Close
About
FAQ
Home
Collections
Login
USC Login
Register
0
Selected
Invert selection
Deselect all
Deselect all
Click here to refresh results
Click here to refresh results
USC
/
Digital Library
/
University of Southern California Dissertations and Theses
/
Investigating the role of STAT3 in mouse and rat embryonic stem cell self-renewal and differentiation
(USC Thesis Other)
Investigating the role of STAT3 in mouse and rat embryonic stem cell self-renewal and differentiation
PDF
Download
Share
Open document
Flip pages
Contact Us
Contact Us
Copy asset link
Request this asset
Transcript (if available)
Content
INVESTIGATING THE ROLE OF STAT3 IN MOUSE AND RAT EMBRYONIC STEM CELL
SELF‐RENEWAL AND DIFFERENTIATION
by
Eric Nathaniel Schulze
A Dissertation Presented to the
FACULTY OF THE USC GRADUATE SCHOOL
UNIVERSITY OF SOUTHERN CALIFORNIA
In Partial Fulfillment of the
Requirements for the Degree
DOCTOR OF PHILOSOPHY
(GENETIC, MOLECULAR, AND CELLULAR BIOLOGY)
August 2010
Copyright 2010 Eric Nathaniel Schulze
ii
Epigraph
Somewhere, something incredible is waiting to be known.
‐ Carl Sagan
iii
Dedication
I wish to dedicate this body of work to those Scientists that have both plied their
essential trade and sought to help others understand its innumerable worth to
humanity and this cosmos.
iv
Acknowledgements
I wish to thank my mentor, Dr. Qi‐Long Ying, for his undying passion, creativity, and
patience in transmitting not just what Science is and how it is performed
professionally, but what it can be and the utter limitlessness application Science has
in this world.
I wish to thank my lovely fiancé and partner, Lori, as she has undoubtedly helped
focus my thinking and sharpened my aspirations in this life with her boundless love,
care, and intellect.
I wish to thank my family and friends. Without your ability to help me place Science
into societal context, to realize that seeking observational and reasoned truths
about this world do matter to non‐Scientists in significant ways, I would have
missed a key portion of my training.
To my Committee Members, Professors Martin Pera, Gregor Adams, and Carolyn
Lutzko: Your Scientific acumen and wisdom are forever appreciated. Your belief in
what I have set out to accomplish is cherished. I hope to make each of you proud and
also improve humanity’s knowledge base in the process.
To my labmates, past and present, and my colleagues: Biology is nothing if not a
chance to demonstrate how each individual might un‐weave the tapestry of life.
Your insights both guide and inspire me to be a better Scientist.
v
Table of Contents
Epigraph ii
Dedication iii
Acknowledgements iv
List of Figures vii
Abstract xii
Introduction: 1
I.1 ‐ An Overview of the History of Embryonic Stem
Cell Research 1
I.2 ‐ Mouse ES Cells Use the LIF/STAT3 Pathway
To Self‐Renew 5
I.3 ‐ Permissiveness Relates to Metastable Pluripotent
Cell‐States 13
I.4 ‐ Rationale for This Investigation 19
I.5 – Conclusions 20
Results: 22
Chapter One ‐ STAT3 Overexpression Enhances Feeder‐
Independent Mouse ES Cell Self‐Renewal 22
Chapter Two ‐ Increased Endogenous STAT3 Activation is
Sufficient to Maintain Feeder‐Independent Mouse ES
Cell Self‐Renewal 53
Chapter Three ‐ Increased and Sustained STAT3 Activation
Promotes Mouse and Rat ES Cell Self‐Renewal 62
Chapter Four ‐ Klf4 Overexpression Promotes Mouse and
Rat ES Cell Self‐Renewal 90
Chapter Five ‐ STAT3 Activation May Promote Tumor
Cell Growth Arrest 101
Discussion: 111
D.1 – STAT3 Is Regulated In a Strain or Species‐Specific
Manner to Promote Rodent ES Cell Self‐Renewal 111
D.2 ‐ The Role of STAT3 In Mediating Mouse and
Rat ES Cell Self‐Renewal 113
D.3 ‐ The Role of ERK1/2 In Mouse ES Cell Self‐Renewal 117
vi
D.4 ‐ Rodent ES Cell Self‐Renewal: A Stratified Model 118
D.5 ‐ Future Directions and Implications 123
D.6 – Conclusions 124
Bibliography 126
Appendices: 140
Appendix A: Materials and Methods 140
Appendix B: Table of Primer Sets 146
vii
List of Figures
Figure I.1a: Embryonic stem cells permit precise gene‐targeting. 3
Figure I.2a: STAT3 regulates mouse ES cell self‐renewal. LIF
mediates activation of STAT3 through the recruitment of Janus‐
kinases (Jaks). 7
Figure I.2b: LIF/Jak/STAT3 signaling regulates mouse ES cell
self‐renewal. 9
Figure I.2c: LIF activates the MAPK/ERK and PI(3)K/Akt
pathways in mouse ES cells. 10
Figure 1.1a: B6 Mouse ES cells enter crisis following feeder depletion. 23
Figure 1.1b: B6 mouse ES cells lose AP positivity in response to
feeder depletion. 24
Figure 1.2a: Feeder‐independent 46C self‐renew effectively in
the absence of feeders. 26
Figure 1.2b: Feeder‐independent 46C mouse ES cells increase
STAT3 expression and activation in response to LIF stimulation
as compared to B6 mouse ES cells. 27
Figure 1.2c: Densiometry indicates that 46C mouse ES cells
increase STAT3 expression following LIF treatment. 28
Figure 1.2d: Densiometry indicates that 46C mouse ES cells
increase STAT3 phosphorylation following LIF treatment. 28
Figure 1.3a: STAT3 overexpression enhances feeder‐free
self‐renewal of B6 mouse ES cells. 30
Figure 1.3b: STAT3 overexpression prevents crisis and
promotes self‐renewal. 32
Figure 1.3c: Removal of STAT3 transgene results in decreased
total cell number and decreased AP positive colony formation. 33
Figure 1.3d: Loss of STAT3 overexpression from B6 STAT3
cells results in decreased AP positivity. 34
viii
Figure 1.3e: STAT3 overexpressing B6 mouse ES cells remain
undifferentiated in long‐term feeder‐free culture. 35
Figure 1.4a: 46C mouse ES cells exhibit lower total ERK1/2
expression and activation as compared to B6 ES cells. 37
Figure 1.4b: Densiometry reveals that 46C mouse ES cells
harbor decreased ERK1/2 expression and phosphorylation
as compared to B6 ES cells. 38
Figure 1.4c: STAT3 overexpression correlates to decreased
ERK1/2 phosphorylation. 39
Figure 1.4d: Densiometry reveals that STAT3 overexpression
correlates to decreased ERK1/2 phosphorylation in B6 ES cells. 40
Figure 1.4e: Inhibition of MAPK improves STAT3 mediated
feeder‐independent self‐renewal. 42
Figure 1.4f: MAPK inhibition increases feeder‐free self‐
renewal when STAT3 is overexpressed. 43
Figure 1.4g: STAT3 overexpression increases RNA expression
of core pluripotency genes. 44
Figure 1.4h: STAT3 overexpression prevents differentiation
in stat3/ mouse ES cells cultured feeder‐free. 46
Figure 1.4i: STAT3 expression is required for LIF‐mediated
self‐renewal feeder‐free. 48
Figure 1.5a: β‐catenin activation does not significantly
enhance feeder‐free B6 self‐renewal in the absence of LIF. 50
Figure 1.5b: β‐catenin Activation Does Not Significantly Increase
Self‐Renewal when despite LIF stimulation and MAPK inhibition. 51
Figure 1.5c: Feeder‐free self‐renewal ability is impaired following
β‐catenin transgene excision. 52
Figure 2.1a: G‐CSF mediated STAT3 activation exerts a dose‐
dependent self‐renewal effect. 55
ix
Figure 2.1b: Densiometry reveals that STAT3 phosphorylation
levels decrease in correlation with decreasing G‐CSF concentrations. 56
Figure 2.1c: Jak inhibition decreases STAT3 mediated mouse ES
cell self‐renewal efficiency feeder‐free. 57
Figure 2.2b: STAT3 activation promotes feeder‐independent self‐
renewal of BalbC1 mouse ES cells. 59
Figure 2.2c: Feeder‐independent 46C mouse ES cells exhibit a
dose‐dependent self‐renewal response to G‐CSF treatment. 61
Figure 3.1a: G‐CSF mediated STAT3 phosphorylation is increased
and sustained over 24 hours. 63
Figure 3.1b: Densiometry reveals that G‐CSF treated mouse ES
cells retain an increased STAT3 phosphorylation level over 24 hours. 64
Figure 3.1c: Densiometry reveals that G‐CSF treated mouse ES
cells retain a stable STAT3 expression level over 24 hours. 64
Figure 3.1d: G‐CSF activates STAT3 in proportion to STAT3
expression level. Equal total cellular protein from B6 mouse
ES cells was compared via western blot analysis. 66
Figure 3.2a: SOCS3 RNA expression levels do not appear to
significantly differ between LIF or G‐CSF treatments. 68
Figure 3.3a: G‐CSF mediated STAT3 activation correlates
to rapid induction of mouse ES cell differentiation. 70
Figure 3.4a: Feeder‐free maintenance of B6 mouse ES cells
in G‐CSF conditions maintains pluripotent identity. 73
Figure 3.4b: G‐CSF treatment of mouse ES cells regulates a
distinct set of genes as compared to LIF. 74
Figure 3.4c: B6 mouse ES cells maintain pluripotency following
long‐term culture feeder‐free. 76
Figure 3.5a: DAc8 rat ES cells activate STAT3 in response
to LIF stimulation. 80
Figure 3.5b: Densiometry comparing rat and mouse ES
cell STAT3 levels. 80
x
Figure 3.5c: STAT3 overexpression promotes LIF mediated
self‐renewal of rat ES cells. 82
Figure 3.5d: G‐CSF supports rat ES cell self‐renewal. 83
Figure 3.5e: G‐CSF mediated STAT3 activation increases rat
ES cell self‐renewal ability over 2i. 84
Figure 3.5f: Rat ES cells expressing the chimerical gp130 receptor
activate STAT3 in response to G‐CSF. 85
Figure 3.5g: Densiometry indicates that G‐CSF and LIF
phosphorylate STAT3 comparably in rat ES cells. 86
Figure 3.5h: G‐CSF stimulation sustains STAT3 activation over
time promoting long‐term self‐renewal. 87
Figure 3.5i: Densiometry indicates that G‐CSF increases STAT3
activation over 24 hours in rat ES cells. 88
Figure 3.5j: Densiometry indicates that G‐CSF maintains STAT3
expression over 24 hours in rat ES cells. 88
Figure 3.5k: G‐CSF maintained rat ES cells maintain a pluripotent
identity similar to canonical 2i maintained rat ES cells. 89
Figure 4.1a: Mouse ES cells upregulate Klf4 in response to
G‐CSF treatment feeder‐free. 93
Figure 4.3a: Klf4 overexpression enhances LIF‐mediated feeder‐
free B6 ES cell self‐renewal. 95
Figure 4.3b: Klf4 overexpression permits long‐term culture
feeder‐free in a LIF‐dependent manner. 96
Figure 4.3c: Klf4 overexpression enhances BalbC1 mouse ES
cell feeder‐free self‐renewal. 97
Figure 4.3d: Klf4 knock‐down is efficient in mouse ES cells. 98
Figure 4.3e: Klf4 expression is not essential for mouse feeder‐
free self‐renewal. 99
xi
Figure 4.3f: Klf4 knock‐down decreases pluripotency gene
expression and increases early differentiation markers. 100
Figure 5.1a: G‐CSF Mediates STAT3 activation and
correlates to inhibited ERK1/2 activation. 103
Figure 5.1b: Densiometry confirms decreased ERK1/2
activation in response to G‐CSF in HT29 tumor cells. 104
Figure 5.1c: Wild‐Type Human Tumor Cell Lines Demonstrate
No Discernible Effect to G‐CSF Treatment. 105
Figure 5.1d: Increased STAT3 Activation Correlates to
Decreased Cell Number in DLD1‐Y118F cells. 106
Figure 5.1e: Increased STAT3 activation correlates to
decreased cell number in HT29‐Y118F cells. 107
Figure 5.2a: G‐CSF stimulation correlates to decreased
cumulative cell number. 109
Figure 5.2b: G‐CSF Prevents Tumor Cell Proliferation at
Low Cell Density and During Exponential Growth. 110
Figure D.2a: STAT3 signaling appears to have a functional
range that effectively promotes ES cell self‐renewal. 112
Figure D.4a: Rodent ES cell self‐renewal is metastable cell‐
state within naive pluripotency. 119
Figure D.4b: Self‐renewal represents a finely‐tuned interplay
between STAT3 and MAPK/ERK signaling in rodent ES cells. 121
xii
Abstract
Pluripotent embryonic stem (‘ES’) cells are typically derived and maintained using
inductive or inhibitory signals that are thought to behave in a binary ‘on/off’
manner. Mouse ES cells are maintained in an excess of leukemia inhibitory factor
(LIF) and rat ES cells in a cocktail of inhibitors that block both GSK3β and MAPK
signaling in a STAT3‐independent manner. Here, we provide evidence that both
mouse and rat ES cell self‐renewal is conserved via a STAT3‐dependent mechanism
that treats STAT3 activation as fluid and dynamic. We observe that increased Klf4
expression enhances STAT3‐mediated self‐renewal in mouse ES cells but is not
essential to prevent differentiation. As well, increased STAT3 activation and ERK1/2
inhibition synergistically enhance self‐renewal. In all, we propose that the self‐
renewal mechanism is a nested metastable cell‐state within naïve pluripotency, and
that further refinements to the self‐renewal mechanism will allow for direct inter‐
species comparisons and derivation of novel ES and induced pluripotent cell
population.
1
Introduction
Modern advances in science are predicated upon the unconventional insights of
previous generations. In order to make clear predictions about the world we
inhabit, scientists must diligently understand how and why the current path has
been laid down.
I.1 An Overview of the History of Embryonic Stem Cell Research
Embryonic stem (ES) cells are in vitro cell artifacts that represent a developmentally
halted tissue type of the pre‐implantation mammalian blastocyst(Evans and
Kaufman, 1981; Martin, 1981). ES cells have the peculiar, yet useful, properties of
nearly infinite in vitro self‐renewal and the ability to differentiate into any tissue
type of the soma and germline, termed ‘pluripotency.’ The concept of harnessing
pluripotency in vitro arose from early work upon the hematopoietic system.
Researchers at that time realized that transplanted hematopoietic cells would give
rise to spleen colonies of hematopoietic cells. In 1964, Till and McCulloch concluded
that these cells were a progenitor, or ‘stem,’ cell type that, from single cells,
reconstitute a whole population within a tissue(Till et al.). With the availability of
mouse ES cells in 1981, researchers hypothesized that precise gene targeting
experiments could be possible, and therefore specific gene functions might be
2
interrogated(Thomas and Capecchi, 1987). For the discovery of mouse ES cells and
the ability to create gene‐targeted animals via homologous recombination using ES
cells, Sir Martin Evans , Mario Capecchi and Oliver Smithies were awarded the Nobel
Prize in medicine in 2007, highlighting the importance of ES cell research and the
translation of these ideas to in vivo systems.
However Sir Evans’ work was just the culmination of many researchers’ previous
efforts toiling away on a curious phenomenon observed in germ cell tumors found
in the 129 strain of mouse. In 1954, Stevens and Little characterized these tumor
cells from this mouse as a relatively frequent phenomenon, occurring at around 1%
in the testes(Brook and Gardner, 1997). The tumor types that resulted from these
cells, termed teratocarcinoma and embryonal carcinoma (‘EC’), were odd in that not
only did they harbor the characteristic of being able to grow indefinitely in a culture
dish in an undifferentiated state, a phenomenon referred to as ‘self‐renewal,’ but
also when single EC cells were transferred from one animal to another, the tumor
could be reconstituted fully and retain the ability to differentiate into tissue types
representative of all germ layers of the soma(Kleinsmith and Pierce, 1964). This
latter characteristic, termed ‘pluripotency,’ is a defining aspect of authentic
embryonic stem cells.
Maturation, or differentiation, of tumor cell types is not in itself a novel finding,
however the investigators working on embryonal carcinomas were astute enough to
note that EC cells share many cell surface antigens and markers of cells of the
3
embryonic inner cell mass(Gachelin et al., 1977; Solter and Knowles, 1978). These
tumors, either by their mechanism of transformation or by their tissue of origin,
exhibited a novel characteristic that conferred both self‐renewal and multipotent
differentiation(Strickland and Mahdavi, 1978).
Researchers were aware that, if transplanted to ectopic sites, mouse embryos could
give rise to teratomas.
Borrowing from oncology
studies, researchers routinely
injected EC cells into the kidney
capsule as a means of providing
an environment for their
pluripotency to be
observed(Bogden et al., 1979;
Castro and Cass, 1974). EC cells
were routinely injected in
developing mouse blastocyts
where the tumor cells would
colonize the developing
conceptus, eventually forming the mosaically patterned chimeras which form the
first portion of the assay used to rigorously demonstrate pluripotency: Germline
transmission(Illmensee and Mintz, 1976; Mintz and Illmensee, 1975). This result
Figure I.1a: Embryonic stem cells permit precise
genetargeting.(1) A preimplantation mouse
blastocyst is subjected to immunosurgery. (2) The
resulting ES colonies emerge and proliferate. (3)
Genetargeting by homologous recombination is
performed. (4) Modified ES cells are injected into a
host blastocyst and transferred into a donor
mouse. (5) Coat color contribution determines ES
penetrance. (6) Mating of chimera results in
germline transmission of transgene in offspring.
4
encouraged researchers because if the rate of transmission could be improved
somehow, it could become feasible to completely generate a genetically‐modified
organism by simply altering the gene‐of‐interest in a single pluripotent embryonic
‘stem’ cell which would then be allowed to re‐enter the normal developmental
pathway (Figure I.1a).
It was Evans and Martin’s insight that led to the discovery of a karyotypically
normal cell type from an in vivo source that could recapitulate the tumor cell’s
abilities without the undesirable side effects of using cancer(Martin, 1980). It is
interesting to note that as of now, the in vivo equivalent of ES cells has yet to be
definitively determined, although most evidence indicates that the pre‐implantation
epiblast is the likely equivalent to ES cells(Gardner and Beddington, 1988; Lanza et
al., 2006). ES cells represent a developmentally frozen cell state that retains nearly
all of the properties of the pluripotent epiblast.
Following fertilization and formation of the zygote, cleavage division begins under
the supervision of mainly inherited factors and imprinted genes(Surani, 2001).
Pluripotency markers, such as Oct4, are inherited transcripts that then give way to
endogenous production as early as the 2‐cell stage (Hansis et al., 2001). Cells then
begin to partition the embryonic compartment, as indicated by differential, outer
cell layer expression of cdx2, a marker for trophectodermal lineage, and Oct4
expression located within the inner cell layer(Suwińska et al., 2008). There is even
evidence that pluripotency markers are partitioned immediately after fertilization,
5
suggesting that the pluripotency compartment is marked and separated early in
development(Lu et al., 2001; Piotrowska‐Nitsche et al., 2005).
Cell potency refers to the total number of germ layers each cell can form. In addition,
potency refers to how many differentiated cell types within each tissue layer each
cell can form. Totipotency indicates the ability for an individual cell to form all
tissue types of the soma including all extraembryonic tissues(Lanza et al., 2006).
Dichorionic identical twins are a natural result of totipotent stem cells that separate
in early cleavage divisions to form separate, genetically identical embryos.
Pluripotency is generally regarded as a property held by the epiblast alone in
normal development(Grubb, 2006).
I.2 Mouse ES Cells Use the LIF/STAT3 Pathway To SelfRenew
Embryonic stem cells have two defining characteristics: Self‐renewal and
pluripotency. While the differentiation potential and characteristics of pluripotency
have remained a popular area of research, investigating the mechanism as to why ES
cells decide to divide into two identical daughter cells as opposed to differentiated
progeny has remained relegated to a small cadre of groups. A thorough
understanding of ES cell self‐renewal will likely result in derivation of ES cells in
novel species. In addition, by investigating self‐renewal, clinical applications that
use ES cells will benefit from the ability to completely terminate and regulate the
self‐renewal program, a finding that may be applied to both ES‐based therapies and
6
cancer studies. And finally, with the recent discovery of induced pluripotent stem
(iPS) cells, understanding how stem cells decide to self‐renew is tantamount to
understanding efficient and complete reprogramming(Takahashi and Yamanaka,
2006).
Self‐renewal is defined as the ability to divide into two identical daughter cells that
retain the pluripotency properties of the parental cell(Lanza et al., 2006).
Interestingly, self‐renewal in ES cells is coupled to unlimited proliferative ability. ES
cells appear to lack a Hayflick limit and never appear to exit the cell cycle,
demonstrating persistent cyclin D expression, spending the majority of the cell cycle
in S phase after an extremely abbreviated G1 stage(Burdon et al., 2002; Dalton,
2009; Singh and Dalton, 2009). Therefore mouse ES cells can be maintained without
the worry of senescence (Smith, 2001). In addition, telomerase is active in ES cells
and is required for cellular reprogramming, suggesting that telomerase activity aids
in proliferation of self‐renewing ES cells(Lee et al., 2005; Niida et al., 2000;
Takahashi and Yamanaka, 2006).
In the mouse, self‐renewal is mediated primarily by the LIF/Jak/STAT3
pathway(Niwa et al., 1998b) (Figure I.2a). Leukemia Inhibitory Factor (LIF) is an IL‐
6 family cytokine that is secreted by the trophectoderm in vivo to promote diapause:
Diapause is the phenomenon whereby a suckling mouse triggers a delay in
blastocyst implantation that can last for several weeks. Estrogen mediates
trophectodermal secretion of LIF, which in turn induces self‐renewal of the inner
7
cell mass (ICM) until implantation occurs and development continues(Mead, 1993;
Renfree and Shaw,
2000).
In vitro, LIF is
secreted by the
support layer of
mitotically‐
inactivated cells
used to anchor and
maintain mouse ES
cells, termed
‘mouse embryonic
fibroblasts’
(‘MEFs’) or ‘feeders’(Rathjen et al., 1990; Smith and Hooper, 1987). LIF secretion
acts in paracrine to bind and co‐localize the two necessary cell surface receptors:
LIFRβ and gp130(Chambers et al., 1997; Gearing et al., 1991). LIFRβ and gp130 are
non‐tyrosine kinase receptors that, in the presence of LIF, heterodimerize(Davis et
al., 1993). The cytosolic domains then catalyze a series of signal transduction
pathways that execute self‐renewal for the mouse.
Figure I.2a: STAT3 regulates mouse ES cell selfrenewal. LIF
mediates activation of STAT3 through the recruitment of
Januskinases (Jaks). STAT3 activation promotes self
renewal and ‘stemness.’ LIF also activates ERK1/2 which
aids STAT3 in proliferation, but promotes differentiation as
well.
8
Gp130 is the common IL‐6 family receptor. It frequently heterodimerizes with other
IL‐6 family members, as well as harboring homodimerization ability(Murakami et
al., 1993). Gp130‐/‐ mutant embryos fail to develop past implantation and give rise
to predominantly parietal endoderm(Nichols et al., 2001). It is therefore important
that gp130 be expressed for both ES maintenance and for normal embryonic
development. LIFRβ appears to be unnecessary for blastocyst formation and the
formation of the pluripotent ICM, as LIFRβ‐/‐ embryos develop to term, but die
perinatally with severe neural defects(Li et al., 1995; Ware et al., 1995). LIF‐/‐
embryos are normal into adulthood, but LIF‐/‐ mothers fail to support blastocyst
diapause and implantation(Stewart et al., 1992). In all, LIF appears to be important
as a secreted factor by both the mother and trophectoderm to support pluripotency.
LIF, once engaged with the receptor complex, recruits a multitude of proteins to the
cytosolic scaffolding. Most importantly, Janus kinase 2 (Jak2) is recruited to the
membrane proximal domain(Narazaki et al., 1994). Once docked, Jak2
phosphorylates Tyk, which in turn recruits inactive STAT3 monomers(Stahl et al.,
1994). While ES cells express several different isoforms, STAT3 is the most common
species present in mouse ES cells. STAT3 is a highly conserved acute‐phase
pleiotropic transcription factor, capable of exacting transcriptional regulation upon
phosphorylation at the cell membrane(Darnell et al., 1994).
9
STAT3 has several important functional domains important to its role in ES cells.
Composed of a DNA binding, SH2, coiled‐coil, and transactivation domains, STAT3
has the ability to both interact directly and as a co‐activator of transcription(Becker
et al., 1998; Zhong et al., 1994a). Within the transactivation domain lie two distinct
phosphorylation sites important to ES cells. In the mouse, serine 727 (‘Ser727’ or
‘S727’) phosphorylation has
been shown to enhance
transcriptional activity, likely by
providing a docking site for
interaction with other
proteins(Wen et al., 1995;
Yokogami et al., 2000).
Tyrosine 705 (‘Tyr705’ or
‘Y705’) phosphorylation is
associated with STAT3
activation(Zhong et al., 1994b).
Upon Jak2 docking, STAT3
monomers are recruited to one of the several STAT3 docking sites distal to the
membrane(Narazaki et al., 1994). Four distinct YXXQ motifs can bind to STAT3.
Y265/275 were found to be critical to maintaining the pluripotent state, as mutation
of these docking sites abolished mouse ES self‐renewal(Niwa et al., 1998a).
Figure I.2b: LIF/Jak/STAT3 signaling regulates
mouse ES cell selfrenewal. (1) LIF ligand binding
induces heterodimerization of both LIFRβ and
gp130. (2) Receptor complex formation
precipitates docking and phosphorylation of Jak2
and Tyk. (3) Jak2 recruits and phosphorylates
STAT3 monomers. (4) Active STAT3 dimer
complexes translocate to the nucleus and effect
gene transcription.
10
Jak2 phosphorylates STAT3 at Y705, which causes STAT3 to develop affinity for
other phosphorylated STATs(Hemmann et al., 1996). STAT3 preferentially
reciprocally binds to other activated STAT3 proteins by phospho‐SH2
interactions(Heim et al., 1995). This homodimer forms the active complex that then
immediately translocates to the nucleus whereby transcriptional regulation occurs
(Figure I.2b).
STAT3’s role in mammalian
cells is broad. In mouse ES cells,
STAT3 is the central protein
that maintains ‘stemness’ and
self‐renewal. However, when
STAT3 was first discovered in
tumor cell lines, this protein
demonstrated it was capable of
responding to a variety of
cytokines, namely, IFNγ, EGF,
LIF, and IL‐6(Zhong et al.,
1994a). In both ES and somatic
tissues, STAT3 activation
mediates cellular proliferation and anti‐apoptosis(Fukada et al., 1996). In ES cells
the G1 checkpoint does not appear to exist, suggesting that mouse ES cells employ
Figure I.2c: LIF activates the MAPK/ERK
and PI(3)K/Akt pathways in mouse ES cells.
LIF engagement promotes docking an
activation of the phosphatase SHP2, which
in turn activates MAPK signaling. MAPK
activation results in ERK1/2 activation,
which regulates proliferative and
differentiation signals. Concurrently, SHP2
activates PI(3)K which phosphorylates Akt.
Akt appears to regulate cell cycle and
ERK1/2 activation.
11
STAT3 to regulate the cell‐cycle in a novel manner(Savatier et al., 1996). Again in
both ES and somatic cells, STAT3 promotes pro‐survival signals by regulating
expression of c‐Myc, Src, and Bcl family genes(Bromberg et al., 1998; Kiuchi et al.,
1999). STAT3 is ubiquitously expressed in mammalian tissues, therefore its
dysregulation is common in specific cancers(Corvinus et al., 2005).
Once activated by Jak2 in mouse ES cells, STAT3 dimers translocate to the nucleus.
Concurrent to this activity, Jak2 also phosphorylates Y759 upon the gp130 receptor
which recruits the protein phosphatase SHP‐2(Symes et al., 1997). SHP‐2 then
activates the Ras/MAPK/ERK pathway(Boulton et al., 1994). In mouse ES cells
MAPK activation is associated with decreased cellular proliferation and
differentiation(Burdon et al., 1999; Kunath et al., 2007). ERK2/ mouse ES cells can
be derived and maintained with reduced proliferative potential(Yao et al., 2003;
Ying et al., 2008a). ERK1/ mice are viable suggesting ES cells viable as well. Yet
ERK1/2 double knock out mouse ES cells have yet to be derived(Selcher et al., 2001).
Unlike STAT3, ERK activation occurs through a step‐wise regulatory pathway
culminating in transcriptional control (Figure I.2c).
The third arm of the LIF signaling pathway begins with SHP‐2 phosphorylation by
Jak2, which activates the phospho‐inositol (3‐OH) kinase (PI(3)K) pathway(Boulton
et al., 1994). PI(3)K is a highly conserved pathway that regulates many cellular
processes. In mouse ES cells PI(3)K activates Akt, which in turn acts as a pro‐
survival signal(Sun et al., 1999). There are conflicting reports as to the definitive
12
role of Akt in mouse ES cells. While promotion of Akt signaling has been
demonstrated to augment mouse ES cell self‐renewal in combination with LIF
signaling, inhibition of Akt shows no significant deleterious effects upon self‐
renewal(Paling et al., 2004). STAT3 inhibition, either by dominant negative
mutation, Jak2 inhibition, or siRNA‐mediated knock‐down causes immediate mouse
ES cell differentiation(Niwa et al., 1998b).
Burdon et al. demonstrated clearly that STAT3 is the key effector in mediating
mouse ES cell self‐renewal(Burdon et al., 1999). By mutating the essential SHP‐2
docking site at tyrosine 118 they determined that STAT3 alone is sufficient to
promote self‐renewal by inhibiting LIF‐mediated ERK1/2 and Akt signaling. When
mutated to a dominant negative form, STAT3 cannot promote self‐renewal in the
absence of ERK1/2 and Akt signaling despite the presence of LIF/Jak signaling.
Burdon et al. also treat STAT3 activation as a binary ‘On/Off’ system. In this case LIF
is either present and active, or absent and off.
It is the aim of this work presented here to address the question surrounding STAT3’s
role in regulating the selfrenewal mechanism. I hypothesize that STAT3 regulates
self‐renewal not by a binary system, but rather as having a range of activity that
promotes self‐renewal. Precedent for this idea has been demonstrated by Niwa et al.
They have demonstrated that Oct4 expression is not binary, but rather tightly
regulated to fall within a specific range, outside of which results in rapid
differentiation or death of mouse ES cells(Niwa et al., 2000). This mechanism
13
however applies specifically to pluripotency regulation. Separate from the
pluripotency mechanism, we propose that STAT3 activation regulates self‐renewal
in a similar manner.
Also, much is known about STAT3 with regards to mouse ES cells, however little has
been done to investigate the role STAT3 plays in regulating rat ES cells. Our group
has reported upon the derivation of germline competent rat ES cells, but to date,
these cells can only be maintained by inhibiting inductive differentiation cues in a
STAT3‐independent manner(Li et al., 2008). This ‘ground state,’ permits self‐
renewal by limiting MAPK activation in the absence of STAT3 activation(Ying et al.,
2008b). Also important to promoting ground state self‐renewal is inhibition of
glycogen synthase kinase 3β (GSK3β) which appears to mimic Wnt/β‐catenin
signaling(Sato et al., 2004). The role of β‐catenin is likely essential for both mouse
and rat ES cell self‐renewal but has yet to be determined mechanistically.
I.3 Permissiveness Relates to Metastable Pluripotent CellStates
The first mouse ES cell lines derived were isolated from 129 strain mice(Evans and
Kaufman, 1981; Martin, 1981). Whether by chance or premeditation, the
investigators chose a strain of mice that have the peculiar ability to be derived and
maintained either in co‐culture with mitotically‐inactivated mouse embryonic
fibroblasts (‘MEFs’ or ‘Feeders’) or simply a gelatinized substrate alone(Nichols et
14
al., 1990). Initially, many successful ES derivations made use of conditioned medium
collected from embryonal carcinoma cells, a transformed pluripotent cell type
capable of self‐renewal and differentiation. Also presumed at the time was that
factors within the conditioned medium and/or the feeders were of importance for
maintenance of the undifferentiated state. However, it was found that the feeders
could be removed when deriving most 129 strain ES cells as long as the conditioned
medium was present. No other mouse nor rat strain has been routinely derived in
the absence of feeders with any significant efficiency and are therefore called
‘feeder‐dependent’(Gardner and Brook, 1997; Kawase et al., 1994; Li et al., 2008).
129 mouse ES cells that are able to self‐renew upon a gelatinized substrate alone are
referred to as ‘feeder‐independent.’
Recently, evidence has postulated that pluripotency itself is a stratified cell‐state
based on each strain’s or species’ inherent ‘permissiveness.’ It is proposed that
mammalian pluripotency consists of at least two metastable cell‐states(Hanna et al.,
2009). These states, termed ‘naïve’ and ‘primed’ pluripotency indicate distinct
molecular biases towards specific in vitro culture preferences(Nichols and Smith,
2009).
Naïve pluripotency refers to cells that express canonical ES cell transcription factors
such as Oct4, Nanog, and Sox2. In addition, self‐renewal is maintained by LIF/STAT3
signaling or in ground‐state culture conditions which are STAT3‐independent. In
addition naïve pluripotent cells retain two active X chromosomes and can
15
contribute to germline chimeras extensively(Nichols and Smith, 2009). Genetic
modification produced by homologous recombination in naïve pluripotency is more
efficient than primed pluripotent cells(Buecker et al., 2010). Mouse and rat
embryonic stem cells are thought to harbor naïve pluripotency.
Primed pluripotent stem cells can be derived from the post‐implantation murine
egg‐cylinder of mice at 6.5 dpc. These cells express the same canonical pluripotency
markers such as Oct4 and Nanog, but also up‐ and downregulate a specific subset of
genes more indicative of stem cells that have prepared to differentiate(Tesar et al.,
2007). Despite this, primed ES cells exhibit robust in vitro self‐renewal. Rex1, a
marker of the inner cell mass and ES cells, is absent. FGF5, a canonical marker of
early differentiation that is not expressed in ES cells, is significantly upregulated in
primed pluripotent cells(Brons et al., 2007). Most telling, however, is that X‐
chromosome inactivation has occurred, suggesting a unique level of epigenetic
changes has occurred that may be distinct from naïve pluripotent cells(Nichols and
Smith, 2009). Primed pluripotent cells becomes refractory to LIF/STAT3‐mediated
self‐renewal and efficient germline transmission has yet to be demonstrated.
However, these cells are at least in vitro pluripotent and can form teratomas. Mouse
primed pluripotent cells, termed ‘EpiSCs,’ self‐renew only in the presence of
bFGF/Activin signaling, and differentiate in the presence of ground state culture
conditions(Tesar et al., 2007). Primed EpiSCs can be derived from ES cells via
bFGF/Activin A induction in culture(Brons et al., 2007). Reprogramming back to
naïve pluripotency has been demonstrated with ectopic expression of Klf4,
16
suggesting that epigenetic changes to the core pluripotency network are essential in
the transition between cell‐states(Guo et al., 2009).
Permissiveness refers to the idea that each mouse strain or species harbors
different inherent levels of restrictions that permit switching between naïve and
primed pluripotency(Hanna et al., 2009). Pertinent to our work, it has been detailed
that 129 mouse ES cells represent the most permissive ES cell line known currently.
Determining permissiveness relies upon a combination of criteria such as the
efficiency of derivation, germline contribution, and reprogramming
efficiencies(Hanna et al., 2009). Permissive lines also appear to demonstrate feeder‐
independent self‐renewal with no apparent ill‐effects to pluripotency.
‘Non‐permissive’ mouse strains are described as historically difficult strains to
derive germline competent ES cells and their relative recalcitrance to retain self‐
renewal in feeder‐free culture systems(Hanna et al., 2009). Examples of such strains
form the majority of the ‘feeder‐dependent’ ES cells such as NOD background,
C57BL/6, and Balb/c. Little work has gone into understanding why such differences
exist.
Currently, the least permissive species is human. Recent work has presented
evidence that human ES cells currently in use may be more accurately characterized
as EpiSC‐like based on the current classification system used in rodents(Nichols and
Smith, 2009). It has been shown that human iPS cells can be reprogrammed back to
17
naïve pluripotency with extensive genetic modification and heavily modified culture
conditions(Buecker et al., 2010; Hanna et al., 2010). This inefficient process results
in cells that become LIF/STAT3 sensitive for self‐renewal, can be cultured in
ground‐state conditions, and express transcriptional markers indicative of mouse
and rat ES cells. In addition, epigenetic changes are apparent, as the reprogrammed
human iPS cells restore their X chromosomes to active states. In addition, gene‐
targeting efficiency via homologous recombination is significantly higher in the
mouse ES‐like human iPS cells as compared to human EpiSC‐like cells. However,
germline transmission testing is impermissible on human pluripotent cells,
therefore a full and comprehensive analysis of the identity of these cells is likely
untenable unless it is shown that ES cell lines can be derived and propagated from
human blastocysts using the culture conditions and concepts proposed by these
investigators.
Permissive mouse strains are currently limited to the 129. One hallmark feature that
makes this strain popular for researchers is their unique ability to self‐renew
feeder‐free. The reason why 129 strain mouse ES cells can be maintained feeder‐
free has remained scientifically understudied. Fundamental advances in the last
twenty years using 129‐derived mouse ES cells have illuminated several key
characteristics of ES cell biology: The identification of LIF as the main arbiter of
mouse ES cell self‐renewal may be the most important thus far. LIF’s isolation and
characterization by Dr. Austin Smith codified the role of extracellular signals in
promoting mouse ES cell self‐renewal by demonstrating that the feeder cell layer is
18
the predominant in vitro source that secretes LIF in paracrine to activate STAT3
within mouse ES cells(Nichols et al., 1990; Smith and Hooper, 1987). The
conditioned medium as well was later determined to contain LIF. LIF activates
STAT3 primarily, which in turn regulates the self‐renewal program and helps to
maintain the undifferentiated state.
Dr. Smith then demonstrated that LIF alone could regulate self‐renewal(Nichols et
al., 1990). LIF was cloned and recombinant versions were found to be sufficient in
promoting self‐renewal when supplemented in serum containing non‐conditioned
medium and feeders. However, it was observed that, again, mainly 129 strain mouse
ES cells could successfully self‐renew in the absence of the feeder cells. All other
strains were observed to either die or differentiate. This phenomenon was first
noticed by Sir Martin Evans, and he appropriately entitled it “ES cell crisis(Nagy et
al., 2003).” No further progress has been made into determining the mechanism by
which the feeders augment self‐renewal in non‐129 mouse ES cells.
By comparison, 129 strain ES cells are employed frequently for basic research
because of their convenience and the simplicity of the culture system. Although ES
strains such as C57BL/6 mice are a popular strain for researchers, their ES cells are
less so due to their feeder‐dependence, sensitivity to minute culture changes, and
relatively low derivation efficiencies when compared to 129 strain ES cells(Brook
and Gardner, 1997).
19
I was charged with investigating the role the feeders played in promoting self‐
renewal. If the feeders are indeed critical in maintaining the self‐renewal of all but
129 strain mouse ES cells, could the function of the feeders be dissected? I reasoned
that 129 mouse ES cells’ ability to selfrenew feederfree was tethered to their
increased permissiveness as compared to other mouse strains. I hypothesized that
either the feeders provide a necessary signaling supplement to feeder‐dependent ES
cells or that 129 strain ES cells have an innate advantage or acquired mutation that
supplements or replaces the feeders’ function. This advantage seemed unique to
129 ES cells, and therefore, I reasoned that if we could dissect this mechanism, ‘non‐
permissive’ mouse ES cells could be rendered feeder‐free with no ill‐effects to their
pluripotency.
I.4 Rationale for This Investigation
It has been established that conditioned medium alone is not sufficient to promote
self‐renewal of feeder‐dependent mouse ES cells(Nichols et al., 1990). It appears
that feeder‐dependent self‐renewal is indeed a contact mediated effect, although we
cannot rule out short‐range paracrine cytokine interactions nor a highly unstable
factor/small molecule. The latter scenario is unlikely however, given the extent of
work performed to analyze conditioned medium(Lim and Bodnar, 2002). However
the dearth of knowledge in this area of self‐renewal and the multitude of possible
factors to assay rendered this line of testing unfeasible. Many cytokines have been
tested as a possible source of self‐renewal, however, none of these cytokines can
20
replace the function of LIF in vitro. We reasoned that studying the differences
between both 129 and feeder‐dependent mouse ES cells a more expedient manner
in which to study the self‐renewal mechanism.
In this work, I will employ a metric referred to as the ‘crisis assay.’ The assay’s main
purpose is to measure self‐renewal ability in feeder‐free culture systems. We
reasoned that efficient long‐term feeder‐free self‐renewal of mouse ES cells in a
simplified culture system is an effective manner to compare different mouse ES cell
strains.
I.5 Conclusions
I conclude that increased STAT3 activation can maintain pluripotent populations of
both mouse and rat ES cells through a shared STAT3‐dependent mechanism.
Increased STAT3 activation that is sustained over time mediates both mouse and rat
ES cell self‐renewal. This effect is synergistically enhanced when the MAPK‐ERK1/2
pathway is inhibited. I posit that STAT3’s role in rodent ES cell self‐renewal is far
more sophisticated and refined that once thought, and lack of knowledge of this
specific level of regulation may have impeded past attempts at deriving rodent ES
cells. We also conclude that increased Klf4 expression enhances mouse ES cell
feeder‐free self‐renewal in a LIF‐dependent fashion. Klf4 depletion renders mouse
ES cells capable of feeder‐free self‐renewal only when STAT3 activation is increased.
As such we note decreased pluripotency marker expression and increased early
21
differentiation marker expression when Klf4 is depleted, suggesting that Klf4
expression reinforces naïve pluripotency and that depletion ‘primes’ mouse ES cells
for differentiation.
We posit that the self‐renewal mechanism, like pluripotency, is itself stratified into
distinct cell‐states. These cell‐states can transition easily between different ranges
of STAT3 activation and STAT3‐independent pathways within naïve pluripotency.
This model predicts that there are different rodent self‐renewal mechanisms which
are themselves stratified within a specific pluripotent cell‐state.
It is our hope the findings presented here accelerate the derivation and simplify the
maintenance of naïve pluripotent rodent cell populations. In addition, when
attempting to derive or maintain naïve pluripotent mammalian cells, one must now
consider using STAT3 in a species‐specific manner rather than uniformly. Also, one
must decide whether employing a STAT3 independent mechanism or a STAT3
dependent system relying upon cytokine induction of self‐renewal is appropriate.
22
Results
Chapter One STAT3 Overexpression Enhances FeederIndependent Mouse ES
Cell SelfRenewal
1.1 – B6 Mouse ES Cells Die or Differentiate Upon Feeder Depletion
We first tested if there was a quantifiable difference when C57BL/6, hereto after
referred to as ‘B6,’ mouse ES cells were maintained upon feeders or feeder‐free. B6
mouse ES cells were seeded at clonal density, approximately 5x10
3
cells/cm
2
, either
upon a gelatinized substrate or in co‐culture with CF‐1 feeder cells and
supplemented with both serum and recombinant LIF. B6 cells exhibit a significant
decrease in alkaline phosphatase (AP) positivity after seven days of incubation
(Figure 1.1a). AP expression is a general marker of undifferentiated, self‐renewing
ES cell colonies. Both cell populations were passaged and AP positive colonies were
undetectable when the feeders were absent (Figure 1.1b). This finding suggests that
LIF is likely insufficient in promoting B6 mouse ES cell self‐renewal feeder‐free.
23
Figure 1.1a: B6 Mouse ES cells enter crisis following feeder depletion. (A)
B6 wildtype mouse ES cells seeded at clonal density (5x10
3
cells/cm
2
) in
coculture with CF1 feeders in serum medium supplemented with
10ng/ml LIF. (B) The same cells one day following passage into feeder
free conditions begin to die or differentiate despite serum and LIF. (C)
Cells from (B) one day following a second passage. Cells appear flattened
with little to no apparent proliferation observed.
24
Figure 1.1b: B6 mouse ES cells lose AP positivity in response to feeder
depletion. (A) 10
3
B6 mouse ES cells were seeded either upon a
gelatinized substrate or in coculture with CF1 feeders in a 4well plate.
Each condition was supplemented with serum medium and 10ng/ml LIF.
Following a seven day incubation, the cells were fixed and stained for
alkaline phosphatase (AP). (B) Photo demonstrating decreased AP
stained B6 mouse ES cells cultured upon gelatin (left panel) as compared
to the same cells cultured with feeders (right). Asterisk indicates pvalue
<0.05, Error bars indicate SD.
25
1.2 – 129 FeederIndependent Mouse ES Cells Increase STAT3 Expression and
Activation In Response to LIF Stimulation
We then investigated how 129 derived mouse ES cells avoid crisis and promote
feeder‐free self‐renewal. We began by directly comparing 46C feeder‐independent
mouse ES cells to B6 mouse ES cells. 46C mouse ES cells are a derivative cell line
from the parental 129 strain(Ying et al., 2003). We observe that these cells exhibit a
significantly higher proportion of AP positive colonies as compared to B6 mouse ES
cells when cultured in a feeder‐independent manner (Figure 1.2a). Following
seeding upon a gelatinized substrate at clonal density (5x10
3
cells/cm
2
), each cell
type was supplemented with 10ng/ml hLIF for 12 hours, after which the cells were
serum and cytokine starved for 6 hours. At this point serum medium was re‐applied
and again 10ng/ml LIF was added to the culture medium. Total protein cell lysates
were collected at one hour and twenty‐four hours post‐stimulation.
Western blotting reveals that the 46C mouse ES cells increase STAT3 expression
levels in response to LIF stimulation by twenty‐four hours post‐induction as
compared to B6 mouse ES cells (Figure 1.2b). In addition, 46C cells demonstrate
increased Tyr705 phosphorylation as compared to B6 mouse ES cells 30 minutes
post‐LIF stimulation. Densiometric analysis confirms this observation (Figure 1.2c‐
d). This finding suggests that feeder‐independent mouse ES cells may survive
feeder‐withdrawal by increasing both STAT3 expression and activation in response
to LIF.
26
Figure 1.2a: Feederindependent 46C selfrenew effectively in the absence of
feeders. 5x10
3
cells/cm
2
each of either B6 or 46C mouse ES cells were plated
upon a gelatinized substrate with either 10ng/ml LIF or medium alone
(‘NT’). Following a seven day incubation, the cells were fixed and AP stained.
46C mouse ES cells were able to significantly retain AP positive colonies as
compared to B6 cells when supplemented with LIF. Asterisk indicates p
value <0.05. Error bars indicate SD.
27
Figure 1.2b: Feederindependent 46C mouse ES cells increase STAT3
expression and activation in response to LIF stimulation as compared to
B6 mouse ES cells. (A) 10
5
cells each of B6 or 46C mouse ES cells were
seeded upon gelatinized substrates. One day later, each cell type was
stimulated with 10ng/ml LIF. Western blotting of the resulting cell
lysates reveals that 46C mouse ES cells increase STAT3 expression by 24
hrs postLIF stimulation as compared to B6 mouse ES cells. (B) 46C
mouse ES cells increase STAT3 Tyr705 phosphorylation in response to
LIF as compared to B6 mouse ES cells at 30 minutes poststimulation.
28
Figure 1.2c: Densiometry indicates that 46C mouse ES cells increase STAT3
expression following LIF treatment. Image analysis of Figure 1.2b indicates
that 46C mouse ES cells increase STAT3 expression almost twofold by 24
hours posttreatment. Data are normalized to tubulin expression. Scale is
linear.
Figure 1.2d: Densiometry indicates that 46C mouse ES cells increase STAT3
phosphorylation following LIF treatment. Image analysis of Figure 1.2b
indicates that 46C mouse ES cells increase STAT3 expression nearly twofold
within one hour of treatment. Data are normalized to tubulin expression.
29
1.3 – STAT3 Overexpression Prevents FeederDependent Mouse ES Crisis
We observe that 46C feeder‐independent mouse ES cells likely increase their STAT3
expression and activation in response to LIF treatment in feeder‐free culture. We
hypothesized that overexpressing STAT3 in feeder‐dependent mouse ES cells may
recapitulate the effects seen in the 46C mouse ES cells.
To test if increasing STAT3 expression may improve feeder‐free self‐renewal, we
stably introduced a constitutively active, high expression murine STAT3α vector
into B6 mouse ES cells. Following drug selection, individual clones were selected
and tested further. We again seeded equal total cell numbers of both 46C and the
STAT3 overexpressing B6 ES cells (‘B6‐STAT3’). Both cell types were seeded upon
feeders or gelatinized substrates and supplemented with 10ng/ml LIF. Seven days
later, AP staining revealed no significant difference between 46C and B6‐STAT3
mouse ES cells (Figure 1.3a). Western blotting confirms that, compared to wild‐type
cells, B6‐STAT3 cells do in fact exhibit increased STAT3 expression, as well as
significantly increased Tyr705 phosphorylation in response to LIF treatment.
30
Figure 1.3a: STAT3 overexpression enhances feederfree selfrenewal of
B6 mouse ES cells. (A) 5x10
3
cells/cm
2
each of either wildtype B6, 46C,
or STAT3 overexpressing B6 mouse ES cells were plated upon a
gelatinized substrate with either 10ng/ml LIF or medium alone (‘NT’).
Following a sevenday incubation, the cells were fixed and AP stained. B6
mouse ES cells overexpressing STAT3 significantly increase AP positive
colony formation as compared to wild type B6 mouse ES cells. 46C and
B6STAT3 mouse ES cells display a similar AP positive colony number
when LIF is applied. (B) Western blot indicating that STAT3 expression is
increased following STAT3α transfection. STAT3 phosphorylation
increased in response to LIF as well. Error bars indicate SD
31
To verify that the effect observed is due to STAT3 overexpression, the transgene
was excised from the genome via Cre recombinase expression. Complete excision
results in GFP positive expression as a result of the removal of a premature stop
codon upstream of GFP (Figure 1.3b). As indicated in Figure 1.3c, when compared to
wild‐type cells upon gelatinized substrates, GFP positive B6 mouse ES cells
demonstrate a marked loss of AP positivity after seven days incubation feeder‐free
despite LIF supplementation. This loss of AP positivity is consistent with wild‐type
B6 mouse ES cells when cultured feeder‐free, suggesting that the increased AP
positive colonies observed correlates to the increased STAT3 expression (Figure
1.3d). STAT3 overexpressing B6 mouse ES cells were able to be maintained in tight,
dome‐like colonies when cultured with LIF feeder‐free for ten passages and stained
positive for both Oct4 and Sox2 (Figure 1.3e). These findings suggest that STAT3
overexpression enhances feeder‐independent self‐renewal of B6 mouse ES cells in a
LIF‐dependent manner.
32
Figure 1.3b: STAT3 overexpression prevents crisis and promotes self
renewal. 5x10
3
cell/cm
2
each of wildtype or STAT3 overexpressing B6
mouse ES cells were seeded upon a gelatinized substrate or in coculture
with CF1 feeders. Cell were fixed and AP stained on Day 7. STAT3
overexpression enhances feederfree selfrenewal as indicated by AP positive
staining as compared to wildtype cells cultured with feeders and LIF.
Feederdepletion results in the loss of AP positivity. Phase contrast pictures
demonstrating STAT3 transgene removal following Cremediated excision.
33
Figure 1.3c: Removal of STAT3 transgene results in decreased total cell
number and decreased AP positive colony formation. (A) Equal total cell
numbers of either B6STAT3 cells or B6STAT3 cells that were
transfected with Cre recombinase. Cells were counted every day. STAT3
overexpressing cells increase their total cell number over five days, while
Cretransfected initially increase then decrease by day five. (B) Cells from
(A) were AP stained after five days. Cretransfected cells were observed
to exhibit decreased AP positive colonies as compared to STAT3
overexpressing cells.
34
Figure 1.3d: Loss of STAT3 overexpression from B6 STAT3 cells results
in decreased AP positivity. 5x10
3
cells/cm
2
of either wildtype or Cre
recombinanse transfected B6STAT3 cells were seeded upon
gelatinized substrates and incubated for seven days. The cells were
then fixed and AP stained. The removal of the STAT3 transgene by Cre
results in an inability to sustain selfrenewal feederfree. AP staining
demonstrates that this loss of selfrenewal is consistent with wild
type cells, suggesting that increased STAT3 expression is critical in
preventing crisis and promoting selfrenewal feederfree.
35
Figure 1.3e: STAT3 overexpressing B6 mouse ES cells remain
undifferentiated in longterm feederfree culture. (A) B6STAT3 cells
maintained upon gelatinized substrates in serum medium supplemented
with 10ng/ml LIF for five passages retain pluripotency markers Oct4 and
Sox2. (B) AP stain comparing entire wells demonstrate STAT3
overexpression mimics 46C mouse ES cell AP positivity.
36
1.4 Inhibition of MAPK and Activation of LIF/STAT3 Have a Synergistic Effect Upon
Preventing Mouse ES Cell Crisis
It has been established that MAPK/ERK signaling is antagonistic to LIF/STAT3
signaling for ES cell self‐renewal(Matsuda et al., 1999). Burdon et al. demonstrated
that ERK1/2 attenuation enhances STAT3 mediated self‐renewal(Burdon et al.,
1999). We hypothesized that ERK1/2 activation may play a significant role in
regulating feeder‐independent self‐renewal. To test this, we stimulated both 46C
and B6 mouse ES cells with LIF in feeder‐free conditions for 45 minutes. We
compared total ERK1/2 and phosphorylated ERK1/2 levels between strains. We
observe in Figure 1.4a that 46C mouse ES cells exhibit decreased total ERK1/2
expression as well as decreased ERK1/2 phosphorylation as compared to B6 ES
cells. Densiometric analysis confirmed this finding and reveals that while 46C ES
cells phosphorylate ERK1/2 in a similar proportion to total ERK expression as
compared to B6 cells, the overall decrease in ERK1/2 phosphorylation observed in
46C ES cells may augment feeder‐free self‐renewal (Figure 1.4b).
We then compared ES cells within the B6 strain to observe if the decreased ERK1/2
signaling was simply a product of strain differences. We compared B6 wild‐type ES
cells to B6‐STAT3 mouse ES cells to interrogate ERK1/2 expression and activation.
Equal total cellular protein was compared via western blot. STAT3 overexpressing
cells demonstrate decreased ERK1/2 phosphorylation regardless of treatment as
37
compared to wild‐type cells (Figure 1.4c). ERK1/2 expression levels remained
grossly equal between cell types, and did not appear to be affected by any treatment.
Densiometry analysis of the western blots suggests that of the two ERK isoforms,
ERK1 is reduced by a factor of approximately five (Figure 1.4d). As well, STAT3
overexpressing B6 cells demonstrate a slightly increased total ERK expression, yet
further testing is needed to firmly confirm or deny this observation. We interpret
this observation to mean that increased STAT3 expression may correlate to
decreased ERK1/2 phosphorylation, which may improve feeder‐free self‐renewal
ability.
Figure 1.4a: 46C mouse ES cells exhibit lower total ERK1/2 expression and
activation as compared to B6 ES cells. Equal total protein from wholecell
lysates were analyzed via western blot of both wildtype B6 and 46C mouse
ES cells. Following the addition of 1uM PD0325901, no detectable levels of
phosphorylated ERK1/2 were observed. 46C mouse ES cells demonstrate a
significantly lower ERK1/2 expression level as compared to B6 mouse ES
cells. NT indicates mediumonly treated cells.
38
Figure 1.4b: Densiometry reveals that 46C mouse ES cells harbor decreased
ERK1/2 expression and phosphorylation as compared to B6 ES cells. Image
analysis of Figure 1.4a indicates that 46C cells harbor half the total ERK1/2
expression of B6 ES cells. As well, 46C cells phosphorylate approximately half
the mount of ERK1/2 as B6 ES cells. The relative proportion of expression:
phosphorylation within each strain appears equal. Data are normalized to
tubulin. NT indicates mediumonly treated cells.
39
Figure 1.4c: STAT3 overexpression correlates to decreased ERK1/2
phosphorylation. Total cell lysates were compared via western blot and
probed for Akt and ERK1/2 phosphorylation following cytokine
stimulation for 30 minutes. STAT3 overexpressing B6 mouse ES cells
demonstrate a significant decrease in ERK1/2 phosphorylation as
compared to wildtype cells.
40
Figure 1.4d: Densiometry reveals that STAT3 overexpression correlates
to decreased ERK1/2 phosphorylation in B6 ES cells. Image analysis of
Figure 1.4c reveals that a fivefold increase in STAT3 expression
correlates to almost a 25 percent reduction in ERK1/2 phosphorylation
as compared to wildtype cells. Further analysis demonstrates that P
ERK1 is reduced by 75 percent as compared to wildtype cell ERK1
phosphorylation. Data are normalized to tubulin.
41
We then tested if decreasing ERK1/2 phosphorylation would enhance feeder‐free
self‐renewal. 5x10
3
cells/cm
2
B6 wild‐type and B6‐STAT3 mouse ES cells were
seeded upon gelatinized substrates. One group received LIF only while the other
received LIF plus the addition of PD0395201, a known specific MAPK inhibitor(Ying
et al., 2008a). Following seven days of incubation, AP staining revealed that, neither
LIF nor PD resulted in a significant proportion of AP positive colonies. However,
when LIF and PD were used in combination, a highly significant increase in AP
positive colonies resulted (Figure 1.4e). We tested whether STAT3 overexpression
or 46C mouse ES cells would respond to MAPK inhibition in a similar fashion. STAT3
overexpression resulted in increased AP positive colonies under LIF stimulation
alone, and PD also resulted in AP positive colonies. However, again, the combination
of LIF and PD resulted in a higher proportion of AP positive colonies than either
condition individually (Figure 1.4f). 46C feeder‐independent mouse ES cells resulted
in a significant increase in AP positive colonies when compared to B6‐STAT3 mouse
ES cells upon MAPK inhibition alone. As well, 46C mouse ES cells increase their AP
positive fraction when both LIF and PD are used in combination, mirroring the B6‐
STAT3 results (Figure 1.4f).
Quantitative PCR reveals that STAT3 overexpression increase RNA expression of the
core pluripotency gene Oct4, Nanog, and Sox2 as compared to wild‐type B6 mouse
ES cells (Figure 1.4g). Together, these data suggest that MAPK inhibition coupled to
42
LIF stimulation provides a synergistic effect that enhances feeder‐free mouse ES cell
self‐renewal.
Figure 1.4e: Inhibition of MAPK improves STAT3 mediated feeder
independent selfrenewal. 5x10
3
cells/cm
2
wildtype B6 mouse ES
cells were seeded upon gelatinized substrates and supplemented
with LIF, PD0395201, both, or medium alone (‘NT’). Following
seven days of incubation, the cells were fixed and AP stained. In the
case of LIF or PD, selfrenewal was impaired. However, when used
in combination, the selfrenewal was synergistically increased.
Asterisk indicates pvalue <0.001. Error bars indicate SD.
43
Figure 1.4f: MAPK inhibition increases feederfree selfrenewal when
STAT3 is overexpressed. Equal total cell amounts (5x10
3
cells/cm
2
) of
either B6Y118FSTAT3 or 46CY118F mouse ES cells were seeded upon
gelatinized substrates and incubated with either 10ng/ml LIF, 1uM
PD0395201, both or 2pg/ml GCSF for seven days. The cells were then
fixed and AP stained. Both cell types demonstrate increased AP positive
staining to either LIF or PD. 46C mouse ES cells significantly increase self
renewal with MAPK inhibition alone in serum medium. Both cell types
exhibit increased selfrenewal when both LIF and MAPK inhibition are
used in combination. Asterisks indicate pvalue <0.001. Error bars indicate
SD. ‘NT’ indicates mediumonly treatment.
44
Figure 1.4g: STAT3 overexpression increases RNA expression of core
pluripotency genes. Total RNA extracts from passage 15 B6STAT3
mouse ES cells maintained upon gelatinized substrates supplemented
with 10ng/ml LIF were converted to cDNA and subjected to
quantitative PCR analysis to determine expression levels of
pluripotency genes. Normalized to wildtype B6 mouse ES cells
maintained in coculture with feeders and supplemented with LIF,
B6STAT3 cells maintained feederfree demonstrate a robust
increase in core pluripotency gene expression. A modest 4fold
increase in STAT3 RNA expression increases Oct4, Nanog, and Sox2
expression levels over 20fold over wildtype cells, suggesting STAT3
may regulate these gene’s expression. Rex1, a marker of the
pluripotent inner cell mass appears unaffected, suggesting it is not
directly regulated by STAT3 overexpression. Data is normalized to
wildtype B6 mouse ES cells maintained with feeders, serum, and LIF.
Data are normalized to GAPDH.
45
We then tested if STAT3 expression was required in the context of ERK1/2
inhibition for enhancing feeder‐free self‐renewal. We employed the use of stat3/
mouse ES cells. Stat3/ mouse ES cells are not able to self‐renew via LIF/STAT3
activation, and can only be maintained as ES cells via chemical inhibition of both
GSK3β and MAPK(Ying et al., 2008a).
To test if STAT3 activation and MAPK inhibition is synergistically promoting mouse
ES cell self‐renewal, we stably transfected the same constitutively active STAT3α
vector into these STAT3 null ES cells. Upon drug selection and colony selection to
isolate individual clones, these stat3/ mouse ES cells overexpressing STAT3
(‘stat3/(STAT3)’) were seeded at clonal density (5x10
3
cells/cm
2
) into co‐culture
with CF‐1 feeders cells. Serum and LIF supplemented medium was applied and, after
three passages, immunoflourescence analysis revealed positive Oct4 and Sox2
expression, suggesting these cells are undifferentiated in the presence of the STAT3
transgene and LIF (Figure 1.4h). Cre‐mediated excision of the STAT3 transgene
resulted in GFP positive colonies. LIF treatment resulted in immediate
differentiation, suggesting that STAT3 expression is crucial for preventing
differentiation.
46
Figure 1.4h: STAT3 overexpression prevents differentiation in stat3/ mouse
ES cells cultured feederfree. Immunofluorescence images staining for both
Oct4 and Sox2 in STAT3 overexpressing stat3/ mouse ES cells cultured
feederfree upon a gelatinized substrate in serum medium supplemented with
10ng/ml LIF for three passages.
47
We then seeded the both stat3/ and stat3/(STAT3) mouse ES cells at clonal
density upon gelatinized substrates. Either LIF, PD0395201, or both were
supplemented into the culture medium. Seven days later, AP staining revealed that
STAT3 overexpressing ES cells exhibited increased AP positive staining after LIF
treatment. MAPK inhibition did not result in significant AP positive staining by itself
in either cell type, however the combination of LIF and PD0395201 promoted a
significantly higher proportion of AP positive colonies than either LIF or
PD03095201 separately. Lack of STAT3 expression correlated with no positive AP
staining could be recovered from LIF, PD0325901, nor the combination of the two
factors (Figure 1.4i). These results suggest STAT3 expression is required for feeder‐
independent self‐renewal, and MAPK inhibition alone is insufficient to promote
mouse ES cell self‐renewal.
48
Figure 1.4i: STAT3 expression is required for LIFmediated selfrenewal
feederfree. (A) Stat3/ and Stat3/(STAT3) mouse ES cells were seeded at
equal density (5x10
3
cells/cm
2
) upon gelatinized substrates. After various
cytokine treatments for a seven day period, the cells were fixed and AP
stained. As indicated, there is no difference between mediumonly (‘NT’)
treated and LIF treated cells when STAT3 is not expressed. However, upon
reintroduction of STAT3 via a high expression vector, selfrenewal is
regained. LIF mediated selfrenewal is enhanced by addition of MAPK
inhibitor PD0395201, but neither LIF nor MAPK inhibition alone promotes
selfrenewal. GCSF treatment recapitulates LIF/PD selfrenewal as well. (B)
AP stain of entire well demonstrates STAT3 expression’s enhances stat3/
ES cell selfrenewal. Error bars indicate SD.
49
1.5 βCatenin Activation Enhances Mouse ES Cell SelfRenewal in LIFDependent
Fashion
β‐catenin likely plays an important role in ES cell self‐renewal. However, most
reports only examine β‐catenin’s role in isolation(Miyabayashi et al., 2007; Sato et
al., 2004). To determine if β‐catenin can enhance STAT3‐mediated self‐renewal, We
hypothesized that increasing nuclear β‐catenin levels would augment STAT3’s
effects. β‐catenin is canonically activated via the inhibition of GSK3β via the Wnt
signal pathway(Miller et al., 1999). Since GSK3β inhibition promotes both mouse
and rat ES cell self‐renewal in the absence of STAT3 signaling, we sought to
investigate whether it could augment self‐renewal if STAT3 activation was present.
To test this, we employed a mouse ES cell line that contained a floxed, stably
expressed β‐catenin‐Estrogen receptor fusion protein which, in the presence of the
chemical 4‐hydroxy tamoxifen (4‐OHT), will cause the nuclear importation of β‐
catenin. This construct was transfected into the B6‐Y118F cell line to take advantage
of G‐CSF signaling. We then seeded an equal number (10
3
cells/cm
2
) ES cells at
clonal density and stimulated this with various permutations of LIF, 4‐OHT, or
PD184352, a known MAPK inhibitor.
50
When LIF was used in combination with 4‐OHT, a significant increase in AP positive
colonies emerged. In the absence of LIF, the combined effects of 4‐OHT and PD18
did not result in a significant proportion of AP positive colonies (Figure 1.5a).
We observed that despite increasing the proportion of AP positive colonies via the
combined effects of STAT3 activation and MAPK inhibition, addition of 4‐OHT to this
condition did not significantly increase the number of AP positive colonies as
compared to omitting it (Figure 1.5b).
Figure 1.5a: βCatenin activation does not significantly enhance feeder
free B6 selfrenewal in the absence of LIF. B6T2ER ES cells were
subjected chemical inhibition of MAPK or activation of βcatenin. AP
positive colonies were quantified at Day 7. βcatenin and MAPK
inhibition alone did not result in significant AP positive colony
formation. The combination of LIF and 4OHT increased AP positive
colony formation only when PD18 was present. Asterisk indicates pvalue
<0.05. Error bars indicates SD.
51
To demonstrate that the increased self‐renewal observed was due to β‐catenin
activation via transgene activation, the transgene was excised using Cre
recombinase. Clones were again selected, tested for transgene excision, and then
subjected to the previous assay. Figure 1.5c demonstrates that removal of the
transgene correlates to a loss of AP positive colonies despite LIF and 4‐OHT
stimulation. These results suggest that β‐catenin may augment LIF‐mediated self‐
renewal, but that this effect is not critical in promoting feeder‐free self‐renewal in
B6 mouse ES cells.
Figure 1.5b: βcatenin Activation Does Not Significantly Increase SelfRenewal
when despite LIF stimulation and MAPK inhibition. When LIF plus MAPK
inhibition was implemented into culture with 4OHT, no significant increase was
observed in selfrenewal.. Error bars indicate SD.
52
Figure 1.5c: Feederfree selfrenewal ability is impaired following β
catenin transgene excision. 10
3
cells/cm
2
of B6 mouse ES cells expressing
the T2ER transgene were seeded upon a gelatinized substrate. B6T2ER
mouse ES cells transfected with Cre recombinase were seeded as well.
10ng/ml LIF, was added and the cell incubated for seven days. The cells
were then fixed and AP stained. Cells treated with LIF and 4OHT
demonstrate selfrenewal feederfree only when the T2ER gene is present.
Cre transfected cells are unable to selfrenew despite LIF and 4OHT
stimulation. Asterisk indicates pvalue <0.05. Error bars indicate SD.
53
Chapter Two Increased Endogenous STAT3 Activation is Sufficient to Maintain
FeederIndependent Mouse ES Cell SelfRenewal
2.1 – Artificial Activation of Endogenous STAT3 Can Functionally Replace LIF in Mouse
ES Cells
Since STAT3 overexpression is not physiologically relevant, we then tested if
endogenous STAT3 activation was sufficient to enhance feeder‐independent mouse
ES cell self‐renewal. We have observed previously that STAT3 overexpression leads
to significantly increased Tyr705 phosphorylation levels as compared to wild‐type
cells despite using an equivalent concentration of LIF. We sought to mimic this
increased STAT3 activation by stable integration and expression of a chimeric
receptor that has been shown to control STAT3 activation in a dose‐dependent
fashion.
The receptor is constructed as the N‐terminal extracellular domain of the
granulocyte‐colony stimulating factor (G‐CSF) receptor fused to the cytosolic
domain of the common IL‐6 receptor gp130(Niwa et al., 1998b). Gp130 has been
shown to be the key mediator in transducing the LIF signal to STAT3, MAPK, and
PI(3)K in mouse ES cells. In addition, this chimeric receptor contains a loss‐of‐
function mutation at Tyr118 of gp130 (as calculated from the fusion boundary
between the GCSF receptor and gp130) that results in the inability to recruit the
54
phosphatase SHP‐2. SHP‐2 has been demonstrated to be the key mediator in
activating the MAPK/ERK and PI(3)K/AKT pathway in mouse ES cells.
The G‐CSF receptor is not expressed in wild‐type mouse ES cells, so we
hypothesized that addition of G‐CSF to the cell culture would activate the chimeric
receptor, which would in turn activate STAT3(Burdon et al., 1999).
We then stably transfected and expressed the chimeric gp130 receptor in wild‐type
B6 mouse ES cells. Following drug and colony selection, individual clones were
tested for their ability to respond to G‐CSF. G‐CSF was applied to transfected clones
seeded at clonal density upon gelatinized substrates in decreasing two‐fold
dilutions, beginning with 10ng/ml. AP staining revealed that, after seven days
incubation, G‐CSF treated cells exhibited a dose‐dependent response in positive
correlation to the concentration applied. It was found that nominal concentrations,
~0.2ng/ml, were sufficient to promote feeder‐independent self‐renewal as
indicated by AP staining (Figure 2.1a). Densiometric analysis revealed that relative
STAT3 phosphorylation levels decreased by approximately one‐half for every ten‐
fold decrease in G‐CSF for the concentrations tested (Figure 2.1b). Below 0.2pg/ml
G‐CSF treatment could not prevent crisis.
55
Figure 2.1a: GCSF mediated STAT3 activation exerts a dosedependent self
renewal effect. (A) 10
3
cells/cm
2
B6Y118F mouse ES cells were seeded upon
a gelatinized substrate in serum medium and supplemented with a
decreasing concentration of GCSF, 10ng/ml LIF, or medium alone.
Following a seven day incubation, the cells were fixed and AP stained. AP
positive staining shows a progressive loss as the GCSF concentration
decreases. LIF appears to be insufficient to promote feederindependent self
renewal even at high concentration. (B) Western blot comparing the effects
of decreasing concentrations of GCSF upon STAT3 phosphorylation. As
indicated, phosphorylated Tyr705 levels decrease in concordance with
decreasing GCSF concentrations.
56
To identify if G‐CSF was exerting the self‐renewal effect through the Jak/STAT3
pathway, we employed the use of a well‐characterized Jak2 inhibitor, AG490. Again,
B6‐Y118F mouse ES cells were plated upon gelatinized substrates at clonal density
and supplemented with 0.2ng/ml G‐CSF, with one well receiving an additional
supplement of 250nM AG490(Eriksen et al., 2001). AP staining reveals that while G‐
CSF by itself is enough to prevent mouse ES cell crisis, the addition of AG490
significantly reduced the amount of self‐renewing colonies (Figure 2.1c). This result
suggests that G‐CSF stimulation of the chimeric receptor is mediating self‐renewal
through a Jak/STAT3 signal pathway.
Figure 2.1b: Densiometry reveals that STAT3 phosphorylation levels decrease in
correlation with decreasing GCSF concentrations. Image analysis of Figure 2.1a
indicate that as GCSF concentration decreases, phosphorylated STAT3
decreases as well. Data are normalized to tubulin expression.
57
Figure 2.1c: Jak inhibition decreases STAT3 mediated mouse ES cell self
renewal efficiency feederfree. 5x10
3
cells/cm
2
B6Y118F cells were
seeded upon a gelatinized substrate and incubated with either 10ng/ml
LIF, 0.2ng/ml GCSF or serum medium alone. One group was
supplemented further with 250 nM Jak inhibitor AG490 and the other
DMSO control. Following a seven day incubation, the cells were fixed and
AP stained. LIF treated cells did selfrenew appreciably with or without
Jak inhibition, however GCSF treated cell selfrenewal were significantly
inhibited by Jak inhibition, suggesting GCSF is exerting its effects
through STAT3. Asterisk indicates pvalue <0.01. Error bars indicate SD.
58
2.2 – Increased Endogenous STAT3 Activation Prevents Crisis in Other Feeder
Dependent Mouse ES Cell Lines
We then tested if endogenous STAT3 activation prevents crisis effect was limited to
the B6 mouse ES cell line. I selected another feeder‐dependent mouse ES cell line,
Balb/C1. I then stably expressed the same chimeric G‐CSF/gp130 receptor, drug
selected the transfectants and isolated individual clones. Balb/C1 mouse ES cells
containing the chimeric receptor (termed ‘Balb/C‐Y118F’) demonstrated AP
positive colonies in the presence of G‐CSF. Balb/C ES cells were not able to self‐
renew effectively in the presence of LIF when seeded at clonal density upon a
gelatinized substrate as indicated by alkaline phosphatase staining (Figure 2.2a).
Our results suggest that the rescue from ES cell crisis is likely not an anomalous
phenomenon relegated to a specific mouse ES cell strain, but rather a general
phenomenon .
59
Figure 2.2b: STAT3 Activation promotes feederindependent selfrenewal of BalbC1
mouse ES cells. (A) 5x10
3
cells/cm
2
of either B6Y118F or Balb/c1Y118F mouse ES
cells were seeded onto a gelatinized substrate in serum medium containing either
10ng/ml LIF, 10ng/ml GCSF, 1ng/ml GCSF, or medium alone. Balb/c mouse ES
cells mirror B6 mouse ES cells in that they exhibit morphology consistent with self
renewal in the presence of GCSF but not LIF. (B) AP staining of Balb/cY118F
mouse ES cells after seven days of culture feederfree in either LIF, GCSF, or
medium alone. Strong AP positive staining of GCSF treated cells suggest that
feederfree selfrenewal is enhanced by GCSF treatment Balb/c1 ES cells.
60
In addition, I assayed a feeder‐independent mouse ES cell line to ascertain whether
G‐CSF mediated STAT3 activation could substitute for LIF in this capacity. Using 46C
mouse ES cells transfected with the chimeric receptor (‘46C‐Y118F’), I added a
gradient of concentrations of G‐CSF with a range of 200pg/ml – 0.78pg/ml, a range
of three orders of magnitude. As seen in Figure 2.2c, G‐CSF treatment results in AP
positive colonies. Peripheral differentiation, as suggested by loss of AP positive
staining, increases as G‐CSF concentration falls. In addition we observe a general
flattening of the individual colonies and gradual loss of AP staining. Previously we
demonstrated that G‐CSF activates STAT3, and that STAT3 activation mediates
mouse ES cell feeder‐free self‐renewal. It can therefore be inferred that STAT3
mediates feeder‐free self‐renewal irrespective of mouse ES cell strain.
61
Figure 2.2c: Feederindependent 46C mouse ES cells exhibit a dosedependent
selfrenewal response to GCSF treatment. 10
3
cells/cm
2
46CY118F mouse ES
cells were seeded upon gelatinized substrates and supplemented with
decreasing concentrations of GCSF, 10ng/ml LIF, or medium alone (‘NT’).
Following seven days of incubation, the cells were fixed and AP stained. GCSF
appear to exert a similar effect as B6 mouse ES cells in response to GCSF
mediated STAT3 activation. AP positive staining decreases with decreasing G
CSF concentration. LIF stimulation produces AP positive colonies, but overt
marginal differentiation is apparent.
62
Chapter Three Increased and Sustained STAT3 Activation Promotes Mouse and
Rat ES Cell SelfRenewal
3.1 GCSF Stimulation Promotes Increased STAT3 Activation Over 24 Hours
LIF/STAT3 signaling reaches a peak activation level approximately thirty minutes
after induction(Niwa, 2001; Niwa et al., 1998a). At which point, negative feedback
loops such as SOCS3 are upregulated by active STAT3 which attenuate STAT3
signaling by binding Jak2 and preventing it from phosphorylating STAT3(Li et al.,
2005; Sasaki et al., 1999). We sought to compare G‐CSF stimulation to LIF induction
to investigate if G‐CSF reached a similar peak of activation. Following stimulation of
B6‐Y118F mouse ES cells upon gelatinized substrates, total protein lysates were
collected at thirty minutes, sixteen hours, and twenty‐four hours post‐induction
with both cytokines. Western blotting revealed that G‐CSF activated an increased
amount of STAT3 at thirty minutes as compared to LIF, but also this activation level
is sustained over twenty‐four hours (Figure 3.1a). Densiometric analysis indicates
that G‐CSF treatment increased STAT3 phosphorylation levels approximately two‐
fold as compared to LIF. This increase degraded slightly over time, but remained at
least two‐fold higher than LIF treated cells (Figure 3.1b). LIF, on the other hand,
shows a return to basal activation levels by sixteen hours, with a maximum at thirty
minutes. STAT3 expression levels remain relatively unchanged during either
63
treatment (Figure 3.1c). These data suggest that increasing STAT3 activation over
time may enhance mouse ES cell feeder‐free self‐renewal.
Figure 3.1a: GCSF mediated STAT3 phosphorylation is increased and
sustained over 24 hours. Total cell lysates extracted from B6Y118F mouse
ES cells cultured feederfree were compared for Tyr705 phosphorylation.
Cells stimulated with 10ng/ml LIF demonstrate peak phosphorylation at 30
minutes poststimulation with a return to basal levels by 16 hours. GCSF
treated cells demonstrate increased phosphorylation at 30 minutes and
relatively stable maintenance of this phosphorylation over 24 hours. Both
cell types appear to modestly increase STAT3 expression levels over time as
well.
64
Figure 3.1b: Densiometry reveals that GCSF treated mouse ES cells retain
an increased STAT3 phosphorylation level over 24 hours. Image analysis of
Figure 3.1a indicates that GCSF stimulation of B6 mouse ES cells increases
STAT3 phosphorylation levels with minimal signal degradation over 24
hours as compared to LIF treated cells. Data are normalized to tubulin
expression.
Figure 3.1c: Densiometry reveals that GCSF treated mouse ES cells retain a
stable STAT3 expression level over 24 hours. Image analysis of Figure 3.1a
indicates that GCSF stimulation of B6 mouse ES cells causes both LIF
treated and GCSF treated cells to retain relatively similar STAT3 expression
levels over 24 hours. Data are normalized to tubulin expression.
65
To compare STAT3 activation level‐profiles between cell lines, we then expressed
the chimeric gp130 receptor in STAT3 overexpressing B6 mouse ES cell lines using
the methods previously outlined, a line now termed ‘B6‐Y118F‐STAT3’. We then
transfected Cre recombinase into the B6‐Y118F‐STAT3 cell line to remove the
STAT3 transgene. Following selection of colonies, we expanded both cell lines to
compare their ability to activate STAT3 in response to G‐CSF. After stimulating each
cell type with either 10ng/ml LIF, 0.2ng/ml G‐CSF or serum medium alone, we
compared each cell line via western blot. When compared to both wild‐type cells or
those only expressing the chimeric receptor alone (B6‐Y118F), B6‐Y118F‐STAT3
mouse ES cells demonstrate a significant increase in Tyr705 phosphorylation in
response to both LIF and G‐CSF. Following removal of the STAT3 transgene, STAT3
expression and activation levels return to wild‐type levels, suggesting that
increasing STAT3 expression allows for increased STAT3 phosphorylation in
response to LIF or G‐CSF (Figure 3.1d).
66
Figure 3.1d: GCSF activates STAT3 in proportion to STAT3 expression level.
Equal total cellular protein from B6 mouse ES cells was compared via
western blot analysis. Wildtype B6 mouse ES cells respond to 10ng/ml LIF
but not GCSF, while all remaining cell lines stably expressing the chimeric
gp130 receptor activate STAT3 in response to 0.2ng/ml GCSF treatment.
STAT3 overexpression results in a significant increase in STAT3 Tyr705
phosphorylation by both 10ng/ml LIF and 0.2ng/ml GCSF. Cremediated
excision of the STAT3 transgene results in a return to wildtype STAT3
expression levels.
67
3.2 – GCSF Stimulation Correlates to Increased Socs3 RNA Expression
We then investigated whether G‐CSF treatment targets similar LIF/STAT3 gene
targets. We chose Socs3 as it is well characterized primary target of STAT3 signaling
in mouse ES cells(Duval et al., 2000; Heinrich et al., 2003; Li et al., 2005). We
decided to ascertain the kinetics of RNA expression in G‐CSF treated mouse ES cells
over time.
Quantitative PCR revealed that G‐CSF treated mouse ES cells increase Socs3 RNA
expression rapidly by one hour and this RNA level decreased after 24 hours of
stimulation (Figure 3.2a). LIF treatment demonstrated a similar expression pattern
in Socs3 RNA expression level as well. Indeed this finding is corroborated by
microarray data that indicates that Socs3 is upregulated in response to G‐CSF and
LIF treatment (Section 3.3). These data suggest G‐CSF treatment may be targeting
similar genes as LIF, suggesting that G‐CSF regulates mouse ES cell self‐renewal by
targeting a similar gene set as LIF, yet possibly regulating this set in a distinct
manner from LIF.
68
Figure 3.2a: SOCS3 RNA expression levels do not appear to
significantly differ between LIF or GCSF treatments. Total RNA
extracts from B6Y118F mouse ES cells stimulated with either
0.2ng/ml GCSF, 10n/ml LIF, or medium alone over 24 hours.
Both GCSF and LIF treated cells exhibit similar expression
patterns over time. Data normalized to mediumonly treatment.
Error bars indicate SEM.
69
3.3 – STAT3 Activation Induces Differentiation When STAT3 is Overexpressed
The duration of the STAT3 activation signal appears to regulate mouse and rat ES
cell self‐renewal feeder‐free. We sought to understand if STAT3 expression has a
functional ceiling. A functional ceiling is defined as increasing either STAT3
expression or activation levels so that the protein no longer increases self‐renewal
ability or possibly begins to harbor a deleterious effect upon self‐renewal.
We then applied 0.2ng/ml G‐CSF, the ideal concentration determined for B6 mouse
ES cells with wild‐type STAT3 expression levels, to B6‐Y118F‐STAT3 mouse ES cells.
Two days later, we observed B6‐Y118F‐STAT3 mouse ES cells were no longer
arranged in colonies, but rather as individual flattened cells that could not be
passaged. RT‐PCR analysis suggests these cells are predominantly primitive
endoderm following G‐CSF treatment (Figure 3.3a). This result suggests that
increasing STAT3 activation in the context of overexpression may induce ES cell
differentiation.
70
Figure 3.3a: GCSF mediated STAT3 activation correlates to rapid
induction of mouse ES cell differentiation. (A) B6Y118FSTAT3 mouse
ES cells maintained in coculture with CF1 feeders and serum medium
supplemented with 10ng/ml LIF. (B) B6Y118FSTAT3 mouse ES cells
passaged into feederfree conditions and supplemented with 0.2ng/ml G
CSF immediately differentiate within 3 days of stimulation. (C) 5x10
3
cells/cm
2
B6Y118FSTAT3 mouse ES cells were seeded upon a
gelatinized substrate in serum medium supplemented with 0.2ng/ml.
Reversetranscription PCR analysis reveals that GCSF mediated
differentiation results in Sparc positive cell types.
71
3.4 Non129 Mouse ES Cells Maintained FeederFree Exhibit Robust In Vitro
Pluripotency
To verify that feeder‐free self‐renewal does not impinge upon pluripotency, B6‐
Y118F mouse ES cells were seeded at clonal density upon a gelatinized substrate.
Serum‐containing medium was supplemented with 0.2ng/ml G‐CSF and the cells
were allowed to proliferate. Following fifteen passages in this condition, the cells
were subjected to in vitro pluripotency assays to verify ES identity:
Embryoid body (EB) formation assays were performed by seeding 5x10
6
B6‐Y118F
mouse ES cells onto a uncoated, bacterial grade petri dish in serum medium.
Medium was changed every other day as the ES cells aggregate and form embryoid
bodies that proceed with differentiation. Following maturation by approximately
Day 12‐14, roughly four or five mature EB’s were seeded upon a gelatinized four‐
well substrate. The cells were allowed to continue differentiating until
spontaneously beating areas were observed by approximately Day 7, post‐plating.
At this point the cells were stained for markers of differentiation.
As indicated in Figure 3.4a, B6‐Y118F mouse ES cells cultured feeder‐free for fifteen
passages demonstrate the ability to form tissue types of the three somatic germ
layers. This finding was confirmed by RT‐PCR and quantitative PCR analysis of both
conventionally LIF/feeder maintained B6 mouse ES cells and the G‐CSF maintained
versions.
72
In addition, we interrogated the B6‐Y118F mouse ES cells for their global RNA
expression patterns to determine if G‐CSF treatment significantly overlaps with LIF
treatment. Since G‐CSF can support feeder‐independent self‐renewal and LIF
cannot, it can be hypothesized that the gene targets may be different for the two
conditions. We subjected B6‐Y118F mouse ES cells to stimulation with either
0.2ng/ml G‐CSF, 10ng/ml LIF, or medium alone for 45 minutes. The total RNA was
analyzed and globally compared. G‐CSF increases expression of a small subset of
genes and downregulates the remainder, as compared to medium‐treated only cells.
LIF treatment presents a gene expression pattern similar to medium‐only treated
cells. This results suggests that LIF’s gene targets are consistent with a
differentiating phenotype when feeder‐dependent mouse ES cells are placed into a
feeder‐free environment (Figure 3.4b). Upon further examination of the
LIF/Jak/STAT3 pathway, G‐CSF treatment reveals that it significantly
downregulates SHP‐2, Ras, and Akt pathway RNA. This result confirms that G‐CSF
mediated STAT3 activation is likely the critical effector of feeder‐independent self‐
renewal, and that inhibition of MAPK likely enhances this effect.
73
Figure 3.4a: Feederfree maintenance of B6 mouse ES cells in GCSF
conditions maintains pluripotent identity. (A) Passage 15 B6Y118F mouse
ES cells maintained in 0.2ng/ml GCSF upon gelatinized substrates in the
absence of feeders were subjected to embryoid body formation. The resulting
differentiated cell types were probed for markers of individual germ layers.
The cells demonstrate positive staining for all three germ layers βIII
tubulin (ectoderm), gata4 (endoderm), and muscle sarcomere MF
20(mesoderm) – suggesting these cells are likely pluripotent. (B) RTPCR
analysis further suggests no significant difference in differentiation ability
exist between feederfree and feederdependent populations.
74
Figure 3.4b: GCSF treatment of mouse ES cells regulates a distinct set of
genes as compared to LIF. (A) Microarray analysis of B6Y118F mouse ES
cells maintained feederfree in either 0.2ng/ml GCSF, 10ng/ml LIF, or
medium alone (‘NT’) reveals that GCSF gene patterning is significantly
distinct from LIF. GCSF upregulated only a small set of genes, while
generally downregulating the remainder as compared to LIF or medium
alone. (B) Specific genes of the LIF/Jak/STAT3 pathway were analyzed in
detail and reveal that GCSF downregulates MAPK and Akt family member
genes, while LIF appears to increase expression of these genes. (C) Table
comparing the total significantly up or downregulated genes for direct
treatment comparisons. Heatmap colors indicate relative foldchange in
expression. Red equals increased expression, while green indicates decreased
expression, and black indicates no change.
75
We interrogated the G‐CSF maintained B6‐Y118F ES cells for pluripotency markers.
Immunofluorescence and western blotting reveal positive staining for Oct4, Nanog,
and Sox2 at passage fifteen. Quantitative PCR was performed to directly compare
pluripotency gene RNA expression levels of G‐CSF B6‐Y118F mouse ES cells
maintained feeder‐free to wild‐type B6 mouse ES cells maintained upon feeders and
supplemented with LIF. qPCR results revealed similar expression patterns between
both cell types (Figure 3.4c). This result indicates that G‐CSF/STAT3 maintained
cells are likely pluripotent populations of ES cells as compared to their feeder‐
maintained counterparts.
76
Figure 3.5c: B6 mouse ES cells maintain pluripotency following long‐term culture
feeder‐free.
77
Figure 3.5c continued
78
Figure 3.5c continued
Figure 3.4c: B6 mouse ES cells maintain pluripotency following longterm
culture feederfree. (A) B6Y118F mouse ES cells were maintained upon
gelatinized substrates in serum medium containing 0.2ng/ml GCSF and
passaged at regular intervals. (B) RTPCR analysis of passage 15 GCSF
maintained B6 mouse ES cells. Feederfree maintenance does not appear to
adversely affect core pluripotency gene expression as compared to wildtype
B6 mouse ES cells maintained with feeders and LIF. (C) Western blot
demonstrating protein expression of Oct4 and Nanog is comparable between
both feederfree and canonical culture conditions. (D,E)
Immunofluorescence demonstrating feederfree maintained mouse ES cells
express Oct4 and Sox2. (F) Quantitative PCR analysis of B6Y118F cells
maintained in 0.2ng/ml GCSF feederfree as compared to cells in 10ng/ml
LIF with feeders or medium alone. Feederfree maintenance results in a
comparable gene expression pattern over 24 hrs as compared to LIF and
feeders. FGF5, a marker of early differentiation is also downregulated in
response to GCSF treatment. Error bars indicate SEM.
79
3.5 Sustaining Rat ES Cell SelfRenewal Through Manipulating STAT3 Signaling
Our group has reported the derivation and characterization of rat embryonic stem
cells(Li et al., 2008). This pluripotent cell type relies upon derivation and
maintenance upon mouse embryonic fibroblasts, serum‐free defined medium, and
the combined inhibition of both GSK3β and MAPK. In direct contrast with their
murine counterparts, rat ES cells rely upon shielding from inductive differentiation
cues in a STAT3‐independent manner. This cell state has been described as the
‘ground state’ of embryonic stem cells, and has been shown to be applicable in the
derivation and maintenance of mouse ES cells as well.
However, we observed in our work that rat ES cells, despite their ability to self‐
renew in a STAT3 independent manner, demonstrated an acute sensitivity to LIF
stimulation. Upon addition of LIF to the culture medium, DAc2 rat ES cells robustly
phosphorylate Tyr705 upon STAT3 in a manner consistent with mouse ES cells. In
addition rat ES cells phosphorylate Ser727 in concordance with mouse ES cells
(Figure 3.5a). Rat ES cells express slightly less STAT3 than mouse ES cells, yet
activate a comparable fraction of when stimulated with LIF (Figure 3.5b).
80
Figure 3.5a: DAc8 rat ES cells activate STAT3 in response to LIF
stimulation. Total protein lystates were directly compared via
western blot to ascertain if LIF causes phosphorylation of STAT3. As
compared to mouse ES cells, rat ES cell do appear to phosphorylate
Tyr705 robustly and demonstrate comparable STAT3 expression
levels to mouse.
Figure 3.5b: Densiometry comparing rat and mouse ES cell STAT3 levels.
Image analysis of Figure 3.5a demonstrates that rat ES cells express
slightly less STAT3, yet activate slightly more as compared to mouse ES
cells. Cells were stimulated with LIF for 30 minutes.
81
Previously, we have established that sustained STAT3 activation can promote
robust feeder‐independent mouse ES cell self‐renewal in non‐129 mouse strains.
We hypothesized that rat ES cells may respond similarly to STAT3 activation as long
as STAT3 is regulated appropriately.
It was demonstrated that rat ES cells overexpressing STAT3 are able to self‐renew
effectively in the presence of LIF and feeders. The STAT3 transgene was stably
expressed in DAc2 rat ES cells. Western blotting reveals that STAT3 expression is
increased and responsive to LIF stimulation. STAT3 overexpressing rat ES cells
remain Oct4 and Sox2 positive in long‐term culture maintained in LIF and serum
supplemented medium (Figure 3.5c). This result suggests that rat ES cells are
sensitive to STAT3 expression and activation levels, akin to mouse ES cells. We then
hypothesized that specific activation and regulation of endogenous STAT3 would
recapitulate the positive effects observed upon self‐renewal observed in the mouse.
To test this hypothesis, we began by stably introducing the chimeric gp130 receptor
into DAc8 rat ES cells. Following drug selection and clone isolation, we tested
individual clones for their ability to self‐renew feeder‐free. Under no condition
(laminin, fibronectin, or matrigel‐coated substrates) could we identify a G‐CSF
concentration that would promote feeder‐independent self‐renewal. This is
unsurprising, as extracellular matrix (ECM) components that have been
demonstrated to promote either mouse or human ES cell self‐renewal in lieu of
82
feeder cells are well‐documented, but in rat ES cell culture, no such condition
faithfully reproduces the effects of feeders.
Figure 3.5c: STAT3 overexpression promotes LIF mediated selfrenewal of rat ES
cells. (A) Total protein lysates were compared via western blot following stable
overexpression of STAT3 in DAc2 rat ES cells. As is demonstrated, Tyr705
phosphorylation is increased in response to LIF following STAT3 overexpression.
Following gene excision via Cre recombinase, STAT3 phosphorylation levels
return to wildtype levels. (B) STAT3 overexpression results in LIF mediated self
renewal of rat ES cells longterm, as indicated by Oct4 and Sox2 expression. (C)
Cremediated excision of the transgene results in the loss of exogenous STAT3
expression and loss of Oct4 and Sox2 expression despite LIF stimulation.
83
We therefore performed our assays upon feeder cell layers. Previous research has
revealed that only the combined use of specific GSK3β and MAPK inhibition (‘2i’)
prevents rat ES cell differentiation. As a point of comparison, we employed both LIF
and 2i treated wild‐type DAc8 rat ES cells to compare self‐renewal efficiency. To test
if G‐CSF could prevent rat ES cell differentiation, we seeded DAc8 rat ES cells
expressing the chimeric gp130 receptor (termed ‘DAc8‐Y118F’) at clonal density
upon a gelatinized substrate pre‐coated with CF‐1 feeder cells. We determined that
0.02ng/ml G‐CSF was optimal for promoting rat ES cell self‐renewal. As seen in
Figure 3.5d, both 2i and 0.02ng/ml G‐CSF promoted rat ES cell self‐renewal, yet
10ng/ml LIF stimulation results in rapid differentiation.
Figure 3.5d: GCSF supports rat ES cell selfrenewal. (A) Phase contrast
photo displaying DAc8 rat ES cells maintained in the canonical 2i culture
system in serumfree N2B27 medium. (B) 10ng/ml LIF application to DAc8
rat ES cells results in differentiation within two passages. (C) 0.02ng/ml G
CSF application to DAc8Y118F rat ES cells are able to selfrenew efficiently
over fifteen passages with no overt differentiation. (D) GCSF maintained
DAc8Y118F rat ES cell AP stain at passage 15.
84
We then seeded equal total Dac8‐Y118F rat ES cells into co‐culture with CF‐1
feeders and stimulated them with either LIF, G‐CSF, 2i, or medium alone. G‐CSF
treated cells demonstrate significantly increased AP positive colonies as compared
to either 2i or LIF. We observed no AP positive colonies in the LIF‐treated condition
(Figure 3.5e). Thus, we hypothesized that the chimeric receptor may be functioning
as in mouse ES cells to promote self‐renewal.
To verify the chimeric receptor’s function, I seeded both wild‐type DAc8 and DA‐
Y118F rat ES cells at clonal density, approximately 5x10
3
cells/cm
2
, upon a layer of
feeder cells. Both cell types were stimulated with either 10ng/ml LIF or 0.02ng/ml
Figure 3.5e: GCSF mediated STAT3 activation increases rat ES cell self
renewal ability over 2i. 5x10
3
cells/cm
2
DAc8Y118F rat ES cells were seeded
in coculture with CF1 feeders in serumfree N2B27 medium. Either 10ng/ml
LIF, 3uM CHIR99201 plus 1uM PD0395201 (‘2i’), or 0.02ng/ml GCSF were
added and allowed to incubate for seven days. The cells were then fixed and
AP stained. GCSF significantly promotes selfrenewing rat ES cell colonies as
compared to 2i or no treatment. LIFtreatment reveals almost no self
renewing colonies. Asterisk indicates pvalue <0.05. Error bars indicate SD.
85
G‐CSF and total cell lysates were compared for protein expression. Western blotting
revealed that both LIF and G‐CSF demonstrate robust Tyr705 phosphorylation in
DA‐Y118F rat ES cells (Figure 3.5f). Densiometric analysis confirms that DAc8‐
Y118F rat ES cells phosphorylate STAT3 comparably to wild‐type cells when either
LIF or G‐CSF is employed (Figure 3.5g). Wild‐type cells did not demonstrate
detectable STAT3 phosphorylation in response to G‐CSF stimulation. This result
suggests that the chimeric receptor functions to activate STAT3 through G‐CSF
solely in rat ES cells.
Figure 3.5f: Rat ES cells expressing the chimerical gp130 receptor
activate STAT3 in response to GCSF. Total protein lysates were collected
from either wildtype or rat ES cells expressing the chimeric gp130
receptor following a 30 minute stimulation with either 10ng/ml LIF or
0.02ng/ml GCSF. Only cells that express the chimeric receptor
phosphorylate Tyr705 in response to GCSF stimulation.
86
We then hypothesized that, like mouse ES cells, STAT3 activation likely exerts its
effects in a temporal fashion. To test this, we stimulated DAc8‐Y118F rat ES cells
with either LIF or G‐CSF and collected cell lysates at thirty minutes, sixteen hours,
and 24 hours post‐induction. As indicated in Figure 3.5h, G‐CSF treated rat ES cells
demonstrate increased STAT3 Tyr705 phosphorylation that is sustained over
twenty‐four hours. LIF‐treated cells reveal maximal Tyr705 phosphorylation at
thirty minutes with a return to basal levels by sixteen hours. Densitometry confirms
Figure 3.5g: Densiometry indicates that GCSF and LIF phosphorylate
STAT3 comparably in rat ES cells. Image analysis of Figure 3.5f suggests
that GCSF and LIF stimulation result in STAT3 phosphorylation. Data
are normalized to tubulin expression.
87
this finding. G‐CSF stimulation maintains almost two‐fold increase in STAT3
activation as compared to LIF over 24 hours (Figure 3.5i‐j). Quantitative PCR
analysis performed upon DA‐Y118F cells maintained in 0.02ng/ml G‐CSF for fifteen
passages suggests that these cells are undifferentiated (Figure 3.5k). Together, these
results suggest that, like feeder‐dependent mouse ES cells, increased and sustained
STAT3 activation promotes rat ES self‐renewal.
Figure 3.5h: GCSF stimulation sustains STAT3 activation over time
promoting longterm selfrenewal. Total protein lysates were compared
via western blot. (A) DAc8Y118F rat ES cells were stimulated with
either 0.02ng/ml GCSF to 10ng/ml LIF for 30 minutes. Analysis reveals
that GCSF induces Tyr705 phosphorylation robustly and sustains the
majority of this activity over 24 hours. LIF rapidly phosphorylates and
returns to basal levels by 16 hours. (B) AP stain demonstrating
undifferentiated cells after fifteen passages stimulated with GCSF (C)
identical cells maintained in 2i after fifteen passages.
88
Figure 3.5i: Densiometry indicates that GCSF increases STAT3 activation
over 24 hours in rat ES cells. Image analysis of Figure 3.5h(A) indicates
that GCSF stimulation increases STAT3 phosphorylation almost twofold
over LIF treated rat ES cells. Both signals degraded over time, yet GCSF
maintains twice as much active STAT3 as LIF at any given moment over
24 hours. Data are normalized to tubulin expression.
Figure 3.5j: Densiometry indicates that GCSF maintains STAT3
expression over 24 hours in rat ES cells. Image analysis of Figure 3.5h(A)
indicates that GCSF and LIF stimulation maintain constant STAT3
expression over 24 hours. Data are normalized to tubulin expression.
89
Figure 3.5k: GCSF maintained rat ES cells maintain a pluripotent identity
similar to canonical 2i maintained rat ES cells. Quantitative PCR analysis of
DAc8Y118F rat ES cells stimulated with either 0.02ng/ml GCSF, 10ng/ml
LIF, 2i (3uM CHIR99201, 1uM PD0395201), or serumfree N2B27 medium
alone (‘NT’) reveals that GCSF treated cells mirror 2i treated cells in their
pluripotency gene expression pattern. LIF treated cells, although expressing
pluripotencyrelated RNA, demonstrates a more aberrant pattern similar to
mediumonly treatedcells, suggesting dysregulation of the core
pluripotency program. Data are normalized to mediumtreated cells. Error
bars indicate SEM.
90
Chapter Four Klf4 Overexpression Promotes Mouse and Rat ES Cell Self
Renewal
There are many proteins that may regulate self‐renewal downstream of STAT3. We
then wished to understand if any STAT3 downstream effectors may recapitulate the
effects of increased STAT3 expression or activation in promoting mouse and rat ES
cell self‐renewal.
4.1 Introduction and Rationale
Our initial choice was to investigate Klf family proteins: Krüppel‐Like Factor (Klf)
family genes are a highly conserved set of transcriptional factors that were
originally discovered in Drosophila studies(Kaczynski et al., 2003; Marin et al.,
1997; Schöck et al., 1999). Klfs are notable for their conserved zinc‐finger motif and
their tendency to regulate GC‐rich promoter regions(Marin et al., 1997). Klfs
regulate specific aspects of embryonic patterning, such as erythroid lineage
commitments (Klf2), gut tissue patterning (Klf4), and epithelial lineage patterning
(Klf5)(Kuo et al., 1997). Klf2, 4, and 5 are expressed in mouse ES cells and appear to
harbor significant functions that promote self‐renewal(Jiang et al., 2008).
Klfs have been known to be important in self‐renewing mouse ES cells for several
years, however, with the discovery of induced pluripotent (iPS) cells, Klf4 was found
to be one of the four core factors needed mediate this process(Takahashi and
91
Yamanaka, 2006). It was determined that four genes, Oct4, Sox2, cMyc, and Klf4,
were needed to reprogram fibroblasts into iPS cells. Oct4 and Sox2 were known to
directly regulate pluripotency in mouse and human ES cells. CMyc was found to be
essential in expediting the remodeling of the chromatin and is a potent mitogen. It
has been determined that Klf4 is critical in remodeling the epigenetic structure of
reprogramming cells and for also regulating the core pluripoteny network(Jiang et
al., 2008).
Recently, it has been established that Klf4 is the necessary factor to fully, and finally
reprogram pluripotent epiblast stem cells back into an ES‐like pluripotent state(Guo
et al., 2009). Since then, Dr. Hitoshi Niwa’s group has determined that Klf4 is likely
an important target of STAT3 upon LIF activation of gp130(Niwa et al., 2009).
Adding to this model, Dr. Austin Smith and colleagues conclude that Klf4 is regulated
by STAT3, with Klf5 as secondary target of regulation as well(Hall et al., 2009). Klf2
has been found to be a direct target of Oct4 in response to LIF/STAT3 signaling in
mouse ES cells. In sum, Klf2, 4, and 5 therefore make attractive targets immediately
downstream of STAT3 as effectors of the self‐renewal program. Therefore, I sought
to investigate the role of Klfs in regulating the self‐renewal of mouse ES cells.
4.2 – Klf4 Is Upregulated in Response to STAT3 Activation in Mouse ES Cells
We hypothesized that since STAT3 activation is sustained over time, it is likely true
that STAT3’s putative downstream targets are up‐regulated and maintained in a
92
sustained fashion. To test this, we began by seeding high density (~10
4
cells/cm
2
)
B6‐Y118F ES cells in co‐culture with CF‐1 feeders. Following serum‐starvation, the
mouse ES cells were stimulated with either 10ng/ml LIF, 0.2ng/ml G‐CSF, or
medium only. Total RNA was extracted at one hour and twenty‐four hours post‐
stimulation.
Quantitative PCR was performed to ascertain relative amounts of mRNA for Klf2,4,
and 5 (Figure 4.2a). As expected, Klf4 mRNA levels increase significantly by one hour
post‐LIF stimulation, and by twenty‐four hours have only increased modestly over
one hour. However, G‐CSF treated cells demonstrate only a slight increase in Klf4
expression by one hour. By twenty‐four hours, G‐CSF treated cells increase Klf4
expression five‐fold above controls. Despite a lagging increase as compared to LIF,
G‐CSF treatment increases expression of Klf4 higher than LIF induction does at
twenty‐four hours post‐induction. This finding suggests that increased and
sustained STAT3 activation may increase the expression of Klf4 RNA which remains
increased over twenty‐four hours .
93
Figure 4.2a: Mouse ES cells upregulate Klf4 in response to GCSF
treatment feederfree. B6Y118F mouse ES cells were treated with
either 0.2ng/ml GCSF, 10ng/ml LIF, 10ng/ml LIF plus 3uM
CHIR99201, or serum medium alone (‘NT’). Quantitative PCR analysis
reveals that LIF induces a significant increase in Klf4 RNA expression
by one hour as compared to GCSF or LIF/CHIR. By 24 hrs, however,
GCSF and LIF/CHIR significantly increase Klf4 RNA expression over
LIF. Both Klf2 and Klf5 show modest increases in response to GCSF.
Data is normalized to nontreated cells. Error bars indicate SEM.
94
4.3 – Klf4 Overexpression Prevents Crisis in Mouse ES Cells But Is Not Essential For
Maintaining SelfRenewal
To determine if overexpression of specific Klf proteins could enhance STAT3
mediated self‐renewal, we transfected wild‐type B6 mouse ES cells with retroviral
constructs containing either Klf2, 4, or 5 isoforms. After viral Klf RNA production
was verified (Figure 4.3a(A)), each cell type was then seeded upon a gelatinized
substrate containing serum medium and LIF. Only cells transfected with Klf4
displayed colonies morphologically consistent with self‐renewing ES cells that could
be passaged. Removal of LIF from Klf4 transfected cells also resulted in cellular
morphology consistent with differentiation (Figure 4.3a(B)). Klf4 overexpression
was confirmed by western blotting and densiometry (Figure 4.3a(C)). After five
passages feeder‐free, Klf4 overexpressing B6 mouse ES cells stain positive for Oct4
(Figure 4.3b).
95
Figure 4.3a: Klf4 overexpression enhances LIFmediated feederfree B6 ES cell self
renewal. (A) Wildtype B6 mouse ES cells were retroviral transfected with Klf2, 4, or
5 isoforms. RTPCR confirms viral RNA production. (B) Clones infected with each Klf
were seeded onto gelatinized substrates and supplemented with LIF. Only Klf4
transfected cells demonstrated colony morphology consistent with selfrenewal. LIF
was then removed from the Klf4 transfected cells and immediate differentiation was
observed. (C) Western blot indicating Klf4 overexpression following transfection.
(D) Densiometric analysis of (C) that demonstrates that Klf4 is expressed almost
twofold higher than over wildtype. Data are normalized to βactin.
96
We then overexpressed Klf4 in BalbC1‐Y118F mouse ES cells. As indicated in Figure
4.3c, wild‐type BalbC1 ES cells fail to form tight, compacted, and dome‐shaped
colonies when seeded into feeder‐free environment despite supplementation with
LIF. However, when transfected with Klf4, LIF supplementation results in colonies
consistent with undifferentiated, self‐renewing ES cells despite the absence of
feeders. These data suggest that Klf4 overexpression can enhance feeder‐free self‐
Figure 4.3b: Klf4 overexpression permits longterm culture feederfree in a LIF
dependent manner. Wildtype B6 mouse ES cell overexpressing Klf4 are able
stain Oct4 positive after five passages in feederfree conditions stimulated by
LIF.
97
renewal in a LIF‐dependent fashion in more than one non‐permissive mouse ES cell
line. Klf4 overexpression plus LIF supplementation results in cells morphologically
consistent with self‐renewing ES cells expressing the chimeric gp130 receptor and
stimulated with G‐CSF. These results suggest that Klf4 overexpression augments
STAT3 mediated self‐renewal feeder‐free.
Figure 4.3c: Klf4 overexpression enhances BalbC1 mouse ES cell feederfree
selfrenewal. (A) Wildtype BalbC1 mouse ES cells seeded feederfree cease
proliferating and appear to differentiate despite LIF supplementation. (B)
Klf4 transfection into wildtype BalbC1 mouse ES cells renders them
consistent with undifferentiated ES cells in feederfree culture. (C) BalbC1 ES
cells expressing the chimeric gp130 receptor fail to selfrenew in the
presence of LIF feederfree. (D) GCSF stimulation renders the same cells
able to selfrenew.
98
To determine if Klf4 expression was essential for STAT3 mediated self‐renewal, we
transfected B6‐Y118F mouse ES cells with shRNA constructs targeted against Klf4
(‘B6‐Y118F‐K4KD’). Quantitative PCR and western blotting verify significant knock‐
down of Klf4 RNA and protein levels as compared to untransfected cells (Figure
4.3d). We then seeded Klf4‐depleted B6‐Y118F and mock‐infected B6‐Y118F mouse
ES cells onto gelatinized substrates supplemented with 0.2ng/ml G‐CSF. Despite
Klf4 knock‐down, B6‐Y118F cells were able to self‐renew comparably to cells with
normal Klf4 expression levels (Figure 4.3e(A)). Immunostaining reveals Oct4
positive cells after five passages feeder‐free in G‐CSF (Figure 4.3e(B)). These data
suggest that Klf4 expression may not be essential in promoting STAT3‐mediated
mouse ES cell self‐renewal.
Figure 4.3d: Klf4 knockdown is efficient in mouse ES cells. (A) Western blot
demonstrating endogenous Klf4 in B6Y118F mouse ES cells is depleted as
compared to scrambled shRNA control. (B) Densiometry indicates that both
shRNA constructs deplete Klf4 protein by at least 50 percent or more. Data
normalized to βactin expression (C) qPCR indicates that Klf4 RNA levels are
depleted as well following shRNA transfection. Data normalized to mock
infected. Error bars indicate SD.
99
Figure 4.3e: Klf4 expression is not essential for mouse feederfree self
renewal. (A) B6Y118F mouse ES cells infected with Klf4 shRNA and
stimulated with GCSF in a feederfree environment selfrenew efficiently.
(B)Immunostaining of B6Y118F transfected with Klf4 shRNA after five
passages in GCSF in feederfree conditions. Both cell lines stain positive for
Oct4.
100
Despite the ability to be passaged with no apparent differentiation, quantitative PCR
analysis reveals that Oct4, Nanog, and Rex‐1 RNA expression levels are significantly
reduced in Klf4 depleted ES cells (Figure 4.3f(A)). In addition, early markers of
differentiation are upregulated as a result of Klf4 knock‐down, yet as observed
earlier, self‐renewal appears to not be adversely affected (Figure 4.3f(B)). These
data suggest that Klf4 expression is likely accessory to, but not critical for
maintaining STAT3‐mediated mouse ES cell self‐renewal.
Figure 4.3f: Klf4 knockdown decreases pluripotency gene expression and
increases early differentiation markers. (A) qPCR analysis demonstrates that
Klf4 knockdown results in decreased RNA levels of Oct4, Nanog, and Rex1.
(B) qPCR analysis upon the same cell lines indicate that Klf4 knockdown
results in increased early differentiation markers. Data are normalized to
untransfected B6Y118F cells. Error bars indicate SD.
101
Chapter Five STAT3 Activation May Promote Tumor Cell Growth Arrest
5.1 – Introduction and Rationale
STAT3 as an oncogene is well‐characterized in the literature(Corvinus et al., 2005;
Kusaba et al., 2005). The most common transformation event related to STAT3 is
constitutive activation. Aberrant STAT3 regulation is often found in specific subsets
of breast, leukemic, and colon cancers. Dysregulated STAT3 has been correlated to
enhanced tumor survival and poor prognosis(Ling and Arlinghaus, 2005). It is
worthwhile then to investigate if specific aspects of STAT3 regulation in ES cells can
translate to tumor cells with known STAT3 dysregulation. Many tumors, like ES
cells, have at least a small population of cells that self‐renew and resist therapeutic
treatment(Villalva et al., 2010; Welte et al., 2010). Is STAT3 integral in mediating
this process? ES cells provide an isolated environment to study STAT3 without the
limitations of other or more numerous mutations found in cancer cell lines(Foster et
al., 2000; Yang et al., 2005).
As previously shown, in specific instances, STAT3 activation can induce
differentiation in mouse ES cells. It is well‐characterized that many cancers
demonstrate aberrant gene behavior or protein production. Of the many oncogenes
activated or mutated, STAT3 is found to be either constitutively activated or exhibit
slightly higher than normal expression levels(Bromberg et al., 1998; Corvinus et al.,
2005; Kusaba et al., 2005; Ling and Arlinghaus, 2005; Ling et al., 2007). We
102
hypothesized that increasing STAT3 activation may have the effect observed in
mouse ES cells, whereby increasing STAT3 activation may actually promote
differentiation in tumor cells.
To test if increasing endogenous STAT3 activation in human tumor cells is able to
induce differentiation, we selected tumor cell lines that had well‐characterized
STAT3 mutations. Two cell lines, DLD‐1 and HT‐29 were selected for their aberrant
STAT3 activity(Corvinus et al., 2005; Kusaba et al., 2005). DLD‐1 and HT‐29 are both
human colon cancer cell lines of epithelial origin(Fogh and Trempe, 1975; Trainer et
al., 1988).
Both cell lines were expanded, transfected stably with the chimeric gp130 receptor,
and drug selected for individual clones. Western blotting revealed a functional
chimeric receptor was able to robustly activate STAT3 in human tumor cells (Figure
5.1a). Western blotting also revealed a significant decrease in ERK1/2 activation
upon high G‐CSF concentration (2.5ng/ml) stimulation. Upon stimulation by G‐CSF,
several clones demonstrated little cellular attachment to the tissue culture dish,
suggesting the cells may be dying. Densiometry confirms that ERK1/2
phosphorylation is decreased in response to high G‐CSF concentrations (Figure
5.1b). To rule out off‐target effects of G‐CSF, wild‐type HT‐29 and DLD‐1 cells were
exposed to 10ng/ml G‐CSF for seven days. No apparent change in growth was
observed as compared to untreated cells (Figure 5.1c).
103
Figure 5.1a: GCSF Mediates STAT3 activation and correlates to
inhibited ERK1/2 activation. HT29Y118F cells were treated with
a gradient of GCSF for 30 minutes. Western blotting reveals a
decrease in STAT3 activation as the GCSF concentration is
decreased. ERK1/2 activation is inhibited to background levels
under the highest GCSF concentration. STAT3 expression appears
to decrease in correlation with STAT3 activation.
104
Figure 5.1b: Densiometry confirms decreased ERK1/2 activation in response
to GCSF in HT29 tumor cells. Image analysis of Figure 5.1a demonstrates
that high STAT3 phosphorylation correlates to decreased ERK1/2
phosphorylation at 2.5 ng/ml GCSF. STAT3 expression also decreases with
GCSF concentration. ERK1/2 activation remains constant below 2.5 ng/ml
GCSF. Data normalized to tubulin.
105
To quantify the effects of G‐CSF in tumor cells, we plated an equal number of HT‐29
and DLD‐1 human tumor cells cells, ~5x10
3
cells/cm
2
into separate wells, and
stimulated them with either 1ng/ml G‐CSF or medium alone. Compared to wild‐type
cells, the clones expressing the chimeric receptor exposed to G‐CSF demonstrated
almost a total reduction in cell number by Day 7 (Figure 5.1d‐e). This result suggests
that STAT3 activation may be correlated to decreased total cell number.
Figure 5.1c: WildType Human Tumor Cell Lines Demonstrate No Discernible
Effect to GCSF Treatment. (A) Wildtype HT29 human colon cancer cells
were subjected to high doses of GCSF for 5 days. No appreciable effect was
observed. (B) Wildtype DLD1 human colon cancer cells were subjected to
high doses of GCSF for 5 days. Again, no appreciable effect was observed.
106
Figure 5.1d: Increased STAT3 Activation Correlates to Decreased Cell
Number in HT29Y118F cells. Top: (A) Nontreated HT29Y118F cells,
day 7. (B) Western blot demonstrating GCSF mediated STAT3
phosphorylation (C) 100 pg/ml GCSF stimulation demonstrating
detached cells, Day 7 (D) 1ng/ml GCSF stimulation demonstrating
complete detachment of cells to substrate, Day 7.
107
STAT3 has been well‐characterized as a positive regulator of cell cycle progression
in somatic tissues, namely by facilitating the G1 to S phase transition(Fukada et al.,
1998). This is accomplished via gp130 activation of STAT3 and the associated
upregulation of cyclins D and A and cdc25a. STAT3 activation is also a potent
inhibitor of the cell‐cycle inhibitor p21. STAT3’s effect upon DLD‐1 and HT‐29
human tumor cells may be through cell‐cycle regulation, therefore it must be
established that proliferation is inhibited first.
Figure 5.1e: Increased STAT3 activation correlates to decreased cell number
in DLD1Y118F cells. (A) Nontreated DLD1Y118F cells, day 7. (B) 1ng/ml
GCSF treatment, Day 7. (C) Western blot demonstrating GCSF meditated
STAT3 phosphorylation.
108
5.2 – STAT3 Activation Correlates to Decreased Tumor Cell Proliferation
To further investigate STAT3’s effects upon tumor cells, we plated 10
5
DLD‐1 or HT‐
29 clones containing the chimeric receptor. The cells were passaged every two days.
10
5
cells of each type were passaged for six passages, or until there were less than
10
5
cell available to passage. Each cell type was stimulated with a gradient of G‐CSF
concentrations or medium only. Cells stimulated with G‐CSF showed a stagnant cell
number that decreased with each passage when compared to no treatment (Figure
5.2a). Non‐treated cells exhibit an initial decrease in total cell number, but recover
by passage five. Supplementation with 1ng/ml G‐CSF correlated to an inability for
the cells to recover, as no cells could be subcultured by passage four. These data
indicate that high G‐CSF concentrations applied to human tumor cells expressing the
chimeric gp130 receptor may increase cellular death, inhibit proliferation, or both
may be occurring .
109
We assayed for cumulative cell number changes in HT29‐Y118F cells seeded at
clonal density (250 cells/cm
2
) supplemented with a gradient of G‐CSF (1ng/ml –
0.01ng/ml G‐CSF) in serum medium for six days. Indeed, when given a multitude of
concentrations, the lower cell density appears to affect total cell number in an
inversely proportional manner as compared to the G‐CSF concentration present
(Figure 5.2b). We observed no adherent cells by Day 4 when 1ng/ml or 0.1ng/ml G‐
CSF was employed. These observations indicate that G‐CSF treatment of chimeric
Figure 5.2a: GCSF stimulation correlates to decreased cumulative
cell number. HT29Y118F cells were seeded at 10
4
cells/cm
2
. GCSF
was added and the cells were counted every other day at passage.
1ng/ml GCSF resulted in complete ablation by passage 4, while all
other treatments did not significantly affect cell number.
110
receptor‐harboring human tumor cells may be adversely affecting cell survival
and/or proliferation.
Figure 5.2b: GCSF Prevents Tumor Cell Proliferation at Low Cell Density and
During Exponential Growth. Top: 250 HT29Y118F cells/cm
2
were seeded
and GCSF was applied in a gradient fashion. Cells were counted everyday
for six days. All treatments prevented significant growth, while 1ng/ml and
0.1ng/ml GCSF concentrations resulted in no surviving cells to count.
111
Discussion
D.1 – STAT3 Is Regulated In a Strain or SpeciesSpecific Manner to Promote
Rodent ES Cell SelfRenewal
Previously, mouse and rat ES cells have shared only a STAT3‐independent self‐
renewal pathway, however this mechanism is poorly understood (Ying et al.,
2008a). Since mouse ES cells can self‐renew in both a STAT3‐dependent and
independent fashion, we hypothesized that rat ES cells must share both features as
well. Our work has presented evidence that rat ES cell do in fact share both a STAT3‐
dependent as well as independent self‐renewal mechanism with the mouse.
To elucidate this claim, we had to discard with the idea that STAT3 activation
functions as a binary ‘on/off’ system. Rather, modulating STAT3 activation in a
strain or species‐specific manner has the effect of enhancing both mouse and rat ES
cell self‐renewal ability. One obstacle that likely prevented previous derivations of
rat ES cells is that rat ES cell STAT3 activation requirements are different from the
mouse, as mouse ES cells require an approximately ten‐fold higher G‐CSF
concentration than rat to maintain self‐renewal. This increased G‐CSF concentration
likely correlates to a significant increase in STAT3 activation. Receptor expression
levels are likely important in regulating STAT3 activation levels as well. In addition,
LIF stimulation has been shown to be refractory to rat ES cell derivation. This
112
apparent discrepancy in response to LIF may not be because LIF does not support
rat ES cell self‐renewal, but rather because STAT3 is regulated differently upon LIF
induction within each species. This evidence suggests that additional species‐
specific levels of STAT3 regulation are likely necessary to promote rodent ES cell
self‐renewal.
We have presented evidence that attempts to determine why specific ES cell lines
are able to self‐renew feeder‐independently. This work demonstrates that STAT3
activation does not function as a binary ‘on/off’ system, but rather more in line with
a rheostat‐like effect composed of intermediate cell‐states aggregately referred to as
‘self‐renewal.’ This finding not only refines our understanding of the role of STAT3
in mouse ES cell self‐renewal, but also provides a signaling connection that explains
how 129 mouse ES cell strains, but not B6 mouse ES cells, manage feeder‐
independent self‐renewal.
Our results suggest that STAT3 expression and activation levels are critically
important in permitting feeder‐independent mouse ES cell self‐renewal. This finding
argues that the differences in feeder‐free self‐renewal ability between mouse ES cell
strains are related to each strain or species’ inherent permissiveness(Hanna et al.,
2009). Our data suggests that feeder‐independent 46C mouse ES cells harbor an
increased STAT3 expression level in response to LIF. This increased expression level
permits an increased proportion of activated STAT3, which correlates to the ability
113
to self‐renew feeder‐free. B6 mouse ES cells, when engineered to either overexpress
STAT3 or artificially increase endogenous STAT3 activation, are also rendered able
to self‐renew feeder‐free, thus enhancing their self‐renewal ability. This result also
implies that the STAT3‐dependent self‐renewal mechanism is more accurately
described as the aggregate of many intermediate STAT3‐phosphorylation levels
which can be finely tuned by the cell or the investigator given the specific culture
milieu.
D.2 The Role of STAT3 In Mediating Mouse and Rat ES Cell SelfRenewal
Interestingly, STAT3 overexpression correlates to increased core pluripotency gene
transcript expression and decreased ERK1/2 phosphorylation. Together, this result
suggests that increasing STAT3 expression levels has a net positive effect in
promoting ES cell self‐renewal feeder‐free. However, in contrast to this finding, we
report that overexpressing STAT3, then hyperactivating such a large pool results in
acute differentiation of mouse ES cells, and may reduce proliferation of specific
tumor cell populations. If STAT3 activation is found to have a differentiation‐
inducing ability as well in specific circumstances, then novel approaches to tumor
suppression may be developed. Instead of complete inhibition of STAT3 activation,
hyperactivation of STAT3 may exacerbate proliferation inhibition. Within ES cells
however, our observation that STAT3 may induce differentiation when
hyperactivated reaffirms that STAT3 likely functions within a self‐renewal range
with a functional ceiling on self‐renewal (Figure D.2a).
114
Building upon this premise, we provide evidence that rat ES cells are able to self‐
renew in a STAT3‐dependent manner. Prior to this work the rat ES cell self‐renewal
had been suggested to be STAT3‐independent(Buehr et al., 2008; Li et al., 2008).
However, STAT3 overexpression or artificial STAT3 activation results in robust self‐
renewal for both mouse and rat ES cells. What remains to be determined is if feeder‐
independent rat ES cell self‐renewal is possible. Currently, rat ES cells cannot self‐
renew in the absence of feeders. Determining an appropriate feeder‐independent
Figure D.2a: STAT3 signaling appears to have a functional range that
effectively promotes ES cell selfrenewal. Based on the work of others and
myself, mouse and rat ES cells harbor a speciesspecific range for STAT3
expression and activation that fundamentally outlines the selfrenewal
program for each celltype. Finding the functional range by modulating
specific aspects of the selfrenewal machinery for each species is tantamount
to understanding how to derive novel mammalian ES cell species, or at least
in the shortterm novel rodent ES cell species.
115
rat ES cell culture system will allow for more accurate direct comparisons of mouse
and rat ES cells in vitro.
We also report on putative downstream targets of STAT. Since it has been reported
that Klf genes are likely critical targets of STAT3 activation, we sought to understand
the relationship specific Klf genes may have with STAT3(Hall et al., 2009; Niwa et al.,
2009). We note that only Klf4 demonstrates a significant LIF‐mediated increase in
RNA expression over time in mouse ES cells. G‐CSF stimulation further bolstered
this finding by demonstrating that increasing STAT3 activation correlates to an
increase in Klf4 RNA expression, suggesting Klf4 expression is potentially regulated
by active STAT3.
Klf4 overexpression in B6 feeder‐dependent mouse ES cells renders the cells able to
self‐renew feeder‐free in a STAT3 dependent manner. This result is in contradiction
to the finding by Hall et al. that forced Klf4 expression promotes LIF‐independent
self‐renewal(Hall et al., 2009). We note that Hall et al. indicate that self‐renewal was
severely impaired in the absence of LIF, and that self‐renewal was improved when
LIF was included. We speculate that ES strain differences or transfection efficiency
may account for the discrepancy that our cells require LIF to self‐renew feeder‐free.
In addition there is evidence that Klf4 overexpression itself has cytostatic or toxic
effects upon self‐renewing mouse ES cells(Niwa et al., 2009). We however, confirm
also the specific finding by Hall et al. that STAT3 expression is necessary to support
self‐renewal despite Klf4 overexpression. This evidence supports the notion that
116
Klf’s are but one aspect of a critical cadre of downstream targets of STAT3 necessary
to execute self‐renewal(Jiang et al., 2008). It has been also demonstrated that either
Klf2 or 4 forced expression is sufficient to reprogram fibroblasts(Nakagawa et al.,
2008). Evidence suggests that unlike STAT3, however, Klf4’s role is a gate that
promotes naïve pluripotency while preventing the transition to the EpiSC
state(Feng et al., 2009). Given that Klf4 augments self‐renewal, but does not appear
to be essential in promoting it, this view likely true. Therefore, further
investigations into Klf4 as it pertains to self‐renewal may be unnecessary, as Klf4
appears to be a reprogramming regulator and developmental gate, but not directly a
self‐renewal regulator.
It has been demonstrated that, of the Klfs expressed in mouse ES cells, only Klf2
overexpression can promote LIF‐independent mouse ES cell self‐renewal(Hall et al.,
2009). However, no one particular Klf expressed in mouse ES cells appears to be
critical for maintenance of the naïve pluripotent state, suggesting that Klf proteins
are but one aspect of the cloud of signal networks mediating self‐renewal (Jiang et
al., 2008).
D.3 The Role of ERK1/2 In Mouse ES Cell SelfRenewal
We confirm the finding that ERK1/2 activation is detrimental to STAT3‐mediated
mouse ES cell self‐renewal(Burdon et al., 1999; Kunath et al., 2007). However we
reveal to what extent ERK1/2 signaling potently inhibits STAT3‐mediated feeder‐
117
independent self‐renewal: Neither LIF nor ERK1/2 inhibition alone increased
feeder‐free self‐renewal capability, yet in combination feeder‐free self‐renewal was
significantly increased. This suggests a synergy between activation and inhibition,
and not simply STAT3 activation, supports naïve pluripotent self‐renewal. This idea
is firmly in line with other reports that MAPK/ERK signaling is directly in opposition
to STAT3 signaling in mouse ES cells (Greber et al., 2010).
Intriguingly, STAT3 overexpression correlates to a significant decrease in ERK1/2
activation, suggesting increased STAT3 expression may indirectly inhibit ERK1/2,
thus reinforcing naïve pluripotency. More work is needed to verify if in fact STAT3
might be regulating ERK1/2 activation. However, if true, our results may shed light
upon the mechanism behind murine diapause. It is known that LIF is secreted by the
trophectoderm which signals to the ICM to self‐renew. However, cell‐cycling in the
diapaused epiblast is slowed and proliferation is halted, but evidence exists that 129
strain epiblasts have a significantly higher proportion of cycling cells as compared
to other mouse strains(Batlle‐Morera et al., 2008). Increased STAT3 expression in
the epiblast in utero may inhibit ERK1/2 activation, decreasing proliferation, which
would promote self‐renewal.
Also, recent evidence suggests that ERK signaling is needed to stabilize the primed
pluripotent state by inhibiting Klf4 expression and therefore acts as a direct
impediment in reprogramming from EpiSC to ES cell pluripotency(Greber et al.,
2010). If true, this suggests that increased STAT3 activation may further increase
118
Klf4 and Klf2 expression, which has been shown to stabilize naïve pluripotency by
inhibiting ERK1/2 activation(Greber et al., 2010). Finally, ERK expression is likely
unnecessary for naïve pluripotency, as erk2 null mouse ES cells appear to be normal
in their capacity to self‐renew and erk1 null mice are viable(Pagès et al., 1999; Yao
et al., 2003).
D.4 Rodent ES Cell SelfRenewal: A Stratified Model
STAT3 also likely plays a critical role in regulating the naïve pluripotent state.
Recent evidence has emerged that indicates that pluripotency itself is stratified into
different stable states. Hanna et al. demonstrate that STAT3 harbors a critical role in
creating human iPS cell populations that mirror mouse ES cell naïve pluripotency by
using the combination of STAT3 activation, the combined inhibition of GSK3β and
MAPK, and the combined ectopic expression of Klf2 and 4 (Hanna et al., 2010).
Indeed, mouse self‐renewal can be maintained using mechanisms that are STAT3‐
dependent (LIF), independent (2i), or both (LIF/2i) (Guo et al., 2009). We propose
that the naïve pluripotent state’s self‐renewal mechanism is itself a stratified,
metastable cell‐state composed of at least two distinct mechanisms that are shared
between mouse and rat (Figure D.4a). Now, due to the conserved nature of STAT3 in
mouse, rat, and human ES cells, direct in vitro comparisons can now be made that
address the significance of downstream self‐renewal targets.
119
The ground state of ES cells fits into this model as well, suggesting that the ground
state can be interconverted to employ the more traditional inductive, or STAT3‐
dependent, self‐renewal mechanism by induction with LIF. Differentiation in this
model may be induced by increasing or decreasing either STAT3 expression,
Figure D.4a: Rodent ES cell selfrenewal is metastable cellstate within naive
pluripotency. The ground state of mouse ES cells is defined as a STAT3 independent
mechanism that relies upon inhibited MAPK/ERK and GSK3β to promote self
renewal. These two selfrenewal mechanisms can be switched depending on the
culture conditions. By increasing STAT3 expression or activation acutely, and by
inhibiting MAPK/ERK signaling slightly less, more traditional inductivebased ES
cells can be cultured. Further increases in MAPK signaling or a decrease in STAT3
activation will result in primed ES cell transition or overt differentiation.
120
activation, or increasing ERK1/2 activation (Figure D.4b). Differentiation may be
averted by chemical or genetic modification to ‘re‐set’ the balance between
competing signal cascades.
We posit that the self‐renewal mechanism is itself stratified into distinct cell‐states.
These cell‐states can transition between different ranges of STAT3 activation and
STAT3‐independent pathways within naïve pluripotency depending on the
permissiveness of the species or strain. This model predicts that there are different
rodent self‐renewal mechanisms which are themselves stratified within a specific
pluripotent cell‐state. Both the STAT3‐dependent and ground‐state rodent self‐
renewal mechanisms are relegated to describing naïve pluripotency (Nichols and
Smith, 2009).
121
Figure D.4b: Selfrenewal represents a finelytuned interplay between STAT3
and MAPK/ERK signaling in rodent ES cells. Selfrenewal can be defined as any
cellstate that maintains a significantly high proportion of expressed and active
STAT3 as compared to active MAPK/ERK signaling. Either increasing ERK or
decreasing STAT3 activation disrupts this balance and results in ES cell
differentiation. In this scenario, no signal protein exerts all the selfrenewal
effect, as some beneficial mitogenic effects are likely important from ERK
signaling.
122
The idea of a multiplicity of self‐renewal mechanisms within naïve pluripotency is
likely transferable to other pluripotent stem cell populations. This model may be
transferable to other species, and may help explain why LIF is insufficient in
deriving and maintaining porcine or bovine ES cells(Alberio et al., 2010; Ezashi et
al., 2009; Gong et al., 2010). In addition, we have shown that STAT3 is regulated
temporally. As well, STAT3‐independent mechanisms may prove to be temporally
regulated as well. Likely, this temporal regulation coupled to conserved
downstream targets may explain why the observed self‐renewal mechanisms
appear separate, but in reality may be highly interconnected. In addition, this model
predicts the existence of additional self‐renewal mechanisms within the primed
pluripotent cell‐state in addition to the bFGF/MAPK pathways already
characterized. There may be a ‘ground‐state’ of primed pluripotency, which is yet to
be observed, but likely exists.
We add that this idea of conserved, metastable self‐renewal mechanisms will also
aid in the derivation and maintenance of novel mammalian ES cell species, and will
underpin future direct comparisons of mouse and rat ES cells. A more thorough
understanding of the self‐renewal mechanisms and their conserved aspects will
likely expedite the derivation of mammals that may less permissive than humans.
123
D.5 Future Directions and Implications
As a general future direction, it is worth noting that currently two distinct ES culture
systems exist: STAT3‐dependent or STAT3‐independent. What is of fundamental
importance is how these two vastly different signaling systems result in the same
outcome: Self‐renewal. If and where these distinct mechanisms converge upon the
self‐renewal program is of great interest to our group, as this point likely represents
the conserved aspect of self‐renewal necessary to facilitate a wider variety of
mammalian ES cell species to be derived. Conversely, these two pathways may
represent two truly independent self‐renewal pathways, acting in a compensatory
fashion when the other is impaired or removed. Therefore transcriptional and
genomic analysis of the downstream targets of both mechanisms in both mouse and
rat ES cells is necessary. It is also worth determining if the suggestion by Niwa et al.
is true that a STAT3‐independent mechanism is running in parallel to the STAT3‐
dependent mechanism, as the ground state model has been extensively studies
within STAT3‐devoid systems(Niwa et al., 2009).
With regards to tumor biology, it seems more than tenuous that the inherent
proclivity to develop pluripotent tumor types in the 129 strain of mouse is not
correlated to the fact that the ES cell lines yielded are capable of feeder‐free
derivation. Since ES cells share many characteristics with tumor cells, it seems
reasonable to posit that 129 ES cells, in their ability to adhere and proliferate
efficiently without the aid of any supporting tissue, likely by their ability to increase
124
STAT3 expression and activation, is behavior consistent with metastasizing tumor
cells. Strains of mice that yield only feeder‐dependent mouse ES cells not only lack
this ability to quickly modify their STAT3 expression levels to retain feeder‐
independence, they lack the significant occurrence of spontaneous pluripotent
tumor cell types. It is worth investigating the connection between the relative
incidence of cancer as compared to efficiency of ES cell derivation, specifically
feeder‐free derivation between different rat and mouse strains. Indeed, evidence
exists that the attempts to connect the 129 strains relatively high tumor
susceptibility rate to its high permissiveness is genetically determined(Anderson et
al., 2009).The ability to derive ES cells feeder‐free may indicate a reasonably
significant relationship between overall susceptibility to germ cell tumors and ease
of ES cell derivation. Whether the rate of ES cell derivation significantly correlates to
other tumor type susceptibilities is less certain, but would be of interest as well. Gail
Martin herself speculated upon this issue tangentially in her 1981 manuscript
outlining the derivation process, hinting that the connection between ES cells and
tumor cells may be indicative of a more fundamental relationship than once thought.
D.6 Conclusions
In rodent ES cell biology, Stat3 has begun to reveal its true pleiotropy. Further
investigations will no doubt uncover the downstream targets, their likely intimate
relationship with STAT3, and the ultimate effect upon the stem cell phenotype. In
all, a more thorough comprehension of STAT3 and the ES cell self‐renewal
125
mechanism will likely aid in the successful derivation of ES cells in novel species. To
the extent of rodent ES cells, our work has laid the foundation for accurate in vitro
inter‐species comparisons, which will likely aid in further refinements to the
fundamental mechanism of ES cell self‐renewal.
126
Bibliography
Alberio, R., Croxall, N., and Allegrucci, C. (2010). Pig Epiblast Stem Cells Depend on
Activin/Nodal Signaling for Pluripotency and Self Renewal. Stem Cells and
Development.
Anderson, P.D., Nelson, V.R., Tesar, P.J., and Nadeau, J.H. (2009). Genetic factors on
mouse chromosome 18 affecting susceptibility to testicular germ cell tumors and
permissiveness to embryonic stem cell derivation. Cancer Research 69, 9112‐9117.
Batlle‐Morera, L., Smith, A., and Nichols, J. (2008). Parameters influencing derivation
of embryonic stem cells from murine embryos. Genesis (New York, NY: 2000) 46,
758‐767.
Becker, S., Groner, B., and Müller, C.W. (1998). Three‐dimensional structure of the
Stat3beta homodimer bound to DNA. Nature 394, 145‐151.
Bogden, A.E., Haskell, P.M., LePage, D.J., Kelton, D.E., Cobb, W.R., and Esber, H.J.
(1979). Growth of human tumor xenografts implanted under the renal capsule of
normal immunocompetent mice. Experimental Cell Biology 47, 281‐293.
Boulton, T.G., Stahl, N., and Yancopoulos, G.D. (1994). Ciliary neurotrophic
factor/leukemia inhibitory factor/interleukin 6/oncostatin M family of cytokines
induces tyrosine phosphorylation of a common set of proteins overlapping those
induced by other cytokines and growth factors. Journal of Biological Chemistry 269,
11648‐11655.
Bromberg, J.F., Horvath, C.M., Besser, D., Lathem, W.W., and Darnell, J.E. (1998).
Stat3 Activation Is Required for Cellular Transformation by v‐src. Mol Cell Biol 18,
2553‐2558.
Brons, I.G.M., Smithers, L.E., Trotter, M.W.B., Rugg‐Gunn, P., Sun, B., Chuva de Sousa
Lopes, S.M., Howlett, S.K., Clarkson, A., Ahrlund‐Richter, L., Pedersen, R.A., et al.
(2007). Derivation of pluripotent epiblast stem cells from mammalian embryos.
Nature 448, 191‐195.
127
Brook, F.A., and Gardner, R.L. (1997). The origin and efficient derivation of
embryonic stem cells in the mouse. Proceedings of the National Academy of Sciences
of the United States of America 94, 5709‐5712.
Buecker, C., Chen, H.‐H., Polo, J.M., Daheron, L., Bu, L., Barakat, T.S., Okwieka, P.,
Porter, A., Gribnau, J., Hochedlinger, K., et al. (2010). A Murine ESC‐like State
Facilitates Transgenesis and Homologous Recombination in Human Pluripotent
Stem Cells. Cell Stem Cell 6, 535‐546.
Buehr, M., Meek, S., Blair, K., Yang, J., Ure, J., Silva, J., McLay, R., Hall, J., Ying, Q.L., and
Smith, A. (2008). Capture of authentic embryonic stem cells from rat blastocysts.
Cell 135, 1287–1298.
Burdon, T., Smith, A., and Savatier, P. (2002). Signalling, cell cycle and pluripotency
in embryonic stem cells. Trends in Cell Biology 12, 432‐438.
Burdon, T., Stracey, C., Chambers, I., Nichols, J., and Smith, A. (1999). Suppression of
SHP‐2 and ERK signalling promotes self‐renewal of mouse embryonic stem cells.
Developmental biology 210, 30–43.
Castro, J.E., and Cass, W. (1974). Maintenance of human tumours and tissues in
immunosuppressed mice. The British Journal of Surgery 61, 421‐426.
Chambers, I., Cozens, A., Broadbent, J., Robertson, M., Lee, M., Li, M., and Smith, A.
(1997). Structure of the mouse leukaemia inhibitory factor receptor gene: regulated
expression of mRNA encoding a soluble receptor isoform from an alternative 5'
untranslated region. The Biochemical Journal 328 ( Pt 3), 879‐888.
Corvinus, F.M., Orth, C., Moriggl, R., Tsareva, S.A., Wagner, S., Pfitzner, E.B., Baus, D.,
Kaufmann, R., Huberb, L.A., Zatloukal, K., et al. (2005). Persistent STAT3 Activation
in Colon Cancer Is Associated with Enhanced Cell Proliferation and Tumor Growth.
Neoplasia (New York, NY) 7, 545‐555.
Dalton, S. (2009). Exposing Hidden Dimensions of Embryonic Stem Cell Cycle
Control. Cell Stem Cell 4, 9‐10.
128
Darnell, J.E., Kerr, I.M., and Stark, G.R. (1994). Jak‐STAT pathways and
transcriptional activation in response to IFNs and other extracellular signaling
proteins. Science (New York, NY) 264, 1415‐1421.
Davis, S., Aldrich, T., Stahl, N., Pan, L., Taga, T., Kishimoto, T., Ip, N., and Yancopoulos,
G. (1993). LIFR beta and gp130 as heterodimerizing signal transducers of the
tripartite CNTF receptor. Science 260, 1805‐1808.
Duval, D., Reinhardt, B., Kedinger, C., and Boeuf, H. (2000). Role of suppressors of
cytokine signaling (Socs) in leukemia inhibitory factor (LIF) ‐dependent embryonic
stem cell survival. The FASEB Journal: Official Publication of the Federation of
American Societies for Experimental Biology 14, 1577‐1584.
Eriksen, K.W., Kaltoft, K., Mikkelsen, G., Nielsen, M., Zhang, Q., Geisler, C., Nissen,
M.H., Röpke, C., Wasik, M.A., and Odum, N. (2001). Constitutive STAT3‐activation in
Sezary syndrome: tyrphostin AG490 inhibits STAT3‐activation, interleukin‐2
receptor expression and growth of leukemic Sezary cells. Leukemia: Official Journal
of the Leukemia Society of America, Leukemia Research Fund, UK 15, 787‐793.
Evans, M.J., and Kaufman, M.H. (1981). Establishment in culture of pluripotential
cells from mouse embryos. Nature 292, 154‐156.
Ezashi, T., Telugu, B.P.V.L., Alexenko, A.P., Sachdev, S., Sinha, S., and Roberts, R.M.
(2009). Derivation of induced pluripotent stem cells from pig somatic cells.
Proceedings of the National Academy of Sciences of the United States of America
106, 10993‐10998.
Feng, B., Jiang, J., Kraus, P., Ng, J.‐H., Heng, J.‐C.D., Chan, Y.‐S., Yaw, L.‐P., Zhang, W.,
Loh, Y.‐H., Han, J., et al. (2009). Reprogramming of fibroblasts into induced
pluripotent stem cells with orphan nuclear receptor Esrrb. Nat Cell Biol 11, 197‐203.
Fogh, J., and Trempe, G. (1975). New human tumor cell lines. Human tumor cells in
vitro, 115–159.
Foster, K.W., Frost, A.R., McKie‐Bell, P., Lin, C.Y., Engler, J.A., Grizzle, W.E., and
Ruppert, J.M. (2000). Increase of GKLF messenger RNA and protein expression
during progression of breast cancer. Cancer Research 60, 6488‐6495.
129
Fukada, T., Hibi, M., Yamanaka, Y., Takahashi‐Tezuka, M., Fujitani, Y., Yamaguchi, T.,
Nakajima, K., and Hirano, T. (1996). Two signals are necessary for cell proliferation
induced by a cytokine receptor gp130: involvement of STAT3 in anti‐apoptosis.
Immunity 5, 449–460.
Fukada, T., Ohtani, T., Yoshida, Y., Shirogane, T., Nishida, K., Nakajima, K., Hibi, M.,
and Hirano, T. (1998). STAT3 orchestrates contradictory signals in cytokine‐induced
G1 to S cell‐cycle transition. The EMBO Journal 17, 6670‐6677.
Gachelin, G., Kemler, R., Kelly, F., and Jacob, F. (1977). PCC4, a new cell surface
antigen common to multipotential embryonal carcinoma cells, spermatozoa, and
mouse early embryos. Developmental Biology 57, 199‐209.
Gardner, R.L., and Beddington, R.S. (1988). Multi‐lineage 'stem' cells in the
mammalian embryo. Journal of Cell Science Supplement 10, 11‐27.
Gardner, R.L., and Brook, F.A. (1997). Reflections on the biology of embryonic stem
(ES) cells. The International Journal of Developmental Biology 41, 235‐243.
Gearing, D.P., Thut, C.J., VandeBos, T., Gimpel, S.D., Delaney, P.B., King, J., Price, V.,
Cosman, D., and Beckmann, M.P. (1991). Leukemia inhibitory factor receptor is
structurally related to the IL‐6 signal transducer, gp130. The EMBO Journal 10,
2839‐2848.
Gong, G., Roach, M.L., Jiang, L., Yang, X., and Tian, X.C. (2010). Culture Conditions and
Enzymatic Passaging of Bovine ESC‐Like Cells. Cloning and stem cells.
Greber, B., Wu, G., Bernemann, C., Joo, J.Y., Han, D.W., Ko, K., Tapia, N., Sabour, D.,
Sterneckert, J., Tesar, P., et al. (2010). Conserved and divergent roles of FGF
signaling in mouse epiblast stem cells and human embryonic stem cells. Cell Stem
Cell 6, 215‐226.
Grubb, B.J. (2006). Developmental Biology, Eighth Edition. Scott F. Gilbert, editor.
Integr Comp Biol 46, 652‐653.
Guo, G., Yang, J., Nichols, J., Hall, J.S., Eyres, I., Mansfield, W., and Smith, A. (2009).
Klf4 reverts developmentally programmed restriction of ground state pluripotency.
Development 136, 1063‐1069.
130
Hall, J., Guo, G., Wray, J., Eyres, I., Nichols, J., Grotewold, L., Morfopoulou, S.,
Humphreys, P., Mansfield, W., Walker, R., et al. (2009). Oct4 and LIF/Stat3 Additively
Induce Krüppel Factors to Sustain Embryonic Stem Cell Self‐Renewal. Cell Stem Cell
5, 597‐609.
Hanna, J., Cheng, A.W., Saha, K., Kim, J., Lengner, C.J., Soldner, F., Cassady, J.P., Muffat,
J., Carey, B.W., and Jaenisch, R. (2010). Human embryonic stem cells with biological
and epigenetic characteristics similar to those of mouse ESCs. Proceedings of the
National Academy of Sciences of the United States of America.
Hanna, J., Markoulaki, S., Mitalipova, M., Cheng, A.W., Cassady, J.P., Staerk, J., Carey,
B.W., Lengner, C.J., Foreman, R., Love, J., et al. (2009). Metastable Pluripotent States
in NOD‐Mouse‐Derived ESCs. Cell Stem Cell 5, 124.
Hansis, C., Tang, Y.X., Grifo, J.A., and Krey, L.C. (2001). Analysis of Oct‐4 expression
and ploidy in individual human blastomeres. Molecular human reproduction 7, 155.
Heim, M., Kerr, I., Stark, G., and Darnell, J. (1995). Contribution of STAT SH2 groups
to specific interferon signaling by the Jak‐STAT pathway. Science 267, 1347‐1349.
Heinrich, P.C., Behrmann, I., Haan, S., Hermanns, H.M., Müller‐Newen, G., and
Schaper, F. (2003). Principles of interleukin (IL)‐6‐type cytokine signalling and its
regulation. The Biochemical Journal 374, 1‐20.
Hemmann, U., Gerhartz, C., Heesel, B., Sasse, J., Kurapkat, G., Grötzinger, J., Wollmer,
A., Zhong, Z., Jr., J.E.D., Graeve, L., et al. (1996). Differential Activation of Acute Phase
Response Factor/Stat3 and Stat1 via the Cytoplasmic Domain of the Interleukin 6
Signal Transducer gp130. Journal of Biological Chemistry 271, 12999‐13007.
Illmensee, K., and Mintz, B. (1976). Totipotency and normal differentiation of single
teratocarcinoma cells cloned by injection into blastocysts. Proceedings of the
National Academy of Sciences of the United States of America 73, 549‐553.
Jiang, J., Chan, Y.‐S., Loh, Y.‐H., Cai, J., Tong, G.‐Q., Lim, C.‐A., Robson, P., Zhong, S., and
Ng, H.‐H. (2008). A core Klf circuitry regulates self‐renewal of embryonic stem cells.
Nat Cell Biol 10, 353‐360.
131
Kaczynski, J., Cook, T., and Urrutia, R. (2003). Sp1‐ and Kruppel‐like transcription
factors. Genome Biology 4, 206.
Kawase, E., Suemori, H., Takahashi, N., Okazaki, K., Hashimoto, K., and Nakatsuji, N.
(1994). Strain difference in establishment of mouse embryonic stem (ES) cell lines.
The International Journal of Developmental Biology 38, 385‐390.
Kiuchi, N., Nakajima, K., Ichiba, M., Fukada, T., Narimatsu, M., Mizuno, K., Hibi, M., and
Hirano, T. (1999). STAT3 Is Required for the gp130‐mediated Full Activation of the
c‐myc Gene. J Exp Med 189, 63‐73.
Kleinsmith, L.J., and Pierce, G.B. (1964). Multipotentiality of Single Embryonal
Carcinoma Cells. Cancer Res 24, 1544‐1551.
Kunath, T., Saba‐El‐Leil, M.K., Almousailleakh, M., Wray, J., Meloche, S., and Smith, A.
(2007). FGF stimulation of the Erk1/2 signalling cascade triggers transition of
pluripotent embryonic stem cells from self‐renewal to lineage commitment.
Development 134, 2895‐2902.
Kuo, C.T., Veselits, M.L., and Leiden, J.M. (1997). LKLF: A transcriptional regulator of
single‐positive T cell quiescence and survival. Science (New York, NY) 277, 1986‐
1990.
Kusaba, T., Nakayama, T., Yamazumi, K., Yakata, Y., Yoshizaki, A., Nagayasu, T., and
Sekine, I. (2005). Expression of p‐STAT3 in human colorectal adenocarcinoma and
adenoma; correlation with clinicopathological factors. Journal of Clinical Pathology
58, 833‐838.
Lanza, R.P., Gearhart, J., and Hogan, B. (2006). Essentials of stem cell biology
(Academic Press).
Lee, M.K., Hande, M.P., and Sabapathy, K. (2005). Ectopic mTERT expression in
mouse embryonic stem cells does not affect differentiation but confers resistance to
differentiation‐ and stress‐induced p53‐dependent apoptosis. J Cell Sci 118, 819‐
829.
Li, M., Sendtner, M., and Smith, A. (1995). Essential function of LIF receptor in motor
neurons. Nature 378, 724‐727.
132
Li, P., Tong, C., Mehrian‐Shai, R., Jia, L., Wu, N., Yan, Y., Maxson, R.E., Schulze, E.N.,
Song, H., Hsieh, C.‐L., et al. (2008). Germline competent embryonic stem cells
derived from rat blastocysts. Cell 135, 1299‐1310.
Li, Y., McClintick, J., Zhong, L., Edenberg, H.J., Yoder, M.C., and Chan, R.J. (2005).
Murine embryonic stem cell differentiation is promoted by SOCS‐3 and inhibited by
the zinc finger transcription factor Klf4. Blood 105, 635.
Lim, J.W.E., and Bodnar, A. (2002). Proteome analysis of conditioned medium from
mouse embryonic fibroblast feeder layers which support the growth of human
embryonic stem cells. PROTEOMICS 2, 1187‐1203.
Ling, X., and Arlinghaus, R.B. (2005). Knockdown of STAT3 Expression by RNA
Interference Inhibits the Induction of Breast Tumors in Immunocompetent Mice.
Cancer Res 65, 2532‐2536.
Ling, X., Konopleva, M., Zeng, Z., Ruvolo, V., Stephens, L.C., Schober, W., McQueen, T.,
Dietrich, M., Madden, T.L., and Andreeff, M. (2007). The Novel Triterpenoid C‐28
Methyl Ester of 2‐Cyano‐3, 12‐Dioxoolen‐1, 9‐Dien‐28‐Oic Acid Inhibits Metastatic
Murine Breast Tumor Growth through Inactivation of STAT3 Signaling. Cancer Res
67, 4210‐4218.
Lu, C.C., Brennan, J., and Robertson, E.J. (2001). From fertilization to gastrulation:
axis formation in the mouse embryo. Current Opinion in Genetics & Development
11, 384‐392.
Marin, M., Karis, A., Visser, P., Grosveld, F., and Philipsen, S. (1997). Transcription
factor Sp1 is essential for early embryonic development but dispensable for cell
growth and differentiation. Cell 89, 619‐628.
Martin, G.R. (1980). Teratocarcinomas and mammalian embryogenesis. Science
(New York, NY) 209, 768‐776.
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
USA 78, 7634–7638.
133
Matsuda, T., Nakamura, T., Nakao, K., Arai, T., Katsuki, M., Heike, T., and Yokota, T.
(1999). STAT3 activation is sufficient to maintain an undifferentiated state of mouse
embryonic stem cells. The EMBO Journal 18, 4261.
Mead, R.A. (1993). Embryonic diapause in vertebrates. Journal of Experimental
Zoology 266, 629‐641.
Miller, J.R., Hocking, A.M., Brown, J.D., and Moon, R.T. (1999). Mechanism and
function of signal transduction by the Wnt/beta‐catenin and Wnt/Ca2+ pathways.
Oncogene 18, 7860‐7872.
Mintz, B., and Illmensee, K. (1975). Normal genetically mosaic mice produced from
malignant teratocarcinoma cells. Proceedings of the National Academy of Sciences of
the United States of America 72, 3585‐3589.
Miyabayashi, T., Teo, J.‐L., Yamamoto, M., McMillan, M., Nguyen, C., and Kahn, M.
(2007). Wnt/β‐catenin/CBP signaling maintains long‐term murine embryonic stem
cell pluripotency. Proceedings of the National Academy of Sciences 104, 5668‐5673.
Murakami, M., Hibi, M., Nakagawa, N., Nakagawa, T., Yasukawa, K., Yamanishi, K.,
Taga, T., and Kishimoto, T. (1993). IL‐6‐induced homodimerization of gp130 and
associated activation of a tyrosine kinase. Science 260, 1808‐1810.
Nagy, A., Gertsenstein, M., Vintersten, K., and Behringer, R. (2003). Manipulating the
mouse embryo: a laboratory manual (Cold Spring Harbor Laboratory Pr).
Nakagawa, M., Koyanagi, M., Tanabe, K., Takahashi, K., Ichisaka, T., Aoi, T., Okita, K.,
Mochiduki, Y., Takizawa, N., and Yamanaka, S. (2008). Generation of induced
pluripotent stem cells without Myc from mouse and human fibroblasts. Nature
biotechnology 26, 101‐106.
Narazaki, M., Witthuhn, B.A., Yoshida, K., Silvennoinen, O., Yasukawa, K., Ihle, J.N.,
Kishimoto, T., and Taga, T. (1994). Activation of JAK2 kinase mediated by the
interleukin 6 signal transducer gp130. Proceedings of the National Academy of
Sciences of the United States of America 91, 2285‐2289.
134
Nichols, J., Chambers, I., Taga, T., and Smith, A. (2001). Physiological rationale for
responsiveness of mouse embryonic stem cells to gp130 cytokines. Development
128, 2333‐2339.
Nichols, J., Evans, E.P., and Smith, A.G. (1990). Establishment of germ‐line‐competent
embryonic stem (ES) cells using differentiation inhibiting activity. Development
(Cambridge, England) 110, 1341‐1348.
Nichols, J., and Smith, A. (2009). Naive and Primed Pluripotent States. Cell Stem Cell
4, 487‐492.
Niida, H., Shinkai, Y., Hande, M.P., Matsumoto, T., Takehara, S., Tachibana, M.,
Oshimura, M., Lansdorp, P.M., and Furuichi, Y. (2000). Telomere Maintenance in
Telomerase‐Deficient Mouse Embryonic Stem Cells: Characterization of an
Amplified Telomeric DNA. Mol Cell Biol 20, 4115‐4127.
Niwa, H. (2001). Molecular mechanism to maintain stem cell renewal of ES cells. Cell
structure and function 26, 137–148.
Niwa, H., Burdon, T., Chambers, I., and Smith, A. (1998a). Self‐renewal of pluripotent
embryonic stem cells is mediated via activation of STAT3. Genes & Development 12,
2048‐2060.
Niwa, H., Burdon, T., Chambers, I., and Smith, A. (1998b). Self‐renewal of pluripotent
embryonic stem cells is mediated via activation of STAT3. Genes & development 12,
2048.
Niwa, H., Miyazaki, J., and Smith, A.G. (2000). Quantitative expression of Oct‐3/4
defines differentiation, dedifferentiation or self‐renewal of ES cells. Nature genetics
24, 372–376.
Niwa, H., Ogawa, K., Shimosato, D., and Adachi, K. (2009). A parallel circuit of LIF
signalling pathways maintains pluripotency of mouse ES cells. Nature 460, 118–122.
Pagès, G., Guérin, S., Grall, D., Bonino, F., Smith, A., Anjuere, F., Auberger, P., and
Pouysségur, J. (1999). Defective thymocyte maturation in p44 MAP kinase (Erk 1)
knockout mice. Science (New York, NY) 286, 1374‐1377.
135
Paling, N.R.D., Wheadon, H., Bone, H.K., and Welham, M.J. (2004). Regulation of
Embryonic Stem Cell Self‐renewal by Phosphoinositide 3‐Kinase‐dependent
Signaling. Journal of Biological Chemistry 279, 48063‐48070.
Piotrowska‐Nitsche, K., Perea‐Gomez, A., Haraguchi, S., and Zernicka‐Goetz, M.
(2005). Four‐cell stage mouse blastomeres have different developmental properties.
Development 132, 479‐490.
Rathjen, P.D., Toth, S., Willis, A., Heath, J.K., and Smith, A.G. (1990). Differentiation
inhibiting activity is produced in matrix‐associated and diffusible forms that are
generated by alternate promoter usage. Cell 62, 1105‐1114.
Renfree, M.B., and Shaw, G. (2000). Diapause. Annual Review of Physiology 62, 353‐
375.
Sasaki, A., Yasukawa, H., Suzuki, A., Kamizono, S., Syoda, T., Kinjyo, I., Sasaki, M.,
Johnston, J.A., and Yoshimura, A. (1999). Cytokine‐inducible SH2 protein‐3
(CIS3/SOCS3) inhibits Janus tyrosine kinase by binding through the N‐terminal
kinase inhibitory region as well as SH2 domain. Genes to Cells: Devoted to Molecular
& Cellular Mechanisms 4, 339‐351.
Sato, N., Meijer, L., Skaltsounis, L., Greengard, P., and Brivanlou, A.H. (2004).
Maintenance of pluripotency in human and mouse embryonic stem cells through
activation of Wnt signaling by a pharmacological GSK‐3‐specific inhibitor. Nat Med
10, 55‐63.
Savatier, P., Lapillonne, H., van Grunsven, L.A., Rudkin, B.B., and Samarut, J. (1996).
Withdrawal of differentiation inhibitory activity/leukemia inhibitory factor up‐
regulates D‐type cyclins and cyclin‐dependent kinase inhibitors in mouse embryonic
stem cells. Oncogene 12, 309‐322.
Schöck, F., Purnell, B.A., Wimmer, E.A., and Jäckle, H. (1999). Common and diverged
functions of the Drosophila gene pair D‐Sp1 and buttonhead. Mechanisms of
Development 89, 125‐132.
Selcher, J.C., Nekrasova, T., Paylor, R., Landreth, G.E., and Sweatt, J.D. (2001). Mice
Lacking the ERK1 Isoform of MAP Kinase Are Unimpaired in Emotional Learning.
Learning & Memory 8, 11‐19.
136
Singh, A.M., and Dalton, S. (2009). The Cell Cycle and Myc Intersect with Mechanisms
that Regulate Pluripotency and Reprogramming. Cell Stem Cell 5, 141‐149.
Smith, A.G. (2001). Embryo‐derived stem cells: of mice and men. Annual Review of
Cell and Developmental Biology 17, 435‐462.
Smith, A.G., and Hooper, M.L. (1987). Buffalo rat liver cells produce a diffusible
activity which inhibits the differentiation of murine embryonal carcinoma and
embryonic stem cells. Developmental Biology 121, 1‐9.
Solter, D., and Knowles, B.B. (1978). Monoclonal antibody defining a stage‐specific
mouse embryonic antigen (SSEA‐1). Proceedings of the National Academy of
Sciences of the United States of America 75, 5565‐5569.
Stahl, N., Boulton, T., Farruggella, T., Ip, N., Davis, S., Witthuhn, B., Quelle, F.,
Silvennoinen, O., Barbieri, G., Pellegrini, S., et al. (1994). Association and activation
of Jak‐Tyk kinases by CNTF‐LIF‐OSM‐IL‐6 beta receptor components. Science 263,
92‐95.
Stewart, C.L., Kaspar, P., Brunet, L.J., Bhatt, H., Gadi, I., Köntgen, F., and Abbondanzo,
S.J. (1992). Blastocyst implantation depends on maternal expression of leukaemia
inhibitory factor. Nature 359, 76‐79.
Strickland, S., and Mahdavi, V. (1978). The induction of differentiation in
teratocarcinoma stem cells by retinoic acid. Cell 15, 393‐403.
Sun, H., Lesche, R., Li, D.‐M., Liliental, J., Zhang, H., Gao, J., Gavrilova, N., Mueller, B.,
Liu, X., and Wu, H. (1999). PTEN modulates cell cycle progression and cell survival
by regulating phosphatidylinositol 3,4,5,‐trisphosphate and Akt/protein kinase B
signaling pathway. Proceedings of the National Academy of Sciences of the United
States of America 96, 6199‐6204.
Surani, M.A. (2001). Reprogramming of genome function through epigenetic
inheritance. Nature 414, 122‐128.
137
Suwińska, A., Czołowska, R., Ozdzeński, W., and Tarkowski, A.K. (2008). Blastomeres
of the mouse embryo lose totipotency after the fifth cleavage division: expression of
Cdx2 and Oct4 and developmental potential of inner and outer blastomeres of 16‐
and 32‐cell embryos. Developmental Biology 322, 133‐144.
Symes, A., Stahl, N., Reeves, S.A., Farruggella, T., Servidei, T., Gearan, T., Yancopoulos,
G., and Fink, J.S. (1997). The protein tyrosine phosphatase SHP‐2 negatively
regulates ciliary neurotrophic factor induction of gene expression. Current Biology
7, 697‐700.
Takahashi, K., and Yamanaka, S. (2006). Induction of Pluripotent Stem Cells from
Mouse Embryonic and Adult Fibroblast Cultures by Defined Factors. Cell 126, 663‐
676.
Tesar, P.J., Chenoweth, J.G., Brook, F.A., Davies, T.J., Evans, E.P., Mack, D.L., Gardner,
R.L., and McKay, R.D.G. (2007). New cell lines from mouse epiblast share defining
features with human embryonic stem cells. Nature 448, 196‐199.
Thomas, K.R., and Capecchi, M.R. (1987). Site‐directed mutagenesis by gene
targeting in mouse embryo‐derived stem cells. Cell 51, 503‐512.
Till, J.E., McCulloch, E.A., and Siminovitch, L. A stochastic model of stem cell
proliferation, based on the growth of spleen colony‐forming cells.
Trainer, D.L., Kline, T., McCabe, F.L., Faucette, L.F., Feild, J., Chaikin, M., Anzano, M.,
Rieman, D., Hoffstein, S., and Li, D.J. (1988). Biological characterization and
oncogene expression in human colorectal carcinoma cell lines. International Journal
of Cancer Journal International Du Cancer 41, 287‐296.
Villalva, C., Martin‐Lannerée, S., Cortes, U., Dkhissi, F., Wager, M., Le Corf, A., Tourani,
J., Dusanter‐Fourt, I., Turhan, A., and Karayan‐Tapon, L. (2010). STAT3 is essential
for the maintenance of neurosphere‐initiating tumor cells in patients with
glioblastomas : A potential for targeted therapy ? International Journal of Cancer
Journal International Du Cancer.
138
Ware, C.B., Horowitz, M.C., Renshaw, B.R., Hunt, J.S., Liggitt, D., Koblar, S.A., Gliniak,
B.C., McKenna, H.J., Papayannopoulou, T., and Thoma, B. (1995). Targeted disruption
of the low‐affinity leukemia inhibitory factor receptor gene causes placental,
skeletal, neural and metabolic defects and results in perinatal death. Development
(Cambridge, England) 121, 1283‐1299.
Welte, Y., Adjaye, J., Lehrach, H.R., and Regenbrecht, C.R. (2010). Cancer stem cells in
solid tumors: elusive or illusive? Cell Communication and Signaling: CCS 8, 6.
Wen, Z., Zhong, Z., and Darnell Jr, J.E. (1995). Maximal activation of transcription by
Stat1 and Stat3 requires both tyrosine and serine phosphorylation. Cell 82, 241–
250.
Yang, J., Chatterjee‐Kishore, M., Staugaitis, S.M., Nguyen, H., Schlessinger, K., Levy,
D.E., and Stark, G.R. (2005). Novel Roles of Unphosphorylated STAT3 in Oncogenesis
and Transcriptional Regulation. Cancer Res 65, 939‐947.
Yao, Y., Li, W., Wu, J., Germann, U.A., Su, M.S.S., Kuida, K., and Boucher, D.M. (2003).
Extracellular signal‐regulated kinase 2 is necessary for mesoderm differentiation.
Proceedings of the National Academy of Sciences of the United States of America
100, 12759‐12764.
Ying, Q.‐L., Stavridis, M., Griffiths, D., Li, M., and Smith, A. (2003). Conversion of
embryonic stem cells into neuroectodermal precursors in adherent monoculture.
Nat Biotech 21, 183‐186.
Ying, Q.‐L., Wray, J., Nichols, J., Batlle‐Morera, L., Doble, B., Woodgett, J., Cohen, P.,
and Smith, A. (2008a). The ground state of embryonic stem cell self‐renewal. Nature
453, 519‐523.
Ying, Q.L., Wray, J., Nichols, J., Batlle‐Morera, L., Doble, B., Woodgett, J., Cohen, P., and
Smith, A. (2008b). The ground state of embryonic stem cell self‐renewal. Nature 453,
519–523.
Yokogami, K., Wakisaka, S., Avruch, J., and Reeves, S.A. (2000). Serine
phosphorylation and maximal activation of STAT3 during CNTF signaling is
mediated by the rapamycin target mTOR. Current Biology 10, 47–50.
139
Zhong, Z., Wen, Z., and Darnell, J. (1994a). Stat3: a STAT family member activated by
tyrosine phosphorylation in response to epidermal growth factor and interleukin‐6.
Science 264, 95‐98.
Zhong, Z., Wen, Z., and Darnell, J.E. (1994b). Stat3 and Stat4: members of the family
of signal transducers and activators of transcription. Proceedings of the National
Academy of Sciences of the United States of America 91, 4806‐4810.
140
Appendix A
Materials and Methods
Cell Culture
C57BL/6, 46C, and Balb/c1 mouse ES cells were routinely cultured in GMEM
(Sigma) supplemented with 10% FBS (HyClone) and 10ng/ml human LIF (Sigma)
on gelatinized culture flasks. In the case of the C57BL/6 and Balb/c1 mouse ES cells,
the flasks were first pre‐coated with gamma‐irradiated mitotically‐inactive CF‐1
mouse embryonic fibroblasts (MEFs) at a density of 2.1 x 10
4
cells/cm
2
. DAc8 rat ES
cells were routinely cultured in serum‐free N2B27 medium (previously described in
Nichols and Ying, 2006) supplemented with 3 uM CHIR99201 and 1 uM PD0325901
(both from Stem Cell Sciences). Rat ES cell culture flasks were gelatinized and pre‐
coated with gamma‐irradiated CF‐1 MEFs at a density of 2.1 x 10
4
cells/cm
2
. Stat3
null mouse ES cells were derived and maintained as described previously in Ying et.
al., 2008.
Plasmid Construction
To over express Klfs genes in ES cells, retrovirus based gene expression vector,
pMIG, was used. The coding region of KLF4 gene was amplified by RT‐PCR from
cDNA of ES cells. Klf2 and Klf5 were kindly gifted by Shinya Yamanaka. The 5’BglII
141
and 3’XhoI sites were introduced into the PCR product and then subcloned into
BglII/XhoI site in pMIG.
For RNA interference, a lentivirus mediated system was used according to the
standard protocol. Briefly, two pair of sense and antisense oligonucleotides of
shRNA were synthesized. After annealing, the duplexes were cloned into AgeI/EcoRI
sites of pLKO.1. The target sequences are as follows: klf4‐shRNA1: 5’ ‐
GCGCTACAATCATGGTCAAGT;
klf4‐shRNA2: 5’ ‐ GTCAGCTTGTGAATGGATAAT.
Virus production and infection of ES cells
Retroviruses or lentiviruses were produced by transfection of the transducing
vector into 293T cells with packaging vectors through Lipofectamine
2000(Invitrogen), The supernatant containing secreted viral particles was
harvested at 48 and 72 hours after transfection and filtered through 0.45um low‐
protein binding syringe filter, and used to infect ES cells.
For virus transduction, ES cells were trypsinized and re‐suspended in growth media
at concentration 5 x 10
5
/ml. Then 1.5 ml cells were mixed with 0.5 ml viral
supernatants and plated onto gelatinized 3.5 cm dishes. Polybrene was added at a
final concentration 8 ug/ml. Cells were incubated with virus overnight and then fed
with fresh media. Two days after infection, drug selection with puromycin (2
142
ug/ml,Sigma) was performed for 1 week. Puromycin‐resistant ES cell pools were
collected and maintained under drug‐selection conditions.
Transfection of Mouse and Rat ES Cells
STAT3 overexpression clones were created by inserting the full‐length stat3
transgene, acquired from Addgene, into the pPyFloxedMTIPgfp vector (previously
described in Ying et al., 2008). 20 ug of linearized pPyFloxedMTSTAT3IPgfp
plasmid was electroporated into 2 x 10
6
mouse or rat ES cells using 500 uF and 280
V on a BioRad GenePulser XCell apparatus. The cells were then cultured with DR4
MEFs for two days before the transfectants were selected using 1 ug/ml puromycin
for 5‐7 days. Individual stably transfected colonies were picked and expanded.
STAT3 transgene excision was mediated by transient Lipofectamine LTX
(Invitrogen) transfection of pCAGCreIP plasmids into STAT3 overexpressing
C57BL/6 mouse ES cells. The resulting GFP positive colonies were picked,
dissociated, re‐picked, and expanded to obtain a pure population of cells. Stat3 null
mouse ES cells were transfected with the STAT3 expression vector as above, but
instead the cells were cultured in serum‐free N2B27 medium supplemented with 3
uM CHIR99201 and 1 uM PD0325901 in co‐culture with DR4 MEFs prior and
following transfection. Creation of Y118F clones were obtained in a similar manner,
except transfection of the GCSFRgp130Y118F expression vector was linearized and
stably introduced instead. Drug selection was enforced using 20 ug/ml Zeocin
(Invitrogen) for 7‐10 days.
143
In Vitro Differentiation
Mouse ES cells were differentiated by induction of embryoid body formation. Mouse
ES cells were plated onto uncoated bacterial grade petri dishes in MEF medium
(GMEM, 10% FBS, 1 mM L‐Glutamine). Cells would form aggregates in suspension
which would expand into embryoid bodies. The embryoid bodies would then be
plated intact upon a gelatinzed cell culture dish in GMEM/10% FBS medium after 7‐
10 days. The embryoid bodies were allowed to continue differentiation for another
5‐7 days before subsequent analysis was performed.
Immunostaining and AP Staining
Immunostaining was performed using conventional methods. Primary antibodies
were used in the following concentrations: Oct4 (C‐10, Santa Cruz, 1:500), Sox2 (Y‐
17, Santa Cruz, 1:1000), Gata‐4 (G‐4, Santa Cruz, 1:200), βIII‐Tubulin (Sigma, 1:200),
and muscle sarcomere (Developmental Studies Hybridoma Bank, MF‐20, 1:3).
Secondary antibodies were all purchased from Alexa Fluor (Invitrogen) and
employed at a 1:2000 dilution. Nuclei were visualized via DAPI staining. Alkaline
phosphatase staining was performed as per manufacturer’s instructions (Sigma).
144
RTPCR and Quantitative PCR
Total RNA was collected and purified using RNA Easy Mini Kit (Qiagen). cDNA was
prepared by reverse transcribing 2 ug total RNA using the Cloned AMV First‐Strand
cDNA Synthesis Kit (Invitrogen) using oligo dT primers and following the
manufacturer’s instructions. PCR reactions were prepared using 1/20 of the reverse
transcription reaction as template and amplified using Taq polymerase (Invitrogen).
qPCR was performed by using SYBR Green PCR Master Mix (Applied Biosystems)
according to the manufacturer’s instructions. Amplicons were detected by the
ABI7900HT Real‐Time PCR System (Applied Biosystems) and analyzed using RQ
Manager v1.2 (Applied Biosystems). Relative expression was determined by the
ΔΔCt method and normalized to the GAPDH expression. Unless otherwise noted, all
reactions were performed in technical triplicate and representative reactions from
biological duplicates were presented.
Western Blotting
Western blotting was performed using conventional methods. In general, cells were
serum starved for 6‐12 hours prior to stimulation. Cells were then stimulated with
either 10 ng/ml human LIF (Sigma) or 0.2 ng/ml human G‐CSF (Peprotech) in the
presence in serum‐free basal medium (GMEM, 1 mM L‐Glutamine) for the amount of
time specified in the assay. PD0325901 was used at 1 uM concentration. Cells were
145
lysed in RIPA buffer. Total cell lysates were compared under denaturing conditions
and probed using the following primary antibodies: Anti‐STAT3 (BD, 1:1000), anti‐
phosphoY705‐STAT3 (Cell Signaling, 1:1000), anti‐ERK1/2 (K‐23, Santa Cruz,
1:1000), anti‐phosphoY204‐ERK1/2 (E‐4, Santa Cruz 1:1000), and anti‐α‐tubulin
(Sigma, 1:2000), anti‐Klf4 (R&D, AF3158, 1:1000), anti‐β‐actin (Sigma, A5441,
1:2000).
Densiometry
Digitized western blot films were analyzed using ImageJ software available through
the National Institutes of Health website. All images were quantified by normalizing
the density of the signal to tubulin expression unless otherwise noted. Data was
compared to a reference signal to derive the relative intensities reported as
Densiometric Units. Scale is linear unless otherwise noted.
146
Appendix B:
Primer Set List
Primer Name Sequence (5' - 3')
Species
Recognized
Klf4 Forward CTGAACAGCAGGGACTGTCA Mouse, Rat
Klf4 Reverse GTGTGGGTGGCTGTTCTTTT Mouse, Rat
Klf4 Forward qPCR CGGCTGTGGGTGGAAATTC Rat
Klf4 Reverse qPCR CCGGTGTGTTTGCGGTAGT Rat
Klf2 Forward qPCR CAACTGCGGCAAGACCTACA Rat
Klf2 Reverse qPCR CAATGATAAGGCTTCTCACCTGTGT Rat
Klf5 Forward qPCR CCACCTACTTTCCCCCATCA Rat
Klf5 Reverse qPCR CTGGAGCATCTCAGCTTGTCTATC Rat
Tbx3 Forward qPCR TCCGGCTCAGTGTCCTTGTC Rat
Tbx3 Reverse qPCR TGCTCTGCAGTTCGCTGGTA Rat
STAT3 Forward qPCR CCTTACTGGGCCTAGGGTCAAC Rat
STAT3 Reverse qPCR GGACATGGGAAGGAGACATACC Rat
FGF4 Forward GGGAGGCTACAGACAGCAAG Mouse
FGF4 Reverse CTGTGAGCCACCAGACAGAA Mouse
FGF5 Forward GCGACGTTTTCTTCGTCTTC Mouse
FGF5 Reverse ACAATCCCCTGAGACACAGC Mouse
HPRT Forward CTCGAAGTGTTGGATACAGG Mouse, Rat
HPRT Reverse TGGCCTATAGGCTCATAGTG Mouse, Rat
Acta2 Forward GGGAGTAATGGTTGGAAT Mouse
Acta2 Reverse TCAAACATAATCTGGGTCA Mouse
AFP Forward CTGGCGATGGGTGTTTAG Mouse
AFP Reverse CCTGGAGGTTTCGGGATT Mouse
Brachyury Forward CTTTCTTGCTGGACTTCG Mouse
Brachyury Reverse TTACATCTTTGTGGTCGTTT Mouse
Cdx2 Forward CTTCCTGCCAGCAACGAC Mouse
Cdx2 Reverse CAGACATACATTCCGCCTACA Mouse
Gata4 Forward CCGAGGGTGAGCCTGTAT Mouse
Gata4 Reverse GCCTGCGATGTCTGAGTG Mouse
Hand1 Forward GGCGAGAAGAGGATTAAAGG Mouse
Hand1 Reverse CGAGAAGGCATCAGGGTAC Mouse
Nestin Forward GTCTGATGGGTTTGCTGA Mouse
Nestin Reverse AATCGCTTGACCTTCCTC Mouse
Plat Forward TCTGCCAGTGCCCTGATG Mouse
Plat Reverse TGGGTGCCACGGTAAGTC Mouse
Sparc Forward GGTGGTTTGGAGTTAGGC Mouse
Sparc Reverse GTAGAAGGTTTCAAGTGGC Mouse
Tuj1 Forward ACGCATCTCGGAGCAGTT Mouse
Tuj1 Reverse CGATTCCTCGTCATCATCTTC Mouse
Oct4 Forward GAAGCAGAAGAGGATCACCTTG Mouse
Oct4 Reverse TTCTTAAGGCTGAGCTGCAAG Mouse
Abstract (if available)
Abstract
Pluripotent embryonic stem (‘ES’) cells are typically derived and maintained using inductive or inhibitory signals that are thought to behave in a binary ‘on/off’ manner. Mouse ES cells are maintained in an excess of leukemia inhibitory factor (LIF) and rat ES cells in a cocktail of inhibitors that block both GSK3β and MAPK signaling in a STAT3-independent manner. Here, we provide evidence that both mouse and rat ES cell self-renewal is conserved via a STAT3-dependent mechanism that treats STAT3 activation as fluid and dynamic. We observe that increased Klf4 expression enhances STAT3-mediated self-renewal in mouse ES cells but is not essential to prevent differentiation. As well, increased STAT3 activation and ERK1/2 inhibition synergistically enhance self-renewal. In all, we propose that the self-renewal mechanism is a nested metastable cell-state within naïve pluripotency, and that further refinements to the self-renewal mechanism will allow for direct inter-species comparisons and derivation of novel ES and induced pluripotent cell population.
Linked assets
University of Southern California Dissertations and Theses
Conceptually similar
PDF
Pleotropic potential of Stat3 in determining self-renewal, apoptosis, and differentiation in mouse embryonic stem cells
PDF
Role of STAT3 phosphorylation in mouse embryonic stem cell self-renewal and differentiation
PDF
Role of beta-catenin in mouse epiblast stem cell, embryonic stem cell self-renewal and differentiation
PDF
Roles of Klf4 in embryonic stem cells
PDF
The role of ERK1/2 in mouse embryonic stem cell fate control
PDF
Molecular basis of mouse epiblast stem cell and human embryonic stem cell self‐renewal
PDF
Hepatic differentiation in human naïve stem cells compared to human embryonic stem cells
PDF
Novel roles for Maf1 in embryonic stem cell differentiation and adipogenesis
PDF
Derivation and characterization of human embryonic stem (hES) cells and human induced pluripotent stem (hiPS) cells in clinical grade conditions
PDF
Leucine-rich amelogenin peptide induces osteogenesis in mouse embryonic stem cells
PDF
Elucidating the role of neural crest specific Stat3 signaling in maintaining coronal suture patency during embryonic development
PDF
The role of neuregulin receptors in cell differentiation and the response to inflammatory cytokines in the intestinal epithelium
PDF
Interaction of epigenetics and SMAD signaling in stem cells and diseases
PDF
The function of BS69 in mouse embryogenesis and embryonic stem cell differentiation
PDF
Harnessing the power of stem cell self-renewal pathways in cancer: dissecting the role of BMI-1 in Ewing’s sarcoma initiation and maintenance
PDF
The role of Prkci in stem cell maintenance and cell polarity using a 3-D culture system
PDF
The role of LGR5 in the pathogenesis of Ewing sarcoma: a marker of aggressive disease and a contributor to the malignant phenotype
PDF
Roles of epithelial-mesenchymal transition and niche in tumorigenesis of tumor-initiating cells
PDF
Polycomb repressive complex 2 subunit stabilizes NANOG to maintain self-renewal in hepatocellular carcinoma tumor-initiating stem-like cells
PDF
Epigenetic plasticity of cultured female human embryonic stem cells and regulation of gene expression and chromatin by PR-SET7 mediated H4K20me1
Asset Metadata
Creator
Schulze, Eric Nathaniel
(author)
Core Title
Investigating the role of STAT3 in mouse and rat embryonic stem cell self-renewal and differentiation
School
Keck School of Medicine
Degree
Doctor of Philosophy
Degree Program
Genetic, Molecular and Cellular Biology
Publication Date
08/11/2010
Defense Date
05/24/2010
Publisher
University of Southern California
(original),
University of Southern California. Libraries
(digital)
Tag
cell,Development,differentiation,embryonic,embryonic stem cell,ERK1/2,ground state pluripotency,GSK3,human,Jak,Klf4,Krüppel-like factor 4,MAPK,metastable pluripotency,mouse,naive pluripotency,OAI-PMH Harvest,pluripotency,pluripotent,rat,self-renewal,Stat3,STEM,stem cell
Language
English
Contributor
Electronically uploaded by the author
(provenance)
Advisor
Ying, Qi-Long (
committee chair
), Adams, Gregor B. (
committee member
), Lutzko, Carolyn (
committee member
), Pera, Martin F. (
committee member
)
Creator Email
eschulze@gmail.com,eschulze@usc.edu
Permanent Link (DOI)
https://doi.org/10.25549/usctheses-m3349
Unique identifier
UC153735
Identifier
etd-Schulze-3501 (filename),usctheses-m40 (legacy collection record id),usctheses-c127-377882 (legacy record id),usctheses-m3349 (legacy record id)
Legacy Identifier
etd-Schulze-3501.pdf
Dmrecord
377882
Document Type
Dissertation
Rights
Schulze, Eric Nathaniel
Type
texts
Source
University of Southern California
(contributing entity),
University of Southern California Dissertations and Theses
(collection)
Repository Name
Libraries, University of Southern California
Repository Location
Los Angeles, California
Repository Email
cisadmin@lib.usc.edu
Tags
cell
differentiation
embryonic
embryonic stem cell
ERK1/2
ground state pluripotency
GSK3
human
Jak
Klf4
Krüppel-like factor 4
MAPK
metastable pluripotency
mouse
naive pluripotency
pluripotency
pluripotent
rat
self-renewal
Stat3
STEM
stem cell