2003).

The factors that were found to be highly expressed in our study are known to have roles

in wound healing. Activins, members of the TGF-β superfamily of proteins, have shown

to regulate various aspects of cell growth and differentiation in many tissues and organs

(Werner & Grose, 2003). Angiopoietin along with VEGF act on the vascular endothelium,

however unlike VEGF, angiopoietins do not regulate endothelial cell proliferation

(Werner & Grose, 2003). Rather angiopoietin-1 causes stabilization of blood vessels,

whereas angiopoietin-2 causes vessel destabilization and remodelling (Werner & Grose,

2003). Other highly expressed factors in our study, such as IL-8, have been found to be

important for neutrophil chemoattraction, stimulation of re-epithelization and

keratinocyte proliferation (Werner & Grose, 2003). Our study did not look at cell

engraftment because we used a model that was equivalent to a non-matched allogeneic

transplant model. We chose this model to specifically study paracrine signalling effects

without confounding factors related to engraftment or differentiation. Initially, it was

thought that MSCs accelerate the wound healing process due to direct participation in the

repair process and incorporation into the regenerated tissue (Shin & Peterson, 2013).

Nonetheless, previous studies have shown that MSCs are not maintained within the

wound bed, and that there is a rapid decline in the number of cells in the wound bed (Shin

Page 126: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

116

& Peterson, 2013). It is now known that MSC act indirectly, facilitating the host repair

process (Shin & Peterson, 2013). Hence, our model uses mice with a functional immune

system, therefore we are mainly looking at the cytokine effects of MSCs rather than cell

engraftment or differentiation.

Observational analysis of immunohistochemistry staining illustrated there was a trend

toward more Ki67 positive cells around the wound bed in CT16-treated mice, illustrating

greater cellular proliferation. The cells staining positive were not found on the newly

formed tissue, but rather in the adjacent or more distal areas from the wound. Staining

was found to be mostly distributed in the epidermis or dermis, rather than the basal layer

of the skin. Ki67 staining during wound healing has also been indicative of the activation

of keratinocytes around the wound lesion (Xu et al., 2012). During wound healing,

keratinocytes are first present at the leading edge of the wound, then migrate across the

wound bed to dissolve the fibrin clot that has been created (P. Martin, 1997). There was

also more collagen IV positive areas that were noticed for CT16 rather than CT15, seen at

the wound margins. Collagen IV is a major protein of the basement membrane, and it

plays important roles in the maintenance of basement membrane integrity, has filtration

functions, and also stores growth factors (Poschl et al., 2004). Collagen IV staining was

expected due to the fact that reproduction of collagen in the dermal layer is an important

part of wound healing (Xu et al., 2012). Keratin 6 staining was found in small amounts

for mice treated with CT16. The keratin 6 gene is activated in cells within the hair

follicles surrounding the wound and are a source of keratinocytes. The keratinocytes

migrate up the hair shaft and form a proliferating pool of cells at the wound edge and then

migrate across the wound.

Page 127: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

117

Alpha-smooth muscle actin staining could not be found in mice treated with either CT15

or CT16. In the more advanced stages of healing, about a week after wound induction,

wound fibroblasts transform into myofibroblasts which have been shown to express

alpha-smooth muscle actin (P. Martin, 1997). These cells are vital for generating strong

contractile forces and bringing the edges of the wound closer together (P. Martin, 1997).

However, in our study no staining for alpha-smooth muscle, a main constituent of large

blood vessels, could be detected in the newly formed tissue. Muscle could be noticed in

the basal layer of the wound for a small number of mice by haematoxylin and eosin

examination. This appears to be skeletal muscle in the transverse plane. Nevertheless, it is

most likely not newly formed muscle, as the tissue is not developed enough for new

muscle to have been generated. β-catenin staining was noticed around the wound edges,

but more positive areas could be seen for CT16, rather than CT15-treated wounds. β-

catenin levels are usually elevated during the proliferative phases of wound healing, and

have been shown to mediate fibroblast proliferation (Cheon et al., 2006). It has also been

suggested to maintain cells in a less differentiated state during wound healing,

particularly mesenchymal progenitor cells (Cheon et al., 2006).

Despite our histological examination that confirmed full wound closure, analysis of

digital pictures did not yield full closure by day 14. This is due to the fact that in some

mice, a large white/yellow substance formed in the middle of the wound by day 3 and

remained thereafter, hence making accurate measurement of wound closure difficult. This

was seen for both control mice as well as MSC treated mice. A possible explanation for

the formation of this could be the accumulation of dead cells in the centre of the wound.

This could also be necrotic fibrin, alternatively known as fibrin eschar (Joseph M

Page 128: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

118

McCulloch, 2010). Necrotic fibrin can be identified by a yellow or tan appearance, which

is in accordance with the observed colour of the yellow pellet formed in the middle of the

wound (Joseph M McCulloch, 2010). The technique of analyzing serial pictures of the

same animal to assess wound closure (macroscopic analysis) does not always represent a

reliable measurement of the repair process, as factors such as inflammation or matrix

deposition cannot be seen visually (Ansell et al., 2014). As a result, histological analysis

is considered the gold standard method to obtain reliable and accurate information

(Ansell et al., 2014). It has been shown that incisional wounds show a greater correlation

between macroscopic wound examination and histology analysis (Ansell et al., 2014).

Also, the results obtained regarding digital analysis of wound closure are subjective and

interpretation may vary from person to person in delineating the margins of new tissue

formation and wound closure. Hence, there may be differences in wound closure

measurement when it is done by different interpreters. This obstacle occurred in our study,

where the results I obtained of wound closure, differed from the results that were

obtained when the analysis was done by a more experienced interpreter (Figure 21A vs.

Figure 21B). Interpretation of data in this thesis was done using the results obtained from

the more experienced interpreter in our lab to ensure accuracy, reliability and

reproducibility of the data. Additionally, two mice for CT15 on days 7, 10 and 14 were

removed from the analysis. This is due to fact that the wound bed contained excessive

amount of exudate, and hence an accurate measurement of wound closure could not be

made (Figure 22).

Page 129: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

119

Figure 21: Wound closure analysis illustrating different interpretations.

A.

B.

Figure 21. Wound closure analysis of digital photographs can be subjective. Figure 21A

is the analysis of wound closure done by a more experienced interpreter in our lab.

However, Figure 21B is the analysis of wound closure I obtained. Values are ± SE. a,

P<0.05 for CT15 versus control; aa, P<0.01 for CT15 versus control, b, P<0.05 for CT16

versus control, bb, P<0.01 for CT16 versus control.

Page 130: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

120

Day 0

Day 3

Day 7

Day 10

Day 14

Figure 22. CT15-treated mouse wound calculations excluded for days 7, 10 and 14 due

to copious exudate formation, hence making wound healing interpretations difficult.

Figure 22: CT15-treated mouse wound healing calculations excluded for days 7, 10,

and 14.

Page 131: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

121

5. Conclusion and Future Studies

MSCs have shown therapeutic potential in a plethora of in vivo animal models, and

positive effects, mainly using BM-MSCs, have also been documented in clinical trials.

Even though various tissues have been used for cell isolation, UCT represents an ideal

source due to the ease of collection and a lack of complications associated with cell

isolation. We have used an explant method to isolate MSCs from 20 different cord tissues,

and have shown that phenotypically, the cells look identical among all the patient

samples analyzed. A heterogeneous population of cells is first isolated, showing some

cells expressing markers indicative of hematopoietic cells, however, with passaging a

more homogenous population of MSCs is obtained. No variation in standard stromal

surface epitopes was noticed with passaging, with the exception of a significant decrease

of CD105 surface expression from early to late passages, although at p10 over 80% of the

cells were positive.

It has been suggested that the wound healing capacity of MSCs differs among various

cell populations, and that TSG-6 gene expression can be used to predict the in vivo

efficacy of the cells. Our results demonstrated that the relative expression of TSG-6

mRNA varied widely among a subset of 15 cord samples, however this variation did not

seem to correlate with the cytokine secretion profile of umbilical cord tissues. Moreover,

TSG-6 mRNA levels were not predicative of the wound healing capacity of UCT-MSC

preparations in a db/db excisional wound healing mouse model since both low and high

expressing UCT-MSC populations demonstrated similar rates of wound closure. Overall,

we can conclude from this study that umbilical cord tissue may be an ideal source of

Page 132: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

122

MSCs, as regardless of patient source or of the gene expression profile, cells seem to

possess the same positive therapeutic potential, which is ideal for clinical settings.

Future studies could look to document cell engraftment. As a result, differentiation

studies could also be done in the future, as this information would be pertinent to

engraftment studies. Perhaps TSG-6 could affect differentiation efficiency into

mesenchymal cell sources such as bone, adipose, and cartilage. Also, future studies could

be done to assess different MSC populations in regards to CD105 expression for wound

healing, comparing the efficacy of CD105 high and CD105 low MSC populations.

Additional studies might be warranted to investigate if other delivery systems, besides the

fibrin matrix used in this study, would be more beneficial for growth factor secretion as

well as cell survival. A number of different delivery systems exist which could be tested

in this model, for example collagen matrices as well as hydrogels, such as collagen-

pullulan hydrogels (Chen et al., 2012). Also, the stage of wound healing that was seen by

histological examination was not advanced. Future studies could perhaps allow healing to

occur for a longer duration of time (three weeks instead of two weeks), so that fully

stratified squamous epithelium might be formed. Lastly, the primary goal of this study is

to be able to apply UCT-MSCs for cell based therapies for cutaneous non-healing wounds

for diabetic patients in the future. Clinical trials could be carried out to test the potential

of UC-MSCs clinically, in diabetic patients with foot ulcers.

Page 133: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

123

6. References

AbD Serotec. (2015). Isotype controls in flow cytometry. Retrieved August 18, 2015,

2015, from https://www.abdserotec.com/flow-cytometry-isotype-controls.html

Aggarwal, S., & Pittenger, M. F. (2005). Human mesenchymal stem cells modulate

allogeneic immune cell responses. Blood, 105(4), 1815-1822. doi: 10.1182/blood-

2004-04-1559

Ankrum, J. A., Ong, J. F., & Karp, J. M. (2014). Mesenchymal stem cells: immune

evasive, not immune privileged. Nat Biotechnol, 32(3), 252-260. doi:

10.1038/nbt.2816

Ansell, D. M., Campbell, L., Thomason, H. A., Brass, A., & Hardman, M. J. (2014). A

statistical analysis of murine incisional and excisional acute wound models.

Wound Repair Regen, 22(2), 281-287. doi: 10.1111/wrr.12148

Armulik, A., Genove, G., & Betsholtz, C. (2011). Pericytes: developmental,

physiological, and pathological perspectives, problems, and promises. Dev Cell,

21(2), 193-215. doi: 10.1016/j.devcel.2011.07.001

Arno, A. I., Amini-Nik, S., Blit, P. H., Al-Shehab, M., Belo, C., Herer, E., . . . Jeschke, M.

G. (2014). Human Wharton's jelly mesenchymal stem cells promote skin wound

healing through paracrine signaling. Stem Cell Res Ther, 5(1), 28. doi:

10.1186/scrt417

Badiavas, E. V., & Falanga, V. (2003). Treatment of chronic wounds with bone marrow-

derived cells. Arch Dermatol, 139(4), 510-516. doi: 10.1001/archderm.139.4.510

Bara, J. J., Richards, R. G., Alini, M., & Stoddart, M. J. (2014). Concise review: Bone

marrow-derived mesenchymal stem cells change phenotype following in vitro

culture: implications for basic research and the clinic. Stem Cells, 32(7), 1713-

1723. doi: 10.1002/stem.1649

Barbara Young, P. W., Geraldine O'Dowd. (2014). Skin Wheater's Functional Histology

(Sixth ed.). Philadelphia, PA: Elsevier Ltd.

Bernardo, M. E., Locatelli, F., & Fibbe, W. E. (2009). Mesenchymal stromal cells. Ann N

Y Acad Sci, 1176, 101-117. doi: 10.1111/j.1749-6632.2009.04607.x

Bianco, P., Robey, P. G., & Simmons, P. J. (2008). Mesenchymal stem cells: revisiting

history, concepts, and assays. Cell Stem Cell, 2(4), 313-319. doi:

10.1016/j.stem.2008.03.002

Bieback, K., Kern, S., Kluter, H., & Eichler, H. (2004). Critical parameters for the

isolation of mesenchymal stem cells from umbilical cord blood. Stem Cells, 22(4),

625-634. doi: 10.1634/stemcells.22-4-625

Boroviak, T., Loos, R., Bertone, P., Smith, A., & Nichols, J. (2014). The ability of inner-

cell-mass cells to self-renew as embryonic stem cells is acquired following

epiblast specification. Nat Cell Biol, 16(6), 516-528. doi: 10.1038/ncb2965

Brem, H., & Tomic-Canic, M. (2007). Cellular and molecular basis of wound healing in

diabetes. J Clin Invest, 117(5), 1219-1222. doi: 10.1172/JCI32169

Briquet, A., Dubois, S., Bekaert, S., Dolhet, M., Beguin, Y., & Gothot, A. (2010).

Prolonged ex vivo culture of human bone marrow mesenchymal stem cells

influences their supportive activity toward NOD/SCID-repopulating cells and

committed progenitor cells of B lymphoid and myeloid lineages. Haematologica,

95(1), 47-56. doi: 10.3324/haematol.2009.008524

Page 134: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

124

Bryder, D., Rossi, D. J., & Weissman, I. L. (2006). Hematopoietic stem cells: the

paradigmatic tissue-specific stem cell. Am J Pathol, 169(2), 338-346. doi:

10.2353/ajpath.2006.060312

Campagnoli, C., Roberts, I. A., Kumar, S., Bennett, P. R., Bellantuono, I., & Fisk, N. M.

(2001). Identification of mesenchymal stem/progenitor cells in human first-

trimester fetal blood, liver, and bone marrow. Blood, 98(8), 2396-2402.

Caplan, A. I., & Dennis, J. E. (2006). Mesenchymal stem cells as trophic mediators. J

Cell Biochem, 98(5), 1076-1084. doi: 10.1002/jcb.20886

Chamberlain, G., Fox, J., Ashton, B., & Middleton, J. (2007). Concise review:

mesenchymal stem cells: their phenotype, differentiation capacity, immunological

features, and potential for homing. Stem Cells, 25(11), 2739-2749. doi:

10.1634/stemcells.2007-0197

Chen, J. S., Wong, V. W., & Gurtner, G. C. (2012). Therapeutic potential of bone

marrow-derived mesenchymal stem cells for cutaneous wound healing. Front

Immunol, 3, 192. doi: 10.3389/fimmu.2012.00192

Cheon, S. S., Wei, Q., Gurung, A., Youn, A., Bright, T., Poon, R., . . . Alman, B. A.

(2006). Beta-catenin regulates wound size and mediates the effect of TGF-beta in

cutaneous healing. FASEB J, 20(6), 692-701. doi: 10.1096/fj.05-4759com

Choi, H., Lee, R. H., Bazhanov, N., Oh, J. Y., & Prockop, D. J. (2011). Anti-

inflammatory protein TSG-6 secreted by activated MSCs attenuates zymosan-

induced mouse peritonitis by decreasing TLR2/NF-kappaB signaling in resident

macrophages. Blood, 118(2), 330-338. doi: 10.1182/blood-2010-12-327353

Choudhery, M. S., Khan, M., Mahmood, R., Mehmood, A., Khan, S. N., & Riazuddin, S.

(2012). Bone marrow derived mesenchymal stem cells from aged mice have

reduced wound healing, angiogenesis, proliferation and anti-apoptosis capabilities.

Cell Biol Int, 36(8), 747-753. doi: 10.1042/CBI20110183

Chua, S. J., Bielecki, R., Yamanaka, N., Fehlings, M. G., Rogers, I. M., & Casper, R. F.

(2010). The effect of umbilical cord blood cells on outcomes after experimental

traumatic spinal cord injury. Spine (Phila Pa 1976), 35(16), 1520-1526. doi:

10.1097/BRS.0b013e3181c3e963

Conget, P. A., & Minguell, J. J. (1999). Phenotypical and functional properties of human

bone marrow mesenchymal progenitor cells. J Cell Physiol, 181(1), 67-73. doi:

10.1002/(SICI)1097-4652(199910)181:1<67::AID-JCP7>3.0.CO;2-C

Czechanski, A., Byers, C., Greenstein, I., Schrode, N., Donahue, L. R., Hadjantonakis, A.

K., & Reinholdt, L. G. (2014). Derivation and characterization of mouse

embryonic stem cells from permissive and nonpermissive strains. Nat Protoc, 9(3),

559-574. doi: 10.1038/nprot.2014.030

da Silva Meirelles, L., Caplan, A. I., & Nardi, N. B. (2008). In search of the in vivo

identity of mesenchymal stem cells. Stem Cells, 26(9), 2287-2299. doi:

10.1634/stemcells.2007-1122

Dash, N. R., Dash, S. N., Routray, P., Mohapatra, S., & Mohapatra, P. C. (2009).

Targeting nonhealing ulcers of lower extremity in human through autologous

bone marrow-derived mesenchymal stem cells. Rejuvenation Res, 12(5), 359-366.

doi: 10.1089/rej.2009.0872

Davidson, J. M. (2001). Experimental Animal Wound Models. Wounds. 13(1). Retrieved

August 17, 2015

Page 135: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

125

De Ugarte, D. A., Alfonso, Z., Zuk, P. A., Elbarbary, A., Zhu, M., Ashjian, P., . . . Fraser,

J. K. (2003). Differential expression of stem cell mobilization-associated

molecules on multi-lineage cells from adipose tissue and bone marrow. Immunol

Lett, 89(2-3), 267-270.

Deans, R. J., & Moseley, A. B. (2000). Mesenchymal stem cells: biology and potential

clinical uses. Exp Hematol, 28(8), 875-884.

Di Nicola, M., Carlo-Stella, C., Magni, M., Milanesi, M., Longoni, P. D., Matteucci,

P., . . . Gianni, A. M. (2002). Human bone marrow stromal cells suppress T-

lymphocyte proliferation induced by cellular or nonspecific mitogenic stimuli.

Blood, 99(10), 3838-3843.

Diegelmann, R. F., Lindblad, W. J., & Cohen, I. K. (1986). A subcutaneous implant for

wound healing studies in humans. J Surg Res, 40(3), 229-237.

Dykstra, B., Kent, D., Bowie, M., McCaffrey, L., Hamilton, M., Lyons, K., . . . Eaves, C.

(2007). Long-term propagation of distinct hematopoietic differentiation programs

in vivo. Cell Stem Cell, 1(2), 218-229. doi: 10.1016/j.stem.2007.05.015

English, K., French, A., & Wood, K. J. (2010). Mesenchymal stromal cells: facilitators of

successful transplantation? Cell Stem Cell, 7(4), 431-442. doi:

10.1016/j.stem.2010.09.009

Evans, M. J., & Kaufman, M. H. (1981). Establishment in culture of pluripotential cells

from mouse embryos. Nature, 292(5819), 154-156.

Falanga, V. (2005). Wound healing and its impairment in the diabetic foot. Lancet,

366(9498), 1736-1743. doi: 10.1016/S0140-6736(05)67700-8

Falanga, V., Iwamoto, S., Chartier, M., Yufit, T., Butmarc, J., Kouttab, N., . . . Carson, P.

(2007). Autologous bone marrow-derived cultured mesenchymal stem cells

delivered in a fibrin spray accelerate healing in murine and human cutaneous

wounds. Tissue Eng, 13(6), 1299-1312. doi: 10.1089/ten.2006.0278

Fraser, J. K., Wulur, I., Alfonso, Z., & Hedrick, M. H. (2006). Fat tissue: an

underappreciated source of stem cells for biotechnology. Trends Biotechnol, 24(4),

150-154. doi: 10.1016/j.tibtech.2006.01.010

Friedenstein, A. J., Chailakhjan, R. K., & Lalykina, K. S. (1970). The development of

fibroblast colonies in monolayer cultures of guinea-pig bone marrow and spleen

cells. Cell Tissue Kinet, 3(4), 393-403.

Gaebel, R., Furlani, D., Sorg, H., Polchow, B., Frank, J., Bieback, K., . . . Steinhoff, G.

(2011). Cell origin of human mesenchymal stem cells determines a different

healing performance in cardiac regeneration. PLoS One, 6(2), e15652. doi:

10.1371/journal.pone.0015652

Galiano, R. D., Michaels, J. t., Dobryansky, M., Levine, J. P., & Gurtner, G. C. (2004).

Quantitative and reproducible murine model of excisional wound healing. Wound

Repair Regen, 12(4), 485-492. doi: 10.1111/j.1067-1927.2004.12404.x

Galkowska, H., Wojewodzka, U., & Olszewski, W. L. (2006). Chemokines, cytokines,

and growth factors in keratinocytes and dermal endothelial cells in the margin of

chronic diabetic foot ulcers. Wound Repair Regen, 14(5), 558-565. doi:

10.1111/j.1743-6109.2006.00155.x

Ghannam, S., Bouffi, C., Djouad, F., Jorgensen, C., & Noel, D. (2010).

Immunosuppression by mesenchymal stem cells: mechanisms and clinical

applications. Stem Cell Res Ther, 1(1), 2. doi: 10.1186/scrt2

Page 136: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

126

Gilbert, S. F. (2000). Developmental Biology (6th ed.): Sunderland (MA): Sinauer

Associates.

Giordano, A., Galderisi, U., & Marino, I. R. (2007). From the laboratory bench to the

patient's bedside: an update on clinical trials with mesenchymal stem cells. J Cell

Physiol, 211(1), 27-35. doi: 10.1002/jcp.20959

Gittel, C., Brehm, W., Burk, J., Juelke, H., Staszyk, C., & Ribitsch, I. (2013). Isolation of

equine multipotent mesenchymal stromal cells by enzymatic tissue digestion or

explant technique: comparison of cellular properties. BMC Vet Res, 9, 221. doi:

10.1186/1746-6148-9-221

Goodwin, H. S., Bicknese, A. R., Chien, S. N., Bogucki, B. D., Quinn, C. O., & Wall, D.

A. (2001). Multilineage differentiation activity by cells isolated from umbilical

cord blood: expression of bone, fat, and neural markers. Biol Blood Marrow

Transplant, 7(11), 581-588.

Guo, S., & Dipietro, L. A. (2010). Factors affecting wound healing. J Dent Res, 89(3),

219-229. doi: 10.1177/0022034509359125

Gurtner, G. C., Werner, S., Barrandon, Y., & Longaker, M. T. (2008). Wound repair and

regeneration. Nature, 453(7193), 314-321. doi: 10.1038/nature07039

Han, Y. F., Tao, R., Sun, T. J., Chai, J. K., Xu, G., & Liu, J. (2013). Optimization of

human umbilical cord mesenchymal stem cell isolation and culture methods.

Cytotechnology, 65(5), 819-827. doi: 10.1007/s10616-012-9528-0

Harris, R. B., Mitchell, T. D., Yan, X., Simpson, J. S., & Redmann, S. M., Jr. (2001).

Metabolic responses to leptin in obese db/db mice are strain dependent. Am J

Physiol Regul Integr Comp Physiol, 281(1), R115-132.

Hass, R., Kasper, C., Bohm, S., & Jacobs, R. (2011). Different populations and sources of

human mesenchymal stem cells (MSC): A comparison of adult and neonatal

tissue-derived MSC. Cell Commun Signal, 9, 12. doi: 10.1186/1478-811X-9-12

Horwitz, E. M., & Dominici, M. (2008). How do mesenchymal stromal cells exert their

therapeutic benefit? Cytotherapy, 10(8), 771-774. doi:

10.1080/14653240802618085

Horwitz, E. M., Prockop, D. J., Fitzpatrick, L. A., Koo, W. W., Gordon, P. L., Neel,

M., . . . Brenner, M. K. (1999). Transplantability and therapeutic effects of bone

marrow-derived mesenchymal cells in children with osteogenesis imperfecta. Nat

Med, 5(3), 309-313. doi: 10.1038/6529

Hsieh, J. Y., Wang, H. W., Chang, S. J., Liao, K. H., Lee, I. H., Lin, W. S., . . . Cheng, S.

M. (2013). Mesenchymal stem cells from human umbilical cord express

preferentially secreted factors related to neuroprotection, neurogenesis, and

angiogenesis. PLoS One, 8(8), e72604. doi: 10.1371/journal.pone.0072604

Hu, Y., Liao, L., Wang, Q., Ma, L., Ma, G., Jiang, X., & Zhao, R. C. (2003). Isolation

and identification of mesenchymal stem cells from human fetal pancreas. J Lab

Clin Med, 141(5), 342-349. doi: 10.1016/S0022-2143(03)00022-2

Huang, S., Feng, C., Wu, Y., Yang, S., Ma, K., Wu, X., & Fu, X. (2013). Dissimilar

characteristics of umbilical cord mesenchymal stem cells from donors of different

ages. Cell Tissue Bank, 14(4), 707-713. doi: 10.1007/s10561-013-9364-2

Hummel, K. P., Coleman, D. L., & Lane, P. W. (1972). The influence of genetic

background on expression of mutations at the diabetes locus in the mouse. I.

C57BL-KsJ and C57BL-6J strains. Biochem Genet, 7(1), 1-13.

Page 137: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

127

Hwang, J. H., Shim, S. S., Seok, O. S., Lee, H. Y., Woo, S. K., Kim, B. H., . . . Park, Y.

K. (2009). Comparison of cytokine expression in mesenchymal stem cells from

human placenta, cord blood, and bone marrow. J Korean Med Sci, 24(4), 547-554.

doi: 10.3346/jkms.2009.24.4.547

in 't Anker, P. S., Noort, W. A., Scherjon, S. A., Kleijburg-van der Keur, C.,

Kruisselbrink, A. B., van Bezooijen, R. L., . . . Fibbe, W. E. (2003). Mesenchymal

stem cells in human second-trimester bone marrow, liver, lung, and spleen exhibit

a similar immunophenotype but a heterogeneous multilineage differentiation

potential. Haematologica, 88(8), 845-852.

Isakson, M., de Blacam, C., Whelan, D., McArdle, A., & Clover, A. J. (2015).

Mesenchymal Stem Cells and Cutaneous Wound Healing: Current Evidence and

Future Potential. Stem Cells Int, 2015, 831095. doi: 10.1155/2015/831095

Isern, J., Garcia-Garcia, A., Martin, A. M., Arranz, L., Martin-Perez, D., Torroja, C., . . .

Mendez-Ferrer, S. (2014). The neural crest is a source of mesenchymal stem cells

with specialized hematopoietic stem cell niche function. Elife, 3, e03696. doi:

10.7554/eLife.03696

Iso, Y., Spees, J. L., Serrano, C., Bakondi, B., Pochampally, R., Song, Y. H., . . . Prockop,

D. J. (2007). Multipotent human stromal cells improve cardiac function after

myocardial infarction in mice without long-term engraftment. Biochem Biophys

Res Commun, 354(3), 700-706. doi: 10.1016/j.bbrc.2007.01.045

Javazon, E. H., Keswani, S. G., Badillo, A. T., Crombleholme, T. M., Zoltick, P. W.,

Radu, A. P., . . . Flake, A. W. (2007). Enhanced epithelial gap closure and

increased angiogenesis in wounds of diabetic mice treated with adult murine bone

marrow stromal progenitor cells. Wound Repair Regen, 15(3), 350-359. doi:

10.1111/j.1524-475X.2007.00237.x

Jeffcoate, W. J., & Harding, K. G. (2003). Diabetic foot ulcers. Lancet, 361(9368), 1545-

1551. doi: 10.1016/S0140-6736(03)13169-8

Jiang, Y., Jahagirdar, B. N., Reinhardt, R. L., Schwartz, R. E., Keene, C. D., Ortiz-

Gonzalez, X. R., . . . Verfaillie, C. M. (2002). Pluripotency of mesenchymal stem

cells derived from adult marrow. Nature, 418(6893), 41-49. doi:

10.1038/nature00870

Joseph M McCulloch, L. C. K. (2010). Wound Healing: Evidence-Based Management:

F.A. Davis.

Joyce, N., Annett, G., Wirthlin, L., Olson, S., Bauer, G., & Nolta, J. A. (2010).

Mesenchymal stem cells for the treatment of neurodegenerative disease. Regen

Med, 5(6), 933-946. doi: 10.2217/rme.10.72

Kahn, S. E., Cooper, M. E., & Del Prato, S. (2014). Pathophysiology and treatment of

type 2 diabetes: perspectives on the past, present, and future. Lancet, 383(9922),

1068-1083. doi: 10.1016/S0140-6736(13)62154-6

Karp, J. M., & Leng Teo, G. S. (2009). Mesenchymal stem cell homing: the devil is in the

details. Cell Stem Cell, 4(3), 206-216. doi: 10.1016/j.stem.2009.02.001

Kawase, E., Suemori, H., Takahashi, N., Okazaki, K., Hashimoto, K., & Nakatsuji, N.

(1994). Strain difference in establishment of mouse embryonic stem (ES) cell

lines. Int J Dev Biol, 38(2), 385-390.

Keating, A. (2012). Mesenchymal stromal cells: new directions. Cell Stem Cell, 10(6),

709-716. doi: 10.1016/j.stem.2012.05.015

Page 138: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

128

Kern, S., Eichler, H., Stoeve, J., Kluter, H., & Bieback, K. (2006). Comparative analysis

of mesenchymal stem cells from bone marrow, umbilical cord blood, or adipose

tissue. Stem Cells, 24(5), 1294-1301. doi: 10.1634/stemcells.2005-0342

King, A. J. (2012). The use of animal models in diabetes research. Br J Pharmacol,

166(3), 877-894. doi: 10.1111/j.1476-5381.2012.01911.x

Klampfer, L., Chen-Kiang, S., & Vilcek, J. (1995). Activation of the TSG-6 gene by NF-

IL6 requires two adjacent NF-IL6 binding sites. J Biol Chem, 270(8), 3677-3682.

Klampfer, L., Lee, T. H., Hsu, W., Vilcek, J., & Chen-Kiang, S. (1994). NF-IL6 and AP-

1 cooperatively modulate the activation of the TSG-6 gene by tumor necrosis

factor alpha and interleukin-1. Mol Cell Biol, 14(10), 6561-6569.

Kraitchman, D. L., Heldman, A. W., Atalar, E., Amado, L. C., Martin, B. J., Pittenger, M.

F., . . . Bulte, J. W. (2003). In vivo magnetic resonance imaging of mesenchymal

stem cells in myocardial infarction. Circulation, 107(18), 2290-2293. doi:

10.1161/01.CIR.0000070931.62772.4E

Le Blanc, K., & Davies, L. C. (2015). Mesenchymal stromal cells and the innate immune

response. Immunol Lett. doi: 10.1016/j.imlet.2015.05.004

Le Blanc, K., & Ringden, O. (2007). Immunomodulation by mesenchymal stem cells and

clinical experience. J Intern Med, 262(5), 509-525. doi: 10.1111/j.1365-

2796.2007.01844.x

Lee, O. K., Kuo, T. K., Chen, W. M., Lee, K. D., Hsieh, S. L., & Chen, T. H. (2004).

Isolation of multipotent mesenchymal stem cells from umbilical cord blood.

Blood, 103(5), 1669-1675. doi: 10.1182/blood-2003-05-1670

Lee, R. H., Pulin, A. A., Seo, M. J., Kota, D. J., Ylostalo, J., Larson, B. L., . . . Prockop,

D. J. (2009). Intravenous hMSCs improve myocardial infarction in mice because

cells embolized in lung are activated to secrete the anti-inflammatory protein

TSG-6. Cell Stem Cell, 5(1), 54-63. doi: 10.1016/j.stem.2009.05.003

Lee, R. H., Yu, J. M., Foskett, A. M., Peltier, G., Reneau, J. C., Bazhanov, N., . . .

Prockop, D. J. (2014). TSG-6 as a biomarker to predict efficacy of human

mesenchymal stem/progenitor cells (hMSCs) in modulating sterile inflammation

in vivo. Proc Natl Acad Sci U S A, 111(47), 16766-16771. doi:

10.1073/pnas.1416121111

Lee, T. H., Wisniewski, H. G., & Vilcek, J. (1992). A novel secretory tumor necrosis

factor-inducible protein (TSG-6) is a member of the family of hyaluronate binding

proteins, closely related to the adhesion receptor CD44. J Cell Biol, 116(2), 545-

557.

Lesley, J., Gal, I., Mahoney, D. J., Cordell, M. R., Rugg, M. S., Hyman, R., . . . Mikecz,

K. (2004). TSG-6 modulates the interaction between hyaluronan and cell surface

CD44. J Biol Chem, 279(24), 25745-25754. doi: 10.1074/jbc.M313319200

Liu, Y., Yin, Z., Zhang, R., Yan, K., Chen, L., Chen, F., . . . Jiang, X. (2014). MSCs

inhibit bone marrow-derived DC maturation and function through the release of

TSG-6. Biochem Biophys Res Commun, 450(4), 1409-1415. doi:

10.1016/j.bbrc.2014.07.001

Loot, M. A., Kenter, S. B., Au, F. L., van Galen, W. J., Middelkoop, E., Bos, J. D., &

Mekkes, J. R. (2002). Fibroblasts derived from chronic diabetic ulcers differ in

their response to stimulation with EGF, IGF-I, bFGF and PDGF-AB compared to

controls. Eur J Cell Biol, 81(3), 153-160.

Page 139: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

129

Lv, F. J., Tuan, R. S., Cheung, K. M., & Leung, V. Y. (2014). Concise review: the

surface markers and identity of human mesenchymal stem cells. Stem Cells, 32(6),

1408-1419. doi: 10.1002/stem.1681

Ma, S., Xie, N., Li, W., Yuan, B., Shi, Y., & Wang, Y. (2014). Immunobiology of

mesenchymal stem cells. Cell Death Differ, 21(2), 216-225. doi:

10.1038/cdd.2013.158

Maehle, A. H. (2011). Ambiguous cells: the emergence of the stem cell concept in the

nineteenth and twentieth centuries. Notes Rec R Soc Lond, 65(4), 359-378.

Mark, P., Kleinsorge, M., Gaebel, R., Lux, C. A., Toelk, A., Pittermann, E., . . . Ma, N.

(2013). Human Mesenchymal Stem Cells Display Reduced Expression of CD105

after Culture in Serum-Free Medium. Stem Cells Int, 2013, 698076. doi:

10.1155/2013/698076

Martin, G. R. (1981). Isolation of a pluripotent cell line from early mouse embryos

cultured in medium conditioned by teratocarcinoma stem cells. Proc Natl Acad

Sci U S A, 78(12), 7634-7638.

Martin, G. R., & Evans, M. J. (1975). Differentiation of clonal lines of teratocarcinoma

cells: formation of embryoid bodies in vitro. Proc Natl Acad Sci U S A, 72(4),

1441-1445.

Martin, P. (1997). Wound healing--aiming for perfect skin regeneration. Science,

276(5309), 75-81.

Maxson, S., Lopez, E. A., Yoo, D., Danilkovitch-Miagkova, A., & Leroux, M. A. (2012).

Concise review: role of mesenchymal stem cells in wound repair. Stem Cells

Transl Med, 1(2), 142-149. doi: 10.5966/sctm.2011-0018

Mayor, R., & Theveneau, E. (2013). The neural crest. Development, 140(11), 2247-2251.

doi: 10.1242/dev.091751

Meirelles Lda, S., Fontes, A. M., Covas, D. T., & Caplan, A. I. (2009). Mechanisms

involved in the therapeutic properties of mesenchymal stem cells. Cytokine

Growth Factor Rev, 20(5-6), 419-427. doi: 10.1016/j.cytogfr.2009.10.002

Meisel, R., Zibert, A., Laryea, M., Gobel, U., Daubener, W., & Dilloo, D. (2004). Human

bone marrow stromal cells inhibit allogeneic T-cell responses by indoleamine 2,3-

dioxygenase-mediated tryptophan degradation. Blood, 103(12), 4619-4621. doi:

10.1182/blood-2003-11-3909

Mellor, A. L., & Munn, D. H. (1999). Tryptophan catabolism and T-cell tolerance:

immunosuppression by starvation? Immunol Today, 20(10), 469-473.

Michaels, J. t., Churgin, S. S., Blechman, K. M., Greives, M. R., Aarabi, S., Galiano, R.

D., & Gurtner, G. C. (2007). db/db mice exhibit severe wound-healing

impairments compared with other murine diabetic strains in a silicone-splinted

excisional wound model. Wound Repair Regen, 15(5), 665-670. doi:

10.1111/j.1524-475X.2007.00273.x

Milner, C. M., & Day, A. J. (2003). TSG-6: a multifunctional protein associated with

inflammation. J Cell Sci, 116(Pt 10), 1863-1873. doi: 10.1242/jcs.00407

Mitalipov, S., & Wolf, D. (2009). Totipotency, pluripotency and nuclear reprogramming.

Adv Biochem Eng Biotechnol, 114, 185-199. doi: 10.1007/10_2008_45

Moretta, A. (2002). Natural killer cells and dendritic cells: rendezvous in abused tissues.

Nat Rev Immunol, 2(12), 957-964. doi: 10.1038/nri956

Page 140: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

130

Morikawa, S., Mabuchi, Y., Niibe, K., Suzuki, S., Nagoshi, N., Sunabori, T., . . .

Matsuzaki, Y. (2009). Development of mesenchymal stem cells partially originate

from the neural crest. Biochem Biophys Res Commun, 379(4), 1114-1119. doi:

10.1016/j.bbrc.2009.01.031

Muller, M., Trocme, C., Lardy, B., Morel, F., Halimi, S., & Benhamou, P. Y. (2008).

Matrix metalloproteinases and diabetic foot ulcers: the ratio of MMP-1 to TIMP-1

is a predictor of wound healing. Diabet Med, 25(4), 419-426. doi: 10.1111/j.1464-

5491.2008.02414.x

Nagy, A., Rossant, J., Nagy, R., Abramow-Newerly, W., & Roder, J. C. (1993).

Derivation of completely cell culture-derived mice from early-passage embryonic

stem cells. Proc Natl Acad Sci U S A, 90(18), 8424-8428.

Nichols, J., Zevnik, B., Anastassiadis, K., Niwa, H., Klewe-Nebenius, D., Chambers,

I., . . . Smith, A. (1998). Formation of pluripotent stem cells in the mammalian

embryo depends on the POU transcription factor Oct4. Cell, 95(3), 379-391.

Nombela-Arrieta, C., Ritz, J., & Silberstein, L. E. (2011). The elusive nature and function

of mesenchymal stem cells. Nat Rev Mol Cell Biol, 12(2), 126-131. doi:

10.1038/nrm3049

Oedayrajsingh-Varma, M. J., van Ham, S. M., Knippenberg, M., Helder, M. N., Klein-

Nulend, J., Schouten, T. E., . . . van Milligen, F. J. (2006). Adipose tissue-derived

mesenchymal stem cell yield and growth characteristics are affected by the tissue-

harvesting procedure. Cytotherapy, 8(2), 166-177. doi:

10.1080/14653240600621125

Ortiz, L. A., Gambelli, F., McBride, C., Gaupp, D., Baddoo, M., Kaminski, N., &

Phinney, D. G. (2003). Mesenchymal stem cell engraftment in lung is enhanced in

response to bleomycin exposure and ameliorates its fibrotic effects. Proc Natl

Acad Sci U S A, 100(14), 8407-8411. doi: 10.1073/pnas.1432929100

Otte, A., Bucan, V., Reimers, K., & Hass, R. (2013). Mesenchymal stem cells maintain

long-term in vitro stemness during explant culture. Tissue Eng Part C Methods,

19(12), 937-948. doi: 10.1089/ten.TEC.2013.0007

Pallin, B., Ahonen, J., Rank, F., & Zederfeldt, B. (1975). Granulation tissue formation in

viscose cellulose sponges of different design. Acta Chir Scand, 141(8), 697-701.

Pandey, A., Chawla, S., & Guchhait, P. (2015). Type-2 diabetes: Current understanding

and future perspectives. IUBMB Life, 67(7), 506-513. doi: 10.1002/iub.1396

Park, C. W., Kim, K. S., Bae, S., Son, H. K., Myung, P. K., Hong, H. J., & Kim, H.

(2009). Cytokine secretion profiling of human mesenchymal stem cells by

antibody array. Int J Stem Cells, 2(1), 59-68.

Paulini, K., Korner, B., Beneke, G., & Endres, R. (1974). A quantitative study of the

growth of connective tissue: investigations on implanted polyester-polyurethane

sponges. Connect Tissue Res, 2(4), 257-264.

Perez, R., & Davis, S. C. (2008). Relevance of animal models for wound healing.

Wounds, 20(1), 3-8.

Phinney, D. G., & Prockop, D. J. (2007). Concise review: mesenchymal stem/multipotent

stromal cells: the state of transdifferentiation and modes of tissue repair--current

views. Stem Cells, 25(11), 2896-2902. doi: 10.1634/stemcells.2007-0637

Page 141: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

131

Pittenger, M. F., Mackay, A. M., Beck, S. C., Jaiswal, R. K., Douglas, R., Mosca, J.

D., . . . Marshak, D. R. (1999). Multilineage potential of adult human

mesenchymal stem cells. Science, 284(5411), 143-147.

Poschl, E., Schlotzer-Schrehardt, U., Brachvogel, B., Saito, K., Ninomiya, Y., & Mayer,

U. (2004). Collagen IV is essential for basement membrane stability but

dispensable for initiation of its assembly during early development. Development,

131(7), 1619-1628. doi: 10.1242/dev.01037

Prockop, D. J. (1997). Marrow stromal cells as stem cells for nonhematopoietic tissues.

Science, 276(5309), 71-74.

Prockop, D. J. (2009). Repair of tissues by adult stem/progenitor cells (MSCs):

controversies, myths, and changing paradigms. Mol Ther, 17(6), 939-946. doi:

10.1038/mt.2009.62

Prosdocimi, M., & Bevilacqua, C. (2012). Impaired wound healing in diabetes: the

rationale for clinical use of hyaluronic acid plus silver sulfadiazine. Minerva Med,

103(6), 533-539.

Puri, M. C., & Nagy, A. (2012). Concise review: Embryonic stem cells versus induced

pluripotent stem cells: the game is on. Stem Cells, 30(1), 10-14. doi:

10.1002/stem.788

Qi, Y., Jiang, D., Sindrilaru, A., Stegemann, A., Schatz, S., Treiber, N., . . . Scharffetter-

Kochanek, K. (2014). TSG-6 released from intradermally injected mesenchymal

stem cells accelerates wound healing and reduces tissue fibrosis in murine full-

thickness skin wounds. J Invest Dermatol, 134(2), 526-537. doi:

10.1038/jid.2013.328

Ramalho-Santos, M., & Willenbring, H. (2007). On the origin of the term "stem cell".

Cell Stem Cell, 1(1), 35-38. doi: 10.1016/j.stem.2007.05.013

Rebelatto, C. K., Aguiar, A. M., Moretao, M. P., Senegaglia, A. C., Hansen, P., Barchiki,

F., . . . Correa, A. (2008). Dissimilar differentiation of mesenchymal stem cells

from bone marrow, umbilical cord blood, and adipose tissue. Exp Biol Med

(Maywood), 233(7), 901-913. doi: 10.3181/0712-RM-356

Reubinoff, B. E., Pera, M. F., Fong, C. Y., Trounson, A., & Bongso, A. (2000).

Embryonic stem cell lines from human blastocysts: somatic differentiation in vitro.

Nat Biotechnol, 18(4), 399-404. doi: 10.1038/74447

Rodriguez, A. M., Elabd, C., Amri, E. Z., Ailhaud, G., & Dani, C. (2005). The human

adipose tissue is a source of multipotent stem cells. Biochimie, 87(1), 125-128.

doi: 10.1016/j.biochi.2004.11.007

Rogers, I., Yamanaka, N., Bielecki, R., Wong, C. J., Chua, S., Yuen, S., & Casper, R. F.

(2007). Identification and analysis of in vitro cultured CD45-positive cells capable

of multi-lineage differentiation. Exp Cell Res, 313(9), 1839-1852. doi:

10.1016/j.yexcr.2007.02.029

Rojas, M., Xu, J., Woods, C. R., Mora, A. L., Spears, W., Roman, J., & Brigham, K. L.

(2005). Bone marrow-derived mesenchymal stem cells in repair of the injured

lung. Am J Respir Cell Mol Biol, 33(2), 145-152. doi: 10.1165/rcmb.2004-

0330OC

Rosova, I., Dao, M., Capoccia, B., Link, D., & Nolta, J. A. (2008). Hypoxic

preconditioning results in increased motility and improved therapeutic potential of

Page 142: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

132

human mesenchymal stem cells. Stem Cells, 26(8), 2173-2182. doi:

10.1634/stemcells.2007-1104

Sarugaser, R., Hanoun, L., Keating, A., Stanford, W. L., & Davies, J. E. (2009). Human

mesenchymal stem cells self-renew and differentiate according to a deterministic

hierarchy. PLoS One, 4(8), e6498. doi: 10.1371/journal.pone.0006498

Sato, K., Ozaki, K., Oh, I., Meguro, A., Hatanaka, K., Nagai, T., . . . Ozawa, K. (2007).

Nitric oxide plays a critical role in suppression of T-cell proliferation by

mesenchymal stem cells. Blood, 109(1), 228-234. doi: 10.1182/blood-2006-02-

002246

Schreml, S., Babilas, P., Fruth, S., Orso, E., Schmitz, G., Mueller, M. B., . . . Prantl, L.

(2009). Harvesting human adipose tissue-derived adult stem cells: resection

versus liposuction. Cytotherapy, 11(7), 947-957. doi:

10.3109/14653240903204322

Secco, M., Zucconi, E., Vieira, N. M., Fogaca, L. L., Cerqueira, A., Carvalho, M. D., . . .

Zatz, M. (2008). Multipotent stem cells from umbilical cord: cord is richer than

blood! Stem Cells, 26(1), 146-150. doi: 10.1634/stemcells.2007-0381

Seita, J., & Weissman, I. L. (2010). Hematopoietic stem cell: self-renewal versus

differentiation. Wiley Interdiscip Rev Syst Biol Med, 2(6), 640-653. doi:

10.1002/wsbm.86

Shake, J. G., Gruber, P. J., Baumgartner, W. A., Senechal, G., Meyers, J., Redmond, J.

M., . . . Martin, B. J. (2002). Mesenchymal stem cell implantation in a swine

myocardial infarct model: engraftment and functional effects. Ann Thorac Surg,

73(6), 1919-1925; discussion 1926.

Shi, Z., Zhao, L., Qiu, G., He, R., & Detamore, M. S. (2015). The effect of extended

passaging on the phenotype and osteogenic potential of human umbilical cord

mesenchymal stem cells. Mol Cell Biochem, 401(1-2), 155-164. doi:

10.1007/s11010-014-2303-0

Shin, L., & Peterson, D. A. (2013). Human mesenchymal stem cell grafts enhance normal

and impaired wound healing by recruiting existing endogenous tissue

stem/progenitor cells. Stem Cells Transl Med, 2(1), 33-42. doi:

10.5966/sctm.2012-0041

Sonnemann, K. J., & Bement, W. M. (2011). Wound repair: toward understanding and

integration of single-cell and multicellular wound responses. Annu Rev Cell Dev

Biol, 27, 237-263. doi: 10.1146/annurev-cellbio-092910-154251

Spaggiari, G. M., Capobianco, A., Becchetti, S., Mingari, M. C., & Moretta, L. (2006).

Mesenchymal stem cell-natural killer cell interactions: evidence that activated NK

cells are capable of killing MSCs, whereas MSCs can inhibit IL-2-induced NK-

cell proliferation. Blood, 107(4), 1484-1490. doi: 10.1182/blood-2005-07-2775

Spangrude, G. J., Heimfeld, S., & Weissman, I. L. (1988). Purification and

characterization of mouse hematopoietic stem cells. Science, 241(4861), 58-62.

Stojadinovic, O., Brem, H., Vouthounis, C., Lee, B., Fallon, J., Stallcup, M., . . . Tomic-

Canic, M. (2005). Molecular pathogenesis of chronic wounds: the role of beta-

catenin and c-myc in the inhibition of epithelialization and wound healing. Am J

Pathol, 167(1), 59-69.

Page 143: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

133

Stolzing, A., Jones, E., McGonagle, D., & Scutt, A. (2008). Age-related changes in

human bone marrow-derived mesenchymal stem cells: consequences for cell

therapies. Mech Ageing Dev, 129(3), 163-173. doi: 10.1016/j.mad.2007.12.002

Stroncek JD, R. W. (2008). Overview of Wound Healing in Different Tissue Types. In R.

WM (Ed.), Indwelling Neural Implants: Strategies for Contending with the In

Vivo Environment.: Boca Raton (FL): CRC Press.

Takahashi, K., & Yamanaka, S. (2006). Induction of pluripotent stem cells from mouse

embryonic and adult fibroblast cultures by defined factors. Cell, 126(4), 663-676.

doi: 10.1016/j.cell.2006.07.024

Terness, P., Bauer, T. M., Rose, L., Dufter, C., Watzlik, A., Simon, H., & Opelz, G.

(2002). Inhibition of allogeneic T cell proliferation by indoleamine 2,3-

dioxygenase-expressing dendritic cells: mediation of suppression by tryptophan

metabolites. J Exp Med, 196(4), 447-457.

Tesar, P. J., Chenoweth, J. G., Brook, F. A., Davies, T. J., Evans, E. P., Mack, D. L., . . .

McKay, R. D. (2007). New cell lines from mouse epiblast share defining features

with human embryonic stem cells. Nature, 448(7150), 196-199. doi:

10.1038/nature05972

The Jackson Laboratory, T. J. (2014). BKS.Cg-Dock7m +/+ Leprdb/J. JAX Mice

database. Retrieved August 17, 2015, from

http://jaxmice.jax.org/strain/000642.html

Thomson, J. A., Itskovitz-Eldor, J., Shapiro, S. S., Waknitz, M. A., Swiergiel, J. J.,

Marshall, V. S., & Jones, J. M. (1998). Embryonic stem cell lines derived from

human blastocysts. Science, 282(5391), 1145-1147.

Tolar, J., Le Blanc, K., Keating, A., & Blazar, B. R. (2010). Concise review: hitting the

right spot with mesenchymal stromal cells. Stem Cells, 28(8), 1446-1455. doi:

10.1002/stem.459

Trivanovic, D., Kocic, J., Mojsilovic, S., Krstic, A., Ilic, V., Djordjevic, I. O., . . .

Bugarski, D. (2013). Mesenchymal stem cells isolated from peripheral blood and

umbilical cord Wharton's jelly. Srp Arh Celok Lek, 141(3-4), 178-186.

Uccelli, A., Moretta, L., & Pistoia, V. (2008). Mesenchymal stem cells in health and

disease. Nat Rev Immunol, 8(9), 726-736. doi: 10.1038/nri2395

Valluru, M., Staton, C. A., Reed, M. W., & Brown, N. J. (2011). Transforming Growth

Factor-beta and Endoglin Signaling Orchestrate Wound Healing. Front Physiol, 2,

89. doi: 10.3389/fphys.2011.00089

Vivier, E., Tomasello, E., Baratin, M., Walzer, T., & Ugolini, S. (2008). Functions of

natural killer cells. Nat Immunol, 9(5), 503-510. doi: 10.1038/ni1582

Vuorisalo, S., Venermo, M., & Lepantalo, M. (2009). Treatment of diabetic foot ulcers. J

Cardiovasc Surg (Torino), 50(3), 275-291.

Wagner, J., Kean, T., Young, R., Dennis, J. E., & Caplan, A. I. (2009). Optimizing

mesenchymal stem cell-based therapeutics. Curr Opin Biotechnol, 20(5), 531-536.

doi: 10.1016/j.copbio.2009.08.009

Wagner, W., Wein, F., Seckinger, A., Frankhauser, M., Wirkner, U., Krause, U., . . . Ho,

A. D. (2005). Comparative characteristics of mesenchymal stem cells from human

bone marrow, adipose tissue, and umbilical cord blood. Exp Hematol, 33(11),

1402-1416. doi: 10.1016/j.exphem.2005.07.003

Page 144: By Vanessa Noemi Raileanu - University of Toronto T-Space · Vanessa Noemi Raileanu Master of Science Department of Physiology University of Toronto 2015 Abstract Mesenchymal stromal

134

Wang, H. S., Hung, S. C., Peng, S. T., Huang, C. C., Wei, H. M., Guo, Y. J., . . . Chen, C.

C. (2004). Mesenchymal stem cells in the Wharton's jelly of the human umbilical

cord. Stem Cells, 22(7), 1330-1337. doi: 10.1634/stemcells.2004-0013

Wang, Y., Zhang, Z., Chi, Y., Zhang, Q., Xu, F., Yang, Z., . . . Han, Z. C. (2013). Long-

term cultured mesenchymal stem cells frequently develop genomic mutations but

do not undergo malignant transformation. Cell Death Dis, 4, e950. doi:

10.1038/cddis.2013.480

Waterman, R. S., Tomchuck, S. L., Henkle, S. L., & Betancourt, A. M. (2010). A new

mesenchymal stem cell (MSC) paradigm: polarization into a pro-inflammatory

MSC1 or an Immunosuppressive MSC2 phenotype. PLoS One, 5(4), e10088. doi:

10.1371/journal.pone.0010088

Werner, S., & Grose, R. (2003). Regulation of wound healing by growth factors and

cytokines. Physiol Rev, 83(3), 835-870. doi: 10.1152/physrev.00031.2002

Whiteley, J., Bielecki, R., Li, M., Chua, S., Ward, M. R., Yamanaka, N., . . . Rogers, I. M.

(2014). An expanded population of CD34+ cells from frozen banked umbilical

cord blood demonstrate tissue repair mechanisms of mesenchymal stromal cells

and circulating angiogenic cells in an ischemic hind limb model. Stem Cell Rev,

10(3), 338-350. doi: 10.1007/s12015-014-9496-1

Wild, S., Roglic, G., Green, A., Sicree, R., & King, H. (2004). Global prevalence of

diabetes: estimates for the year 2000 and projections for 2030. Diabetes Care,

27(5), 1047-1053.

Wisniewski, H. G., Maier, R., Lotz, M., Lee, S., Klampfer, L., Lee, T. H., & Vilcek, J.

(1993). TSG-6: a TNF-, IL-1-, and LPS-inducible secreted glycoprotein

associated with arthritis. J Immunol, 151(11), 6593-6601.

Wong, V. W., Sorkin, M., Glotzbach, J. P., Longaker, M. T., & Gurtner, G. C. (2011).

Surgical approaches to create murine models of human wound healing. J Biomed

Biotechnol, 2011, 969618. doi: 10.1155/2011/969618

Wu, S. C., Driver, V. R., Wrobel, J. S., & Armstrong, D. G. (2007). Foot ulcers in the

diabetic patient, prevention and treatment. Vasc Health Risk Manag, 3(1), 65-76.

Xu, W., Jong Hong, S., Jia, S., Zhao, Y., Galiano, R. D., & Mustoe, T. A. (2012).

Application of a partial-thickness human ex vivo skin culture model in cutaneous

wound healing study. Lab Invest, 92(4), 584-599. doi: 10.1038/labinvest.2011.184

Yoon, J. H., Roh, E. Y., Shin, S., Jung, N. H., Song, E. Y., Chang, J. Y., . . . Jeon, H. W.

(2013). Comparison of explant-derived and enzymatic digestion-derived MSCs

and the growth factors from Wharton's jelly. Biomed Res Int, 2013, 428726. doi:

10.1155/2013/428726

Young, H. E., Duplaa, C., Young, T. M., Floyd, J. A., Reeves, M. L., Davis, K. H., . . .

Black, A. C., Jr. (2001). Clonogenic analysis reveals reserve stem cells in

postnatal mammals: I. Pluripotent mesenchymal stem cells. Anat Rec, 263(4),

350-360.

Zucconi, E., Vieira, N. M., Bueno, C. R., Jr., Secco, M., Jazedje, T., Costa Valadares,

M., . . . Zatz, M. (2011). Preclinical studies with umbilical cord mesenchymal

stromal cells in different animal models for muscular dystrophy. J Biomed

Biotechnol, 2011, 715251. doi: 10.1155/2011/715251