You are on page 1of 14

Phil. Trans. R. Soc.

B (2011) 366, 2208–2221


doi:10.1098/rstb.2011.0006

Review

The evolving biology of small molecules:


controlling cell fate and identity
Jem A. Efe and Sheng Ding*,†
Department of Chemistry, The Scripps Research Institute, 10550 North Torrey Pines Road,
La Jolla, CA 92037, USA
Small molecules have been playing important roles in elucidating basic biology and treatment of a
vast number of diseases for nearly a century, making their use in the field of stem cell biology a com-
paratively recent phenomenon. Nonetheless, the power of biology-oriented chemical design and
Downloaded from https://royalsocietypublishing.org/ on 26 May 2021

synthesis, coupled with significant advances in screening technology, has enabled the discovery of
a growing number of small molecules that have improved our understanding of stem cell biology
and allowed us to manipulate stem cells in unprecedented ways. This review focuses on recent
small molecule studies of (i) the key pathways governing stem cell homeostasis, (ii) the pluripotent
stem cell niche, (iii) the directed differentiation of stem cells, (iv) the biology of adult stem cells, and
(v) somatic cell reprogramming. In a very short period of time, small molecules have defined a per-
haps universally attainable naive ground state of pluripotency, and are facilitating the precise, rapid
and efficient differentiation of stem cells into somatic cell populations relevant to the clinic. Finally,
following the publication of numerous groundbreaking studies at a pace and consistency unusual
for a young field, we are closer than ever to completely eliminating the need for genetic modification
in reprogramming.
Keywords: small molecules; stem cells; signalling; metabolites; reprogramming;
transdifferentiation

1. INTRODUCTION coincidental. Their advantages are compelling: first,


It took much of the second half of the twentieth cen- the current regulatory environment remains more-or-
tury for the monumental discoveries in molecular less aligned with the discovery and development of
biology and genetics to usher in the era of modern small molecules as therapeutics. Second, logistically,
biology. But even as these fields of inquiry were in they are easily manufactured, stored and administered.
their infancy, the design and the synthesis of small- Third, and perhaps most importantly, from a biochemi-
molecule compounds to elicit desired biological cal standpoint their effects are specific, dose-dependent,
outcomes were a decades-old practice [1]. Over the past rapid and reversible. This set of attributes allows for
century, in both academic and industrial settings, very precise temporal and functional control in vivo.
an extraordinarily wide range of compounds—based on Small molecules also offer a distinct advantage in devel-
naturally occurring substances or entirely man-made— opment; chemical synthesis of compounds based on
have been produced and screened for activity in diverse biologically active molecular ‘scaffolds’ is a very effec-
organisms, cell culture systems and molecular pathways. tive means to quickly generate large ‘libraries’ of
Among the earlier and most noteworthy fruits of such potentially effective small molecules [3,4]. However,
efforts were classic small-molecule antibiotics, which since it is impossible to saturate chemical space and
were so effective in curing previously life-threatening compound libraries are very large by necessity, the
infections that they single-handedly and dramatically actual identification of a highly active and specific com-
extended lifespans around the globe. Small-molecule pound remains a herculean task [5]. Compounding this
drugs to treat a myriad of ailments from heart disease to difficulty is the fact that many effective compounds are
depression eventually followed. To this day, the phar- ultimately discovered to be unsuitable for clinical use,
maceutical industry as a whole still displays a strong e.g. because of the toxicity or other dangerous
preference for small molecules over macromolecular off-target effects. Remarkably, over the years, screening
biologics in the drug-development process [2]. technology has advanced to the point that millions
The development and the use of small molecules as of compounds can be rapidly and reliably tested
the therapeutic method of choice are of course not in numerous biological contexts [6]. Such ‘ultra-
high-throughput’ screening and related technological
* Author for correspondence (sheng.ding@gladstone.ucsf.edu).
advances in design and manipulation have allowed

Present address: The Gladstone Institute of Cardiovascular us to overcome truly daunting odds on a fairly
Disease, 1650 Owens Street, San Francisco, CA 94158, USA. regular basis.
One contribution of 15 to a Discussion Meeting Issue ‘What next for However, it is becoming increasingly obvious that
stem cell biology? The evolving biology of cell reprogramming’. effective development of drugs to combat diseases
2208 This journal is q 2011 The Royal Society
Review. Stem cells and small molecules J. A. Efe & S. Ding 2209

with complex biological underpinnings (e.g. many can- be taken: in the hypothesis-driven (i.e. target-based)
cers and ageing-related disorders of the nervous system approach, prior knowledge of signalling pathways
such as Parkinson’s disease) will require a very detailed and their small-molecule modulators is used to impli-
understanding of the molecular pathways involved, cate them in key regulatory areas. Conversely, the
and no single small molecule is likely to represent a discovery-driven (i.e. phenotype-based) approach
‘magic bullet’ in our efforts to cure such diseases. None- assumes no prior knowledge of regulatory pathways,
theless, small molecules as a class will continue to be one and instead uses unbiased high-throughput screening
of the most effective tools in biomedical research. At this of small-molecule libraries to elicit a certain pheno-
juncture, it is clear that the signal transduction field will type. The cellular targets of effective molecules or
continue to use small molecules in the mapping of ‘hits’ can then be determined using follow-up assays,
molecular pathways relevant to the development and such as affinity chromatography. Eventually, a suffi-
progression of multi-factorial diseases. Importantly, ciently large number of hits could conceivably allow
small molecules will probably be key players in the for the extensive annotation of chemical libraries and
quest to develop novel cell-based therapies for rege- the a priori prediction of small-molecule targets and/
nerative medicine, i.e. for repair and/or replacement or activity. Both methods have their own distinct
of diseased tissues. Stem cells are currently at the fore- advantages, and have been quite effective in the past
front of such efforts, and small molecules—thanks in (reviewed in [6]).The following studies highlight
Downloaded from https://royalsocietypublishing.org/ on 26 May 2021

part to a large body of literature detailing their effects how these approaches—individually, in tandem, or in
on cellular signalling pathways—are playing critical combination with other cell-biological and genomics
roles in stem cell derivation, maintenance and manipu- techniques—continue to improve our understanding of
lation. As such, they offer perhaps the best hope for the complex biology of stem cells (figure 1). We identify
expeditious, effective and safe implementation of novel five key categories: (i) elucidating key pathways of stem
stem cell-based technologies in a clinical setting. cell homeostasis, (ii) defining the stem cell niche, (iii)
How exactly will small molecules help us accomplish directed differentiation of stem cells, (iv) harnessing
these goals? Our review will address this question by the potential of adult stem cells, and (v) somatic cell
covering, with an emphasis on the recent advances, a de-differentiation and fate switching (reprogramming).
diverse array of small molecule-driven studies and their
implications for the future. Uniquely positioned at the
intersection of signal transduction, development and 3. DEFINING AND MAINTAINING A PLURIPOTENT
cellular plasticity, these studies offer a glimpse into one ‘GROUND STATE’
of the fastest developing areas of modern biology. Traditionally, mESCs and hESCs have been cultured on
a monolayer of mitotically inactivated mouse embryonic
fibroblasts (MEFs) termed ‘feeders’, with an additional
2. STEM CELLS AND SMALL MOLECULES: requirement for leukaemia inhibitory factor (LIF; for
AN OVERVIEW mESCs) or basic fibroblast growth factor (bFGF or
Embryonic stem cells (ESCs), isolated from the inner FGF2; for hESCs). In the mouse system, the down-
cell mass (ICM) of the preimplantation embryo [7,8], stream effectors of LIF were eventually identified as
are pluripotent: they possess theoretically boundless signal transducer and activator of transcription 3
self-renewal capacity as well as the potential to generate (STAT3) and the pluripotency-associated genes Klf4
nearly all cells of the organism from which they were and c-Myc [12–14]. The critical component in the unde-
derived. Other naturally occurring pluripotent cell fined portion of the serum-containing media was found
types include epiblast stem cells (EpiSCs; derived from to be bone morphogenetic protein 4 (BMP4), which
post-implantation embryos) and germline cells [9,10]. lies upstream of the inhibitor of differentiation(Id)genes
As these pluripotent cells progressively differentiate, [15]. Feeder-conditioned media supplemented with LIF
they give rise to developmentally more restricted multi- can thus inhibit spontaneous differentiation and allow
potent progenitor cell populations that may persist into indefinite self-renewal, but downstream experimental
adulthood. While the existence and/or exact develop- results can be erratic owing to intrinsic variability in
mental potential of these ‘adult stem cells’ is not serum and MEF quality, as well as varied manipulation
equally well documented for all tissues, there is a signifi- techniques. Furthermore, even though a combination
cant body of literature demonstrating their critical roles of purified BMP4 and LIF does support growth of
in tissue homeostasis and regeneration [11]. mESCs in chemically defined media without feeders,
Since the first derivation of ESCs from mice experimental variability in differentiation potential and
(mESCs)—but especially after the successful isolation propensity remains an issue.
of human ESCs (hESCs)—their culture and directed In an effort to eliminate such variability and sim-
differentiation have presented a unique set of chal- plify media requirements for mESC culture, we
lenges. The loss of pluripotency (i.e. spontaneous carried out a phenotype-based screen of 50 000 small
differentiation) and cell death under prolonged culture molecules to identify compounds that could support
conditions, as well as the inability to generate homo- robust mESC culture in the absence of feeders,
geneous populations of differentiated cells, has been serum and LIF. For facile high-content imaging-
frustrating. Isolating and growing tissue-specific multi- based screening, we used a transgenic mESC line
potent progenitors in culture have proved still more expressing green fluorescent protein (GFP) under con-
difficult. trol of the promoter of Oct4, the pluripotency master
In using small molecules to study and address pro- regulator. Using compact, domed colony morphology
blems in stem cell biology, two distinct approaches can as a secondary phenotype, pluripotin (also known as
Phil. Trans. R. Soc. B (2011)
2210 J. A. Efe & S. Ding Review. Stem cells and small molecules

R3
(a) X
R5 N
follow-up N
optimization X R1
assays R4
N N X
compound
collection R2
?
R3 ?
X
?
R5 high-throughput target/mechanism
N N
X R1 screening determination
R4
N N X
R2
desired phenotypic
known readout and ID of hits
target/pathway
R3 transcription
(b) X
Downloaded from https://royalsocietypublishing.org/ on 26 May 2021

R5 evaluate synergy with other


N N
X R1 R4 compounds and/or cytokines
N N X

R2
test effect on
stem cells
complex
phenotypic
response

transcription
Figure 1. (a) Depicts the outline of a phenotypic screen using small molecules, whereas (b) illustrates the target-based
approach. Their combined application in stem cell biology has proved very useful, as there is often a need to elicit, and
eventually understand the molecular underpinnings of complex phenotypes.

SC1) was identified as a potent and specific small kinase 3 (GSK3) also supports long-term mESC propa-
molecule supporting mESC expansion in the undiffer- gation [17]. By using LIF and BMP4, which negatively
entiated state [16]. Cells cultured without feeders, regulate each other’s differentiation-inducing activities,
serum and cytokines in pluripotin-supplemented traditional culture methods appear to accomplish the
chemically defined media maintained homogeneous same; however, such continuous input can have the
self-renewal and retained their ability for in vivo undesirable side effect of activating certain differen-
germline transmission over many passages, a standard tiation-inducing mechanisms at detectable levels (e.g.
requirement for the demonstration of pluripotency. the ERK pathway mediates ectoderm induction, while
Using affinity chromatography experiments, the targets BMP signalling mainly induces mesoderm) [18–20].
of pluripotin were identified as extracellular-signal- These secondary effects, in turn, lead to high exper-
regulated kinase 1 (ERK1) and Ras GTPase activating imental variability by creating a heterogeneous and
protein (RasGAP), both of which are known positive unstable culture of cells possessing different, and
regulators of differentiation. continuously fluctuating, degrees of pluripotency. The
These findings have two very important implications: more stable and homogeneous nature of cultures main-
first, from a technical standpoint, they demonstrate that tained in the presence of inhibitors indicates that the
a phenotypic screen can identify a single small molecule core pluripotency network is inherently stable, as long
capable of eliciting a very complex response/pheno- as it is not perturbed by external signalling.
type—in this case, maintenance of pluripotency, by Most recently, Wagner et al. [21] used a combination
virtue of pluripotin having specific and synergistic poly- of pharmacological and genetic methods to implicate
pharmacological activity. This is a remarkable feat, and liver receptor homolog-1 (Lrh-1) as a novel b-catenin
one that bodes well for future such efforts. Second, in target gene required for maintaining proper levels of
terms of the underlying biology, it is now obvious that Oct4 and Nanog in mESCs. They have postulated
inhibition of targets/mechanisms promoting differen- that this signalling axis might represent the in vivo
tiation is sufficient to keep mESCs in a pluripotent counterpart of the LIF – STAT3 pathway (which is
‘ground state’. This finding has been corroborated only required in vitro) [21].
by others’ subsequent demonstration that concurrent In the interim, our improved understanding of the
inhibition of the FGF4–ERK1/2 mitogen-activated pluripotent ground state has been successfully applied
protein kinase (MAPK) pathway and glycogen synthase to the derivation of ESCs from mouse strains that had

Phil. Trans. R. Soc. B (2011)


Review. Stem cells and small molecules J. A. Efe & S. Ding 2211

hitherto proven refractory to ESC isolation. We have e.g. by trypsinization, Rho – ROCK signalling is upre-
shown that germline-competent ESCs can be derived gulated, resulting in destabilization of E-cadherin-
from non-obese diabetic (NOD) mice through com- mediated cell – cell adhesion. The loss of cell –cell
bined treatment with LIF and pluripotin [22]. adhesion, in turn, increases Rho – ROCK activity
Subsequently, Hanna et al. [23] published a study show- even further. This positive feedback loop culminates
ing that the same could be accomplished by constitutive in the hyperactivation of ROCK signalling and the
expression of Klf4 or c-Myc, or small molecules that can complete, irreversible disruption of cell – cell and
replace these factors. Finally, Nichols et al. [24] used LIF cell – ECM adhesion. The integrin-dependent survival
in combination with the small molecules PD0325901 niche for hESCs is thus disturbed, leading to cell
and CHIR99021 (CHIR) to derive NOD mouse ESCs death. Conversely, this irreversible disruption of
by inhibiting GSK3 and ERK pathway signalling, cell – cell adhesion and upregulation of ROCK signal-
respectively. ling are not observed after trypsinization of mESCs.
In summary, small-molecule discoveries have Upon closer investigation, we found that slower endo-
enabled the stable long-term culture of homogeneous cytosis of newly synthesized E-cadherin was the main
mESC cultures, and the derivation of ESCs from reason that cell – cell and cell – ECM interactions are
refractory mouse strains. Perhaps most importantly, not adversely affected following single-cell dissociation
insights gleaned from the small-molecule studies out- of mESCs. This finding also raised the possibility that
Downloaded from https://royalsocietypublishing.org/ on 26 May 2021

lined above are currently being applied to hESCs. the signalling environment of the stem cell niche might
For example, Burton et al. [25] have very recently suc- be a determining factor in the post-dissociation rate of
ceeded in maintaining hESCs in the absence of both endocytosis. Indeed, when hESCs were grown under
feeders and cytokines—including bFGF—by using mESC-like culture and signalling conditions using
the compound erythro-9-(2-hydroxy-3-nonyl)adenine the small molecules PD0325901 (for ERK inhibition)
(EHNA) and its analogues. It is tempting to speculate and SB203580 (a p38 kinase inhibitor) [29], cell-sur-
that we may soon be able to use chemically defined face E-cadherin became much more stable and cell
conditions to derive and maintain more robust death upon trypsin treatment was greatly reduced.
hESCs with greatly improved therapeutic potential. The above example is an elegant, multi-faceted
small-molecule study. While the first part of the
paper takes a phenotype-based approach, using a
4. SMALL MOLECULES HELP ELUCIDATE THE screen to identify two compounds that implicate a
STEM CELL SIGNALLING NICHE known pathway, the second part makes use of a differ-
One of the striking differences between mESCs and ent set of small molecules to conduct a target-based
hESCs is that the latter are very sensitive to single cell follow-up. Together, they demonstrate a novel way in
dissociation (e.g. trypsin treatment). The inability to which the signalling environment of the stem cell
dissociate colonies into single cells drastically limits niche can specify multiple pluripotent states, each
the scope of experiments that can be carried out with characterized by different modes of cell survival and
them; for example, genetic manipulation or any study self-renewal.
requiring clonal analysis becomes quite difficult. We Another, perhaps underappreciated, component of
sought to find a small-molecule solution to this problem the stem cell niche comprises metabolites, the multitude
by performing a high-throughput phenotypic screen of of endogenous small molecules generated as by-pro-
50 000 synthetic compounds, looking for molecules that ducts of intracellular reactions. Given how potent
could promote survival following dissociation by trypsin. synthetic small molecules’ effects on stem cell signalling
We ultimately found two: thiazovivin (Tzv; a 2,4- and homeostasis can be, it is hard to imagine that
disubstituted thiazole) and pyrintegrin (Ptn; a 2,4-disub- metabolites would not in some fashion affect stem cell
stituted pyrimidine). Both compounds enhance survival homeostasis. For example, might there be a connection
following dissociation more than 30-fold, without between the characteristics of stem cells’ metabolomes
adversely affecting pluripotency over long-term culture and the fate decisions they make?
in chemically defined medium [26]. To answer this question, we collaborated with
Neither compound has an appreciable effect on cell Siuzdak and co-workers to take an untargeted meta-
proliferation; rather, both substantially increase bolomics approach: using liquid chromatography–
adhesion to extracellular matrix (ECM, e.g. Matrigel electrospray ionization–time of flight mass spectrometry
or laminin), but not gelatin. The increased cell – (LC-ESI-TOF-MS), we quantitatively characterized
ECM interaction, in turn, promotes cell survival and and compared the metabolomes of mESCs and
growth by activating integrin signalling and the down- mESC-derived neurons or cardiomyocytes [30]. The
stream phosphatidylinositol-3-kinase (PI3K) and chemical formulae for over 150 differentially regulated
MAPK pathways [27,28]. In short, integrin-mediated (difference of greater than 2 ) metabolites and their
cell – ECM interactions form the basis of an essential degree of unsaturation were determined. Interestingly,
survival niche for hESCs. the pluripotent metabolome was found to have a signifi-
We found that survival in the niche is regulated by a cantly higher degree of unsaturation (more than fivefold
positive feedback loop involving E-cadherin-mediated higher relative abundance) compared with that of either
cell – cell interactions and the Rho – ROCK signalling terminally differentiated cell type.
pathway: in a healthy, growing hESC colony, cell – Hypothesizing that the activation of oxidative
cell interactions inhibit Rho–ROCK signalling, which pathways might be linked to the induction of differen-
then further enhances both cell – cell and cell – ECM tiation, we inhibited the pro-oxidative eicosanoid
adhesion. However, when cells are fully dissociated, signalling pathway. Indeed, this inhibition promoted a
Phil. Trans. R. Soc. B (2011)
2212 J. A. Efe & S. Ding Review. Stem cells and small molecules

pluripotent state, even under differentiation conditions. general destabilizer of the pluripotent state, and was
Conversely, individual metabolites downstream of shown to facilitate mesodermal and endodermal differ-
oxidative pathways improved cardiac or neural differen- entiation as well [36]. In a different study, the
tiation three to 15-fold. The same effect was observed to molecules IDE1 and IDE2 were identified as potent
a somewhat lesser degree (50–100% improvement) in inducers of definitive endoderm—even in the absence
hESC differentiation, implying that the role of these of Activin A treatment. While both compounds’ direct
metabolites is conserved. Our results are corroborated targets remain unknown, they were shown to induce
by the observations that hypoxia sustains the pluripotent Smad2 phosphorylation in mESCs [37]. Further, the
state [31,32] and that ESCs maintain lower levels of cells generated using IDE1 and 2 can be subsequently
reactive oxygen species by mostly using non-oxidative differentiated into pancreatic cells when treated with
glycolysis (rather than oxidative phosphorylation) the small molecule indolactam V, which is known to
[33]. Intriguingly, inflammation-resolving metabo- activate protein kinase C (PKC). Provided the relevant
lites appear to accelerate differentiation, potentially developmental pathways are relatively well character-
implicating metabolite regulation in the regenerative ized, the stepwise nature of developmental processes
process during wound healing. lends itself well to multi-step screens that can produce
Prior to our study, these abundant, naturally- highly synergistic and specific effects. Such efforts
occurring endogenous small molecules had been have led to the development of protocols that success-
Downloaded from https://royalsocietypublishing.org/ on 26 May 2021

almost completely overlooked in the context of stem cell fully generate mature neurons [38,39], cardiomyocytes
biology. In another study, Garcia-Gonzalo & Izpisua [40,41] and pancreatic cell types [42] from hESCs.
Belmonte [34] demonstrated that albumin-associated On the other hand, a phenotype-based screening
lipids contained in knock-out serum replacer (KSR) are approach can be taken to identify the contributions of
responsible for its promotion of hESC self-renewal. In one or more pathways to a specific developmental out-
short, it is now clear that metabolites have active role(s) come. For example, our earlier efforts in this realm
in balancing self-renewal and differentiation. include a cell-based screen that identified TWS119, a
neurogenic compound that was found to inhibit GSK3
[43]. We also identified the compounds cardiogenol
5. DIRECTED DIFFERENTIATION OF EMBRYONIC (A–D) and purmorphamine as effective inducers of
STEM CELLS the cardiogenic and oesteogenic programmes, respect-
To realize the true potential of pluripotent stem cells, ively [44,45]. More recently, we have discovered a
significant improvements will be required in the synthetic compound dubbed neuropathiazol in a high-
methods used to differentiate them into functional content image-based screen for inducers of neurons
cells relevant from both a basic research and clinical from hippocampal progenitor cells. Neuropathiazol is
standpoint. Traditionally, the first step in the process highly neurogenic, even under unfavourable gliogenic
has been the formation of embryoid bodies (EBs) in conditions [46].
suspension and spontaneous differentiation. The cells These advances underscore the relevance of small
of interest must then be isolated from this very hetero- molecule-driven approaches to the elucidation of basic
geneous mass comprising numerous cell types from developmental biology as well as the eventual clinical
all three germ layers. This method leaves much to be use of ESC-derived differentiated cells. From a clinical
desired in terms of efficiency, speed and reproducibil- perspective, small molecules will impact not only tech-
ity. The resulting cells are not therapeutically useful, nical feasibility, but also cost-effectiveness (e.g. by
and do not lend themselves to detailed analysis of par- replacing expensive growth factors, etc.) In any case,
ticular differentiation pathways. Likewise, the use of as we make progress on the clinical front, it will be
co-culture set-ups or conditioned media brings with imperative that questions regarding cellular equivalence
it many of the same issues. Ideally, directed differen- (i.e. small molecule versus naturally generated) and the
tiation protocols would use only chemically defined in vivo relevance of pathways characterized in vitro be
media to avoid these problems. Tremendous head- answered clearly and definitively.
way has no doubt been made over the years
(reviewed in [35]), but much remains unknown, and
small molecule approaches can make critical, lasting 6. SMALL MOLECULES CAN MODULATE THE
contributions. Specifically, they can play inductive BEHAVIOUR OF TISSUE-SPECIFIC STEM CELLS
roles in differentiation by synergizing with growth fac- Many mature tissues harbour rare populations of
tors and cytokines to incrementally recapitulate in vivo multi- and oligopotent adult stem/progenitor cells.
development in a precise and efficient fashion. These lineage-restricted precursors can play critical
In addition, they may allow the controlled expansion roles in tissue homeostasis and, more importantly,
of desired precursor populations. Finally, they can inhi- the regenerative process following injury and disease.
bit certain processes, such as the self-renewal of stem These qualities make adult stem cells excellent candi-
cells or progression along an unwanted developmental dates for cell-based therapy approaches, provided their
route at the intermediate stages of differentiation. potential can be harnessed as required. This approach
For example, the compound stauprimide can pro- was pioneered decades ago with the advent of bone
mote the induction of Sox17þ endoderm by acting marrow transplants, of which haematopoietic stem
synergistically with the cytokine Activin A. Follow-up cells (HSCs) are the critical component. However,
biochemical studies indicated that it inhibits the subsequent attempts using other cell types in various
nuclear localization of NME2, a transcription factor organ/tissue systems have typically not met with the
that lies upstream of c-Myc. Stauprimide is thus a same level of success. Aside from issues relating to
Phil. Trans. R. Soc. B (2011)
Review. Stem cells and small molecules J. A. Efe & S. Ding 2213

the histocompatibility of allogeneic cell transplants, The company Nuvelo is developing a drug called
difficulties in determining the cell type-specific R-spondin 1 that treats inflammatory bowel disease
requirements for the ex vivo survival and proliferation by inducing stem cell proliferation in the intestinal
of adult stem cells have universally impeded progress. crypt and a corresponding increase in epithelial cells.
Fortunately, phenotypic screens for compounds Interestingly, like BIO, both BCI-540 and R-spondin
mediating survival, proliferation and differentiation 1 exert their effects through modulation of the Wnt
of these cells have yielded promising results. Two pathway [52– 54].
studies representing successful implementation of
small-molecule approaches are outlined below. Ulti-
mately, such research should lead to the development 7. FACILITATING SOMATIC CELL
of drugs that either facilitate ex vivo manipulation of REPROGRAMMING
adult stem cells or enable/enhance the regenerative In late 2006, Shinya Yamanaka et al. [55] demon-
process by exerting precise control over these cells strated that it was possible to revert murine
in vivo. We have also provided examples of current fibroblasts in vitro to a pluripotent mESC-like state
drug-development efforts to underscore the immediate by retrovirally overexpressing just four genes: Oct4,
clinical relevance of adult stem cell studies that use Sox2, Klf4 and c-Myc. Given the dogma-shattering
small molecules. implications of this finding for cell and developmental
Downloaded from https://royalsocietypublishing.org/ on 26 May 2021

Three years ago, our laboratory did a collaborative biology—and an astonishing simplicity that could
study on the small-molecule control of cardiac precur- eventually lend itself to the development of autologous
sor cell proliferation and differentiation [47]. At this cell-based therapies—reprogramming of somatic cells
time, it was known that multi-potent Isl1 þ cardiac pre- to induced pluripotent stem cells (iPSCs) quickly
cursors could be expanded in co-culture with cardiac became an intensely studied area of cell biology.
mesenchymal cells (CMCs) following their isolation Over the past 4 years, iPSCs have been generated
from post-natal hearts. In order to gain a better under- from human fibroblasts and a multitude of other cell
standing of the extracellular signals regulating this types, some using fewer or different sets of genes
expansion, we used genetically-marked (LacZ) Isl1 þ [7,56 – 58]. A number of non-viral methods (reviewed
precursors in a screen of approximately 15 000 com- in [59]) have also been developed, including the use of
pounds. We identified three small molecules that recombinant proteins [60].
could dramatically expand this small population, However, despite such major progress in the field,
among which 6-bromoindirubin-30 -oxime (BIO) at least two aspects of reprogramming remain proble-
stood out as a known inhibitor of GSK3 and hence a matic. First, a requirement for genetic modification
positive regulator of the canonical Wnt pathway [48]. greatly restricts the downstream utility of virally gener-
Treatment with BIO reproducibly induced a greater ated iPSCs, i.e. they are not therapeutically viable as
than sevenfold increase in Isl1 þ cell number. Follow- long as insertional mutagenesis and reactivation of
up experiments revealed that the Wnt/b-catenin exogenous genes—especially oncogenes like c-Myc—
pathway has a multi-phasic role in cardiac develop- remain a possibility. And while the aforementioned
ment. Importantly, the proliferation-inducing effects use of non-integrating vectors or purified proteins
of BIO, and hence of the canonical Wnt pathway, appears to circumvent these issues, these methods dra-
appear to be conserved in humans. matically lower the efficiency and speed of iPSC
Around the same time, North et al. [49] published generation. This trade-off raises a second concern,
the results of a phenotypic screen for modulators of namely that the rarity of iPSC formation might indi-
HSC numbers in vivo using zebrafish embryos. They cate that cells with (epi)genetic abnormalities are
identified small-molecule regulators of prostaglandin being selected for in the process [61]. Alarmingly,
E2 (PGE2) synthesis as potent regulators of HSC there is substantial evidence that downregulation of
number, and went on to show that 16,16-dimethyl tumour suppressor genes improves reprogramming
PGE2 (a stable PGE2 analogue) improved kidney outcomes [62 – 64].
marrow recovery following irradiation injury in adult Small molecules have the potential to alleviate both
zebrafish. Furthermore, this effect was found to be of these concerns by (i) functionally replacing the
conserved in mice, where PGE2 treatment enhanced Yamanaka factors, and (ii) improving speed and effi-
progenitor cell proliferation in ESC differentiation ciency to minimize the potential for acquisition of
assays. Finally, ex vivo treatment of bone marrow undesirable traits in culture. Importantly, the induc-
cells with 16,16-dimethyl PGE2 was observed to tion of pluripotency using small molecules represents
increase successful homing of HSCs in the body. a radically different (i.e. indirect) approach when com-
This study and others (reviewed in [50]) indicated pared with the derivation of iPSCs or somatic cell
that small molecules may have important role(s) to nuclear transfer (SCNT), which undoubtedly overlap
play in the ex vivo manipulation of HSCs prior to considerably from a mechanistic standpoint. Finding
transplantation, a possibility borne out only 2 years such compounds is an admittedly difficult task, but
later when Fate Therapeutics, Inc. announced promis- the combined power of modern synthetic chemistry
ing results using a compound named FT1050 in and high-throughput screening technology should
clinical trials [51]. Other companies have been pursu- not be underestimated. Moreover, certain cell types,
ing similar strategies: for example, Brain Cells, Inc. has e.g. neural precursor cells (NPCs) and keratinocytes,
found that its compound BCI-540 specifically acti- naturally express high levels of one or more of the
vates neurogenesis in the hippocampus, and has Yamanaka factors, a feature that can be taken advan-
potential for treatment of anxiety and depression. tage of in the generation of iPSCs [56,65]. Such cells
Phil. Trans. R. Soc. B (2011)
2214 J. A. Efe & S. Ding Review. Stem cells and small molecules

are especially good candidates for chemical reprogram- blastocyst, and can generate chimeric or even entirely
ming, as they may require as few as one factor for mESC-derived animals when transplanted back into
reprogramming (Oct4 is the only pluripotency factor the blastocyst using the appropriate methods [76].
that is expressed exclusively in stem cells). Finally, EpiSCs, on the other hand, are obtained from post-
reprogramming appears to involve a mesenchymal- implantation egg cylinder-stage epiblasts of mice and
to-epithelial transition (MET) step, suggesting that rats [10]. Just like mESCs, EpiSCs appear to be pluripo-
the use of cells with epithelial phenotypes might be tent, in that they express the same core pluripotency
preferable [57]. transcription factors (Oct4, Sox2 and Nanog) and can
Small molecules that improve reprogramming give rise to tissues of all three germ layers in vitro or in
generally fall into one of two categories. Some com- vivo (only in teratoma assays). However, they exhibit a
pounds have a global effect on cellular plasticity; number of striking differences in cell morphology, cul-
these include global epigenetic modifiers like valproic ture traits and signalling mechanisms [77,78]: mESCs
acid (VPA; a histone deacetylase (HDAC) inhibitor), form domed colonies, have a doubling time of approxi-
RG108 (a DNA methyltransferase inhibitor) and mately 12 h, and are not appreciably affected by single-
parnate (an H3K4 histone demethylase inhibitor). cell dissociation, whereas EpiSCs grow in flattened
In human cells, low levels of the naturally occurring sheets, double roughly every 36 h, and do not tolerate
HDAC inhibitor butyrate have been recently shown single-cell dissociation well. Moreover, treatment with
Downloaded from https://royalsocietypublishing.org/ on 26 May 2021

to non-specifically improve reprogramming efficiency LIF (JAK–STAT3 pathway) and BMP4 (SMAD–Id
as much as 50-fold [66]. Other compounds exert pathway) have traditionally been used to promote
their effects by modulating a specific signalling pathway mESC self-renewal, while EpiSCs depend on FGF2
to replace one or more of the reprogramming factors. and Activin A for long-term maintenance in culture
Our work has identified compounds in both categories. [12,79]. Most importantly, though, EpiSCs’ poor con-
For example, we have shown that inhibition of the tribution to chimerism suggested that they represent a
histone methyltransferase G9a with the compound different and less pluripotent state than mESCs.
BIX01294, when combined with the L-calcium channel This hypothesis has recently been confirmed by the
agonist BayK8644, enables the reprogramming of finding that overexpression of Klf4, Nr5a and Nanog
fibroblasts transduced with only Oct4 and Klf4 [67]. can revert EpiSCs to an mESC-like state [80,81].
Interestingly, BIX01294 is also capable of replacing Furthermore, we have identified a combination of
Oct4 in NPCs; however, this requires exogenous overex- four small molecules that induce the same reversion:
pression of all three remaining Yamanaka factors [68]. concomitant treatment of EpiSCs with inhibitors of
Strikingly, Huangfu et al. [69] have demonstrated that the histone demethylase LSD1, ALK5, GSK3 and
VPA treatment is sufficient to replace both Klf4 and the MEK pathway produces chimerism-competent
c-Myc in neonatal human fibroblasts. mESC-like cells [82].
In the context of directed pathway modulation, we Interestingly, hESCs very closely resemble EpiSCs
have shown that the GSK3 inhibitor CHIR can replace in all aspects of their phenotype and culture require-
Sox2 in the reprogramming of mouse fibroblasts trans- ments. This observation raises the possibility that,
duced only with Oct4 and Klf4. When combined with despite also being derived from the ICM, hESCs are
parnate, CHIR permits the generation of iPSCs from not equivalent to mESCs in terms of their pluripo-
human fibroblasts overexpressing the same two factors tency state. While mESCs have been successfully
[70]. Ichida et al. [71] were also able to replace Sox2 in cultured in a naive ground state, one might speculate
the reprogramming of mouse fibroblasts with the com- that hESCs have hitherto eluded capture at this
pound E-616452 (renamed RepSox), which inhibits stage, undergoing limited differentiation—equivalent
the transforming growth factor-beta (TGF-b) receptor to mouse EpiSCs—prior to establishment in culture.
to upregulate the expression of Nanog. Another likely Given the field’s recent success in reverting EpiSCs
way in which the inhibition of TGF-b signalling con- back to a more naive mESC-like pluripotent state,
tributes to reprogramming success is by facilitating we and others have asked whether the same conver-
MET [72]. It is thus not surprising that Maherali & sion might work for hESCs. This question can be
Hochedlinger [73] could replace both Sox2 and addressed in two ways: by starting with somatic cells
c-Myc by using an inhibitor of TGF-b receptor I and using reprogramming to artificially induce the
kinase/activin-like kinase 5 (Alk5). Lyssiotis et al. ground state, or by attempting to derive naive pluripo-
[74] have shown that kenpaullone, a GSK3 inhibitor, tent cells directly from human blastocysts. Taking the
can replace Klf4 in the reprogramming of MEFs former approach, we found that four-factor trans-
overexpressing Oct4, Sox2 and c-Myc—albeit in an duced human fibroblasts gave rise to mESC-like
as-of-yet unknown, GSK3-independent manner. pluripotent cells when cultured in mESC medium
Finally, in the human system, we have shown that containing human LIF. The nascent colonies could
combined treatment of fibroblasts with the Alk5 be reliably expanded in the presence of a small-mol-
inhibitor SB431542, the MEK inhibitor PD0325901 ecule cocktail containing PD0325901, A-83-01 and
and thiazovivin improves four-factor reprograming CHIR99021—inhibitors of MEK, ALK5 and GSK3,
efficiency greater than 200-fold [75]. respectively [83]. In a related study, Hanna et al.
Aside from re-establishing the pluripotency pro- [84] were able to isolate the same type of naive cells
gramme in somatic cells, reprogramming has also been by ectopically expressing Oct4 in hESCs and simul-
used to transition between different pluripotent states, taneously treating the cells with LIF, inhibitors of
specifically those of ESCs and EpiSCs. The former GSK3 and ERK1/2, and forskolin, a protein kinase
functionally represent the ICM of the pre-implantation A (PKA) pathway agonist that can induce Klf2 and
Phil. Trans. R. Soc. B (2011)
Review. Stem cells and small molecules J. A. Efe & S. Ding 2215

Klf4 expression. The same conditions were also process would undoubtedly represent a completely
permissive for the derivation of human iPSCs. novel path to reprogramming compared with SCNT
Unfortunately, despite these important advances, or defined transcription factor-based induction of
attempts to derive mESC-like hESCs from embryos pluripotency. These two established methods of repro-
have thus far not succeeded. Lengner et al. [85] have gramming both rely on maternally stored or
shown that it is possible to derive female hESCs that ectopically expressed transcription factors, the highly
have not inactivated one of their X chromosomes, a specific functions of which have evolved over millions
hallmark of mESCs and fully reprogrammed mouse of years. We unfortunately cannot escape the fact that
iPSCs. However, in every other way, these cells still man-made small molecules, as effective as they can be,
resemble regular hESCs. The fact that relatively few do not possess any functional entitie(s) analogous to
mouse strains have spontaneously given rise to stable the highly specific DNA recognition and transcription
mESC lines suggests that their naive pluripotent domains that render transcription factors so extra-
state is a metastable one. This metastability is at ordinarily powerful. Attaining the same degree of
least in part owing to genetic and epigenetic differ- potency and specificity with small molecules, which by
ences between various mouse strains, which have yet necessity must indirectly bring about the desired out-
to be fully characterized. A more detailed understand- come, represents both a fundamental challenge and an
ing of the impact of genetic and epigenetic background opportunity to achieve truly groundbreaking progress.
Downloaded from https://royalsocietypublishing.org/ on 26 May 2021

on pluripotency may be needed before we can derive Small molecules have also helped define distinct
and maintain the mESC-like naive pluripotent stem metastable states of pluripotency and self-renewal,
cells from humans and other mammalian species. thereby facilitating transitions from one state to
A recent study elegantly demonstrates the dramatic another. A related future role for small molecules in
impact a slight difference in genetic makeup can have reprogramming might be to ensure that the epigen-
on the way different species regulate the stability of omes of iPSCs are fully reset, as it was very recently
the pluripotent state [86]: mESCs, with their uniquely shown that certain iPSCs may retain an epigenetic
stunted cell cycle, were found to be critically depen- ‘memory’ of their origins, developmentally restricting
dent on high levels of threonine dehydrogenase or biasing them towards these particular lineages [91].
activity as a source of one-carbon metabolism for
purine synthesis. Intriguingly, humans do not have a
functional threonine dehydrogenase gene, a deficiency 8. SMALL MOLECULES HELP ESTABLISH A NEW
that would probably put hESCs at a significant disad- TRANSDIFFERENTIATION PARADIGM
vantage for self-renewal under mESC-like conditions. Most recently, our laboratory has been focusing on a
Consistent with our hypothesis, we found that our hitherto unexplored aspect of somatic cell reprogramm-
above cocktail of compounds (PD0325901, A-83-01 ing. We and others have observed that reprogramming
and CHIR99021), when supplemented with LIF, did using the Yamanaka factors often gives rise to rare side
allow for the generation of naive, mESC-like rat iPSCs populations of cells that do not yield any iPSCs. While
that were capable of contributing to chimerism [83]. some of these clusters consist of nondescript granular
Similarly, others have found that rat ESCs can be cells reported to be unproductive iPSC intermediates
derived from blastocysts under chemically defined [92,93], others exhibit more complex morphological
conditions that include PD0325901, CHIR99021 and traits. Intriguingly, this latter group may express marker
LIF [87,88]. More recently, another study generated genes characteristic of partially or fully differentiated
rat ESCs using four small molecules, including cells entirely unrelated to the starting population—the
PD0325901, A-83-01, CHIR99021 and Y27632 [89]. hallmark of transdifferentiation. We have, for example,
Y27632 is a Rho-associated protein kinase inhibitor observed spontaneously contracting foci of cells and/or
that can stabilize E-cadherin [26]. These derived cells the presence of neurons in a very small numbers.
successfully contributed to the generation of transgenic This unexpected observation led us to ask the
rats via germline transmission, proving their occupancy following question: might it be possible to more
of the naive ground state. These advances have paved specifically instruct reprogramming towards one or
the way for the use of homologous recombination in more of these side populations by altering reprogram-
rat ESCs to generate knock-in strains or loss-of-function ming conditions? Implicit in this question is the idea
mutants. Recently, a p53 knockout rat has been success- that iPSCs may represent but one of many possible
fully generated using this strategy [90]. Such strains outcomes of the Yamanaka factor-based reprogram-
represent invaluable genetic tools, as rats are potentially ming process. We speculated that overexpression of
better suited as a model to study human biology and dis- the Yamanaka factors—especially early on—could be
eases, especially multi-factorial ones. Furthermore, the priming the cells for rapid lineage switching by transi-
successful establishment of rat ESC cultures may pro- ently creating an epigenetically very unstable and
vide the technical framework for the derivation of plastic state. To test this hypothesis, we modified stan-
naive ESCs from other model organisms (e.g. primates) dard reprogramming procedures in ways we thought
and livestock. might ‘unmask’ alternative outcomes. First, we
In summary, significant progress has been made employed a doxycycline-inducible system to limit
towards ‘chemical reprogramming’, insofar as all four Yamanaka factor expression to the bare minimum
Yamanaka factors have now been individually replaced required for reprogramming to take place, as indicated
by small molecules. The real challenge will be to find a by various morphological changes culminating in
combination that can replace all four simultaneously, colony formation. We simultaneously tested condi-
and do so rapidly and efficiently. Mechanistically, this tions conducive to the generation and/or survival of
Phil. Trans. R. Soc. B (2011)
2216 J. A. Efe & S. Ding Review. Stem cells and small molecules

small molecule
unstable inhibition of
intermediates JAK/STAT signalling

induced
pluripotency
prolo
nged
t
expre ransgene
varying ssion
epigenetic instability

treatments

alternative cell
fates
Isl1+

Nkx2.5+ heart
fibroblasts
progenitors
Downloaded from https://royalsocietypublishing.org/ on 26 May 2021

(Re-differentiation)
cardiomyocytes
developmental potential

Figure 2. A new transdifferentiation paradigm. In our model, transient high-level reprogramming factor expression leads to the
formation of epigenetically unstable (i.e. more naive and ‘activated’) intermediates, which subsequently ‘relax’ back into more
stable state(s). In this scheme, the generation of iPSCs is unlikely, as complete de-differentiation requires prolonged overex-
pression of pluripotency factors. On the other hand, direct reprogramming to various progenitor cells—and perhaps
terminally differentiated cells as well—is a feasible outcome, especially if (i) culture conditions allow and/or promote their
formation and (ii) establishment of pluripotency is prevented by using a small-molecule inhibitor of JAK/STAT signalling.

stem cell homeostasis and


maintenance

defining the improving


cellular niche and directed
signalling R3 differentiation
X
R5 N N
X R1 R4
N N X
R2

modulating adult stem chemical reprogramming


cell behaviour and transdifferentiation

Figure 3. A schematic of stem cell-related small molecule efforts as presented in this review.

lineage-restricted cells, e.g. cardiomyocytes, rather successful in generating spontaneously contracting


thaniPSCs: among other things, this entailed omitting cardiomyocytes from embryonic and adult fibroblasts
LIF from the media, and switching to chemically defined in 12 – 13 days [94]. A recent study has accompli-
conditions (including growth factors/cytokines) early on. shed much the same using cardiac lineage-specific
Importantly, we again enlisted the help of small transcription factor overexpression [95]; however, it
molecules in the process. Not only did we use an is important to note that the time required for the
inhibitor of the JAK/STAT pathway to suppress signal- development of contraction is considerably longer in
ling critical to the establishment of pluripotency, but this case.
we also employed a Wnt pathway modulator to pro- Since we have achieved transdifferentiation using only
mote cardiogenesis during an empirically determined the Yamanaka factors, it has broad ramifications and
optimal time window. This strategy has been very applicability. Accordingly, we were able to successfully

Phil. Trans. R. Soc. B (2011)


Review. Stem cells and small molecules J. A. Efe & S. Ding 2217

Table 1. Small molecules used in the studies reviewed and their relevance to stem cell biology.

reference
name of compound target(s) effect(s) no.

(2) indolactam V PKC pancreatic differentiation of ESC-derived [37]


endoderm
(þ) Bayk 8644 L-type Ca2þ channel enhances reprogramming of MEFs [67]
16,16-dimethyl EP receptor(s) increases frequency of long-term HSCs; [49]
prostaglandin E2 improved kidney regeneration
6-bromoindirubin-30 - GSK3 self-renewal of mESCs and proliferation [47,48]
oxime (BIO) of Isl1 þ cardiac precursors
A-83-01 ALK4, ALK5 and rat iPSC self-renewal, in combination with [83,89]
ALK7 CHIR99021 and PD0325901
BCI-540 Wnt pathway neurogenesis in the hippocampus [51]
BIX-01294 G9a HMTase reprogramming of NPCs and MEFs [67,68]
transduced with only Oct4 and Klf4
cardiogenols (A –D) unknown efficient induction of cardiogenesis in mESCs [44]
CHIRON99021 (CHIR) GSK3 activates canonical Wnt signalling, promotes [24,70,83,
Downloaded from https://royalsocietypublishing.org/ on 26 May 2021

mESC self-renewal 87– 89]


erythro-9-(2-hydroxy-3- phosphodiesterase allows hESC maintenance in chemically [25]
nonyl)adenine (EHNA) (PDE2) defined media
forskolin PKA promotes neurogenesis along with retinoic [84]
acid
FT1050 Wnt and prostaglandin improves HSC function and engraftment [51]
E2
IDE1/2 unknown Smad2 phosphorylation in ES cells; [37]
differentiation into endoderm
kenpaullone GSK3 and CDKs replaces Klf4 in the reprogramming of MEFs [74]
neuropathiazol unknown induces neuronal differentiation of multi- [46]
potent adult hippocampal NPCs
parnate lysine-specific reprogramming of human keratinocytes [70]
demethylase I transduced with Oct4/Klf4
PD0325901 MEK inhibits differentiation of mESCs [24,29,75,
83,87–
89]
pluripotin (SC1) RasGAP and ERK1 promotes mESC self-renewal [16,22]
purmorphamine Hedgehog induction of osteogenesis [45]
RG108 DNA MTase facilitates reprogramming of MEFs [66]
R-spondin 1 Wnt pathway stem cell proliferation in the intestinal crypt [54]
SB203580 p38-MAPK used in conjunction with PD0325901 to [29]
maintain hESCs in an mESC-like
signalling niche
SB431542 ALK4, ALK5 and efficient neural differentiation of hES [75]
ALK7 cells in combination with Noggin
stauprimide NME2 enhanced differentiation of ES cells [36]
thiazovivin (Tzv) ROCK improved survival of hESCs upon dissociation [26,75]
TWS119 GSK3 neurogenesis enhancer [43]
pyrintegrin (Pyr) unknown improved survival of hESCs upon dissociation [26]
valproic acid (VPA) histone deacetylase enables reprogramming of human fibroblasts [66,69]
transduced with only Oct4 and Sox2;
globally acting factor
Y27632 ROCK improved survival of hESCs upon dissociation [26,89]

adapt the process to the rapid and efficient generation of appear to specifically modulate enzymatic action or
NPCs from fibroblasts [96]. We believe that we have thus receptor signalling may have as-of-yet undetected sec-
established a new transdifferentiation paradigm, and ondary effects. It is important that dogma does not
small molecules have once again played a prominent become embedded around such compounds as this
role in the process (figure 2). may cloud understanding of their true mechanism of
action, a sine qua non for their deployment clinically.
As has been the case in the past few years, the
9. CONCLUSIONS AND PERSPECTIVES coming decade will no doubt see numerous advances
Our understanding of the biology of stem cells, in these areas, which should lead to the development
especially with respect to hESCs and somatic cell of effective novel drugs.
reprogramming, remains relatively superficial. Like- In this review, we have summarized the ways in which
wise, our knowledge of small-molecule mechanisms small molecules have contributed to the advancement of
of action is also incomplete; some compounds that stem cell research (figure 3 and table 1), and how they
Phil. Trans. R. Soc. B (2011)
2218 J. A. Efe & S. Ding Review. Stem cells and small molecules

will continue to shape these efforts. By allowing us to 6 Emre, N., Coleman, R. & Ding, S. 2007 A chemical
systematically probe known signalling pathways, small- approach to stem cell biology. Curr. Opin. Chem. Biol.
molecule studies are defining very specific requirements 11, 252 –258. (doi:10.1016/j.cbpa.2007.04.024)
for the robust propagation of pluripotent cells. Such 7 Thomson, J. A., Itskovitz-Eldor, J., Shapiro, S. S.,
Waknitz, M. A., Swiergiel, J. J., Marshall, V. S. &
optimization and standardization of growth conditions,
Jones, J. M. 1998 Embryonic stem cell lines derived
in turn, are greatly facilitating the effective and reprodu- from human blastocysts. Science 282, 1145–1147.
cible induction of directed differentiation (one of the key (doi:10.1126/science.282.5391.1145)
goals being large/industrial-scale generation of desired 8 Evans, M. J. & Kaufman, M. H. 1981 Establishment in
populations). Small molecules have also come to play culture of pluripotential cells from mouse embryos.
prominent roles in this next step, e.g. by mimicking Nature 292, 154 –156. (doi:10.1038/292154a0)
niche signalling to more precisely recapitulate in vivo 9 Kanatsu-Shinohara, M. et al. 2004 Generation of pluri-
development and/or by inhibiting unwanted signalling potent stem cells from neonatal mouse testis. Cell 119,
input that invariably reduces efficiency. At the level of 1001–1012. (doi:10.1016/j.cell.2004.11.011)
basic biology, small molecules have proved useful in 10 Brons, I. G. et al. 2007 Derivation of pluripotent epiblast
defining how particular elements of the niche influence stem cells from mammalian embryos. Nature 448,
191 –195. (doi:10.1038/nature05950)
stem cell survival and proliferation. Consequently, their
11 Wagers, A. J. & Weissman, I. L. 2004 Plasticity of adult
utility in activating or potentiating adult stem cells is stem cells. Cell 116, 639– 648. (doi:10.1016/S0092-
Downloaded from https://royalsocietypublishing.org/ on 26 May 2021

already being tested in clinical trials. Finally, small mol- 8674(04)00208-9)


ecules represent a highly desirable alternative to genetic 12 Niwa, H., Burdon, T., Chambers, I. & Smith, A. 1998
modification in reprogramming and transdifferentia- Self-renewal of pluripotent embryonic stem cells is
tion; true ‘chemical reprogramming’ would be easier, mediated via activation of STAT3. Genes Dev. 12,
more effective and much safer from a disease treatment 2048–2060. (doi:10.1101/gad.12.13.2048)
perspective. Many, if not all, of the above small molecule 13 Nichols, J., Chambers, I., Taga, T. & Smith, A. 2001
approaches could conceivably lend themselves to both Physiological rationale for responsiveness of mouse
in vivo and ex vivo clinical implementation. Which embryonic stem cells to gp130 cytokines. Development
128, 2333–2339.
route—or combination of routes—is ultimately chosen
14 Li, Y., McClintick, J., Zhong, L., Edenberg, H. J., Yoder,
will largely depend on empirical success, and is very M. C. & Chan, R. J. 2005 Murine embryonic stem cell
difficult to ascertain at this point. It is almost certain, differentiation is promoted by SOCS-3 and inhibited
however, that the interest in these efforts will continue by the zinc finger transcription factor Klf4. Blood 105,
to grow at its current rapid pace for the foreseeable 635 –637. (doi:10.1182/blood-2004-07-2681)
future. 15 Ying, Q. L. & Smith, A. G. 2003 Defined conditions for
As it becomes increasingly clear that advances in neural commitment and differentiation. Methods Enzymol.
stem cell biology have the potential to radically 365, 327–341. (doi:10.1016/S0076-6879(03) 65023-8)
transform the way diseases are treated in the 16 Chen, S., Do, J. T., Zhang, Q., Yao, S., Yan, F., Peters,
not-so-distant future, small molecules may be poised E. C., Scholer, H. R., Schultz, P. G. & Ding, S. 2006
to once again take centre stage in a profound paradigm Self-renewal of embryonic stem cells by a small molecule.
Proc. Natl Acad. Sci. USA 103, 17 266 –17 271. (doi:10.
shift, just as they did decades ago with the discovery of
1073/pnas.0608156103)
antibiotics. They might be considered small in the 17 Ying, Q. L., Wray, J., Nichols, J., Batlle-Morera, L.,
molecular realm, but their impact on humanity has Doble, B., Woodgett, J., Cohen, P. & Smith, A. 2008
been, and continues to be, enormous. The ground state of embryonic stem cell self-renewal.
We thank all members of the Ding laboratory for their Nature 453, 519 –523. (doi:10.1038/nature06968)
tireless work and stimulating discussions. S.D. is supported 18 Yuan, H., Corbi, N., Basilico, C. & Dailey, L. 1995
by funding from NICHD, NHLBI and NIMH/NIH, Developmental-specific activity of the FGF-4 enhancer
California Institute for Regenerative Medicine, Prostate requires the synergistic action of Sox2 and Oct-3. Genes
Cancer Foundation, Fate Therapeutics, Esther B. O’Keeffe Dev. 9, 2635– 2645. (doi:10.1101/gad.9.21.2635)
Foundation and The Scripps Research Institute. J.A.E. is a 19 Stefanovic, S., Abboud, N., Desilets, S., Nury, D.,
Lowe Family Foundation fellow. Cowan, C. & Puceat, M. 2009 Interplay of Oct4 with
Sox2 and Sox17: a molecular switch from stem cell plur-
ipotency to specifying a cardiac fate. J. Cell Biol. 186,
665 –673. (doi:10.1083/jcb.200901040)
20 Snyder, M., Huang, X. Y. & Zhang, J. J. 2010 Stat3
REFERENCES directly controls the expression of Tbx5, Nkx2.5, and
1 Drews, J. 2000 Drug discovery: a historical perspective. GATA4 and is essential for cardiomyocyte differentiation
Science 287, 1960–1964. (doi:10.1126/science.287.5460. of P19CL6 cells. J. Biol. Chem. 285, 23 639–23 646.
1960) (doi:10.1074/jbc.M110.101063)
2 Cayen, M. N. 2010 Early drug development: strategies and 21 Wagner, R. T., Xu, X., Yi, F., Merrill, B. J. & Cooney, A. J.
routes to first-in-human trials. Hoboken, NJ: Wiley. 2010 Canonical Wnt/b-catenin regulation of liver receptor
3 Dandapani, S. & Marcaurelle, L. A. 2010 Current strat- homolog-1 mediates pluripotency gene expression. Stem
egies for diversity-oriented synthesis. Curr. Opin. Chem. Cells. 28, 1794–1804. (doi:10.1002/stem.502)
Biol. 14, 362 –370. (doi:10.1016/j.cbpa.2010.03.018) 22 Yang, W. et al. 2009 Pluripotin combined with leukemia
4 Wilk, W., Zimmermann, T. J., Kaiser, M. & Waldmann, inhibitory factor greatly promotes the derivation of
H. 2010 Principles, implementation, and application embryonic stem cell lines from refractory strains. Stem
of biology-oriented synthesis (BIOS). Biol Chem. 391, Cells 27, 383 –389. (doi:10.1634/stemcells.2008-0974)
491–497. (doi:10.1515/BC.2010.013) 23 Hanna, J. et al. 2009 Metastable pluripotent states in
5 Dobson, C. M. 2004 Chemical space and biology. Nature NOD-mouse-derived ESCs. Cell Stem Cell 4, 513 –524.
432, 824–828. (doi:10.1038/nature03192) (doi:10.1016/j.stem.2009.04.015)

Phil. Trans. R. Soc. B (2011)


Review. Stem cells and small molecules J. A. Efe & S. Ding 2219

24 Nichols, J., Jones, K., Phillips, J. M., Newland, S. A., 40 Kattman, S. J., Huber, T. L. & Keller, G. M. 2006 Mul-
Roode, M., Mansfield, W., Smith, A. & Cooke, A. tipotent flk-1þ cardiovascular progenitor cells give rise to
2009 Validated germline-competent embryonic stem the cardiomyocyte, endothelial, and vascular smooth
cell lines from nonobese diabetic mice. Nat. Med. 15, muscle lineages. Dev. Cell. 11, 723 –732. (doi:10.1016/
814 –818. (doi:10.1038/nm.1996) j.devcel.2006.10.002)
25 Burton, P. et al. 2010 Identification and characterization 41 Laflamme, M. A. et al. 2007 Cardiomyocytes derived
of small-molecule ligands that maintain pluripotency of from human embryonic stem cells in pro-survival factors
human embryonic stem cells. Biochem. Soc. Trans. 38, enhance function of infarcted rat hearts. Nat. Biotechnol.
1058–1061. (doi:10.1042/BST0381058) 25, 1015–1024. (doi:10.1038/nbt1327)
26 Xu, Y., Zhu, X., Hahm, H. S., Wei, W., Hao, E., Hayek, 42 D’Amour, K. A., Agulnick, A. D., Eliazer, S., Kelly,
A. & Ding, S. 2010 Revealing a core signaling regulatory O. G., Kroon, E. & Baetge, E. E. 2005 Efficient differen-
mechanism for pluripotent stem cell survival and self- tiation of human embryonic stem cells to definitive
renewal by small molecules. Proc. Natl Acad. Sci. USA endoderm. Nat. Biotechnol. 23, 1534–1541. (doi:10.
107, 8129–8134. (doi:10.1073/pnas.1002024107) 1038/nbt1163)
27 Armstrong, L. et al. 2006 The role of PI3K/AKT, 43 Ding, S., Wu, T. Y., Brinker, A., Peters, E. C., Hur, W.,
MAPK/ERK and NFkappabeta signalling in the main- Gray, N. S. & Schultz, P. G. 2003 Synthetic small
tenance of human embryonic stem cell pluripotency molecules that control stem cell fate. Proc. Natl
and viability highlighted by transcriptional profiling and Acad. Sci. USA 100, 7632– 7637. (doi:10.1073/pnas.
functional analysis. Hum. Mol. Genet. 15, 1894 –1913. 0732087100)
Downloaded from https://royalsocietypublishing.org/ on 26 May 2021

(doi:10.1093/hmg/ddl112) 44 Wu, X., Ding, S., Ding, Q., Gray, N. S. & Schultz, P. G.
28 Paling, N. R., Wheadon, H., Bone, H. K. & Welham, 2004 Small molecules that induce cardiomyogenesis in
M. J. 2004 Regulation of embryonic stem cell embryonic stem cells. J. Am. Chem. Soc. 126, 1590–1591.
self-renewal by phosphoinositide 3-kinase-dependent (doi:10.1021/ja038950i)
signaling. J. Biol. Chem. 279, 48 063–48 070. (doi:10. 45 Wu, X., Ding, S., Ding, Q., Gray, N. S. & Schultz, P. G.
1074/jbc.M406467200) 2002 A small molecule with osteogenesis-inducing
29 Burdon, T., Stracey, C., Chambers, I., Nichols, J. & activity in multipotent mesenchymal progenitor cells.
Smith, A. 1999 Suppression of SHP-2 and ERK signalling J. Am. Chem. Soc. 124, 14 520 –14 521. (doi:10.1021/
promotes self-renewal of mouse embryonic stem cells. ja0283908)
Dev. Biol. 210, 30–43. (doi:10.1006/dbio.1999.9265) 46 Lu, Q., Paredes, M., Zhang, J. & Kosik, K. S. 1998 Basal
30 Yanes, O. et al. 2010 Metabolic oxidation regulates extracellular signal-regulated kinase activity modulates
embryonic stem cell differentiation. Nat. Chem. Biol. 6, cell-cell and cell-matrix interactions. Mol. Cell Biol. 18,
411 –417. (doi:10.1038/nchembio.364) 3257 –3265.
31 Ezashi, T., Das, P. & Roberts, R. M. 2005 Low O2 ten- 47 Qyang, Y. et al. 2007 The renewal and differentiation of
sions and the prevention of differentiation of hES cells. Isl1þ cardiovascular progenitors are controlled by a Wnt/
Proc. Natl Acad. Sci. USA 102, 4783–4788. (doi:10. beta-catenin pathway. Cell Stem Cell 1, 165 –179. (doi:10.
1073/pnas.0501283102) 1016/j.stem.2007.05.018)
32 Lin, Q., Lee, Y. J. & Yun, Z. 2006 Differentiation arrest 48 Meijer, L. et al. 2003 GSK-3 –selective inhibitors deri-
by hypoxia. J. Biol. Chem. 281, 30 678 –30 683. (doi:10. ved from Tyrian purple indirubins. Chem. Biol. 10,
1074/jbc.C600120200) 1255 –1266. (doi:10.1016/j.chembiol.2003.11.010)
33 Tsatmali, M., Walcott, E. C. & Crossin, K. L. 2005 New- 49 North, T. E. et al. 2007 Prostaglandin E2 regulates ver-
born neurons acquire high levels of reactive oxygen species tebrate haematopoietic stem cell homeostasis. Nature
and increased mitochondrial proteins upon differentiation 447, 1007 –1011. (doi:10.1038/nature05883)
from progenitors. Brain Res. 1040, 137–150. (doi:10. 50 Broxmeyer, H. E. 2008 Chemokines in hematopoiesis.
1016/j.brainres.2005.01.087) Curr. Opin. Hematol. 15, 49–58. (doi:10.1097/MOH.
34 Garcia-Gonzalo, F. R. & Izpisua Belmonte, J. C. 2008 0b013e3282f29012)
Albumin-associated lipids regulate human embryonic 51 Grayson, P., Mendlein, J., Thies, S. & Yingling, J. 2009
stem cell self-renewal. PLoS ONE 3, e1384. (doi:10. Stem cell modulators (SCMs): a small molecule and bio-
1371/journal.pone.0001384) logic approach to stem cell modulators. Drug Discov.
35 Murry, C. E. & Keller, G. 2008 Differentiation of Today Ther. Strateg. 6, 141–145. (doi:10.1016/J.DD
embryonic stem cells to clinically relevant populations: STR.2009.06.005)
lessons from embryonic development. Cell 132, 52 Li, X., Zhang, Y., Kang, H., Liu, W., Liu, P., Zhang, J.,
661 –680. (doi:10.1016/j.cell.2008.02.008) Harris, S. E. & Wu, D. 2005 Sclerostin binds to LRP5/
36 Zhu, S., Wurdak, H., Wang, J., Lyssiotis, C. A., Peters, 6 and antagonizes canonical Wnt signaling. J. Biol.
E. C., Cho, C. Y., Wu, X. u & Schultz, P. G. 2009 A Chem. 280, 19 883 –19 887. (doi:10.1074/jbc.M41327
small molecule primes embryonic stem cells for differen- 4200)
tiation. Cell Stem Cell 4, 416 –426. (doi:10.1016/j.stem. 53 Semenov, M., Tamai, K. & He, X. 2005 SOST is a
2009.04.001) ligand for LRP5/LRP6 and a Wnt signaling inhibitor.
37 Borowiak, M., Maehr, R., Chen, S., Chen, A. E., Tang, J. Biol. Chem. 280, 26770–26775. (doi:10.1074/jbc.
W., Fox, J. L., Schreiber, S. & Melton, D. A. 2009 M504308200)
Small molecules efficiently direct endodermal differen- 54 Binnerts, M. E. et al. 2007 R-Spondin1 regulates Wnt
tiation of mouse and human embryonic stem cells. Cell signaling by inhibiting internalization of LRP6. Proc.
Stem Cell 4, 348–358. (doi:10.1016/j.stem.2009.01.014) Natl Acad. Sci. USA 104, 14 700 –14 705. (doi:10.
38 Ying, Q. L., Stavridis, M., Griffiths, D., Li, M. & Smith, 1073/pnas.0702305104)
A. 2003 Conversion of embryonic stem cells into neuro- 55 Takahashi, K. & Yamanaka, S. 2006 Induction of pluripo-
ectodermal precursors in adherent monoculture. Nat. tent stem cells from mouse embryonic and adult fibroblast
Biotechnol. 21, 183–186. (doi:10.1038/nbt780) cultures by defined factors. Cell 126, 663–676. (doi:10.
39 Li, X. J., Du, Z. W., Zarnowska, E. D., Pankratz, M., 1016/j.cell.2006.07.024)
Hansen, L. O., Pearce, R. A. & Zhang, S.-C. 2005 Specifi- 56 Kim, J. B. et al. 2009 Oct4–induced pluripotency in
cation of motoneurons from human embryonic stem cells. adult neural stem cells. Cell 136, 411 –419. (doi:10.
Nat. Biotechnol. 23, 215–221. (doi:10.1038/nbt1063) 1016/j.cell.2009.01.023)

Phil. Trans. R. Soc. B (2011)


2220 J. A. Efe & S. Ding Review. Stem cells and small molecules

57 Aasen, T. et al. 2008 Efficient and rapid generation of chemical complementation of Klf4. Proc. Natl Acad.
induced pluripotent stem cells from human keratinocytes. Sci. USA 106, 8912–8917. (doi:10.1073/pnas.09038
Nat. Biotechnol. 26, 1276–1284. (doi:10.1038/nbt.1503) 60106)
58 Loh, Y. H. et al. 2009 Generation of induced pluripotent 75 Lin, T. et al. 2009 A chemical platform for improved
stem cells from human blood. Blood 113, 5476–5479. induction of human iPSCs. Nat. Methods 6, 805–808.
(doi:10.1182/blood-2009-02-204800) (doi:10.1038/nmeth.1393)
59 Abujarour, R. & Ding, S. 2009 Induced pluripotent stem 76 Nagy, A., Gocza, E., Diaz, E. M., Prideaux, V. R., Ivanyi,
cells free of exogenous reprogramming factors. Genome E., Markkula, M. & Rossant, J. 1990 Embryonic stem
Biol. 10, 220. (doi:10.1186/gb-2009-10-5-220) cells alone are able to support fetal development in the
60 Zhou, H. et al. 2009 Generation of induced pluripotent mouse. Development 110, 815 –821.
stem cells using recombinant proteins. Cell Stem Cell 4, 77 Wei, C. L. et al. 2005 Transcriptome profiling of human
381– 384. (doi:10.1016/j.stem.2009.04.005) and murine ESCs identifies divergent paths required to
61 Ohm, J. E. et al. 2010 Cancer-related epigenome changes maintain the stem cell state. Stem Cells 23, 166–185.
associated with reprogramming to induced pluripotent (doi:10.1634/stemcells.2004-0162)
stem cells. Cancer Res. 70, 7662– 7673. (doi:10.1158/ 78 Ohtsuka, S. & Dalton, S. 2008 Molecular and biological
0008-5472.CAN-10-1361) properties of pluripotent embryonic stem cells. Gene
62 Marion, R. M., Strati, K., Li, H., Murga, M., Blanco, R., Ther. 15, 74–81. (doi:10.1038/sj.gt.3303065)
Ortega, S., Fernandez-Capetillo, O., Serrano, M. & Blasco, 79 Qi, X., Li, T. G., Hao, J., Hu, J., Wang, J., Simmons, H.,
M. A. 2009 A p53–mediated DNA damage response limits Miura, S., Mishina, Y. & Zhao, G.-Q. 2004 BMP4 supports
Downloaded from https://royalsocietypublishing.org/ on 26 May 2021

reprogramming to ensure iPS cell genomic integrity. Nature self-renewal of embryonic stem cells by inhibiting mitogen-
460, 1149–1153. (doi:10.1038/nature08287) activated protein kinase pathways. Proc. Natl Acad. Sci.
63 Kawamura, T., Suzuki, J., Wang, Y. V., Menendez, S., USA 101, 6027–6032. (doi:10.1073/pnas.0401367101)
Morera, L. B., Raya, A., Wahl, G. M. & Belmonte, 80 Guo, G. & Smith, A. 2010 A genome-wide screen in
J. C. I. 2009 Linking the p53 tumour suppressor pathway EpiSCs identifies Nr5a nuclear receptors as potent indu-
to somatic cell reprogramming. Nature 460, 1140– 1144. cers of ground state pluripotency. Development 137,
(doi:10.1038/nature08311) 3185–3192. (doi:10.1242/dev.052753)
64 Li, H., Collado, M., Villasante, A., Strati, K., Ortega, S., 81 Guo, G., Yang, J., Nichols, J., Hall, J. S., Eyres, I., Mansfield,
Canamero, M., Blasco, M. A. & Serrano, M. l 2009 The W. & Smith, A. 2009 Klf4 reverts developmentally pro-
Ink4/Arf locus is a barrier for iPS cell reprogramming. grammed restriction of ground state pluripotency.
Nature 460, 1136–1139. (doi:10.1038/nature08290) Development 136, 1063–1069. (doi:10.1242/dev.030957)
65 Hester, M. E., Song, S., Miranda, C. J., Eagle, A., Schwartz, 82 Zhou, H. et al. 2010 Conversion of mouse epiblast stem
P. H. & Kaspar, B. K. 2009 Two factor reprogramming of cells to an earlier pluripotency state by small molecules.
human neural stem cells into pluripotency. PLoS ONE 4, J. Biol. Chem. 285, 29 676 –29 680. (doi:10.1074/jbc.
e7044. (doi:10.1371/journal.pone.0007044) c110.150599)
66 Mali, P. et al. 2010 Butyrate greatly enhances derivation 83 Li, W. et al. 2009 Generation of rat and human induced
of human induced pluripotent stem cells by promoting pluripotent stem cells by combining genetic reprogram-
epigenetic remodeling and the expression of pluripo- ming and chemical inhibitors. Cell Stem Cell 4, 16–19.
tency-associated genes. Stem Cells 28, 713 –720. (doi:10.1016/j.stem.2008.11.014)
(doi:10.1002/stem.402) 84 Hanna, J. et al. 2010 Human embryonic stem cells
67 Shi, Y., Desponts, C., Do, J. T., Hahm, H. S., Scholer, with biological and epigenetic characteristics similar to
H. R. & Ding, S. 2008 Induction of pluripotent stem those of mouse ESCs. Proc. Natl Acad. Sci. USA 107,
cells from mouse embryonic fibroblasts by Oct4 and 9222–9227. (doi:10.1073/pnas.1004584107)
Klf4 with small-molecule compounds. Cell Stem Cell 3, 85 Lengner, C. J. et al. 2010 Derivation of pre-X inacti-
568– 574. (doi:10.1016/j.stem.2008.10.004) vation human embryonic stem cells under physiological
68 Shi, Y., Do, J. T., Desponts, C., Hahm, H. S., Scholer, oxygen concentrations. Cell 141, 872 –883. (doi:10.
H. R. & Ding, S. 2008 A combined chemical and genetic 1016/j.cell.2010.04.010)
approach for the generation of induced pluripotent stem 86 Wang, J., Alexander, P., Wu, L., Hammer, R., Cleaver,
cells. Cell Stem Cell 2, 525 –528. (doi:10.1016/j.stem. O. & McKnight, S. L. 2009 Dependence of mouse
2008.05.011) embryonic stem cells on threonine catabolism. Science
69 Huangfu, D., Osafune, K., Maehr, R., Guo, W., 325, 435 –439. (doi:10.1126/science.1173288)
Eijkelenboom, A., Chen, S., Muhlestein, W. & Melton, 87 Buehr, M. et al. 2008 Capture of authentic embryonic
D. A. 2008 Induction of pluripotent stem cells from pri- stem cells from rat blastocysts. Cell 135, 1287 –1298.
mary human fibroblasts with only Oct4 and Sox2. Nat. (doi:10.1016/j.cell.2008.12.007)
Biotechnol. 26, 1269–1275. (doi:10.1038/nbt.1502) 88 Li, P. et al. 2008 Germline competent embryonic stem
70 Li, W. et al. 2009 Generation of human-induced pluripo- cells derived from rat blastocysts. Cell 135, 1299–1310.
tent stem cells in the absence of exogenous Sox2. Stem (doi:10.1016/j.cell.2008.12.006)
Cells 27, 2992–3000. (doi:10.1002/stem.240) 89 Kawamata, M. & Ochiya, T. 2010 Generation of geneti-
71 Ichida, J. K. et al. 2009 A small-molecule inhibitor of cally modified rats from embryonic stem cells. Proc. Natl
tgf-Beta signaling replaces sox2 in reprogramming by Acad. Sci. USA 107, 14 223 –14 228. (doi:10.1073/pnas.
inducing nanog. Cell Stem Cell 5, 491 –503. (doi:10. 1009582107)
1016/j.stem.2009.09.012) 90 Tong, C., Li, P., Wu, N. L., Yan, Y. & Ying, Q. L. 2010
72 Zhou, H. & Ding, S. 2010 Evolution of induced pluri- Production of p53 gene knockout rats by homologous
potent stem cell technology. Curr. Opin. Hematol. 17, recombination in embryonic stem cells. Nature 467,
276– 280. (doi:10.1097/MOH.0b013e328339f2ee) 211 –213. (doi:10.1038/nature09368)
73 Maherali, N. & Hochedlinger, K. 2009 Tgfbeta signal 91 Kim, K. et al. 2010 Epigenetic memory in induced
inhibition cooperates in the induction of iPSCs and pluripotent stem cells. Nature 467, 285–290. (doi:10.
replaces Sox2 and cMyc. Curr. Biol. 19, 1718– 1723. 1038/nature09342)
(doi:10.1016/j.cub.2009.08.025) 92 Mikkelsen, T. S. et al. 2008 Dissecting direct reprogram-
74 Lyssiotis, C. A. et al. 2009 Reprogramming of murine ming through integrative genomic analysis. Nature 454,
fibroblasts to induced pluripotent stem cells with 49–55. (doi:10.1038/nature07056)

Phil. Trans. R. Soc. B (2011)


Review. Stem cells and small molecules J. A. Efe & S. Ding 2221

93 Silva, J., Barrandon, O., Nichols, J., Kawaguchi, J., 95 Ieda, M., Fu, J. D., Delgado-Olguin, P., Vedantham, V.,
Theunissen, T. W. & Smith, A. 2008 Promotion of repro- Hayashi, Y., Bruneau, B. G. & Srivastava, D. 2010 Direct
gramming to ground state pluripotency by signal reprogramming of fibroblasts into functional cardiomyo-
inhibition. PLoS Biol. 6, e253. (doi:10.1371/journal. cytes by defined factors. Cell 142, 375–386. (doi:10.
pbio.0060253) 1016/j.cell.2010.07.002)
94 Efe, J. A., Hilcove, S., Kim, J., Zhou, H., Ouyang, K., 96 Kim, J., Efe, J. A., Zhu, S., Talantova, M., Yuan, X.,
Wang, G., Chen, J. & Ding, S. 2011 Conversion of Wang, S., Lipton, S. A., Zhang, K. & Ding, S. 2011
mouse fibroblasts into cardiomyocytes using a direct Direct reprogramming of mouse fibroblasts to neural
reprogramming strategy. Nat. Cell Biol. 13, 215–222. progenitors. Proc. Natl Acad. Sci. USA 108, 7838–
(doi:10.1038/ncb2164) 7843. (doi:10.1073/pnas.1103113108)
Downloaded from https://royalsocietypublishing.org/ on 26 May 2021

Phil. Trans. R. Soc. B (2011)

You might also like