Está en la página 1de 12

REVIEWS Nature Reviews Molecular Cell Biology | AOP, published online 15 August 2005; doi:10.

1038/nrm1713

THE MOLECULAR REPERTOIRE


OF THE ‘ALMIGHTY’ STEM CELL
Craig E. Eckfeldt*, Eric M. Mendenhall* and Catherine M. Verfaillie
Abstract | Stem cells share the defining characteristics of self-renewal, which maintains or
expands the stem-cell pool, and multi-lineage differentiation, which generates and
regenerates tissues. Stem-cell self-renewal and differentiation are influenced by the
convergence of intrinsic cellular signals and extrinsic microenvironmental cues from the
surrounding stem-cell niche, but the specific signals involved are poorly understood.
Recently, several studies have sought to identify the genetic mechanisms that underlie the
stem-cell phenotype. Such a molecular road map of stem-cell function should lead to an
understanding of the true potential of stem cells.

PLURIPOTENT Multicellular organisms such as humans develop from monkey5,6, however, in this review we will focus on the
The ability to give rise to all PLURIPOTENT stem cells that are present in the INNER CELL pluripotent ESCs from mouse and human.
embryonic tissues, but not MASS (ICM) of the blastocyst and that generate the tril- The self-renewal and multi-lineage differentiation
extra-embryonic tissues.
lions of mature cells that make up the adult individual. characteristics of stem cells from embryonic and most
Totipotent cells can give rise to
all embryonic and extra- Such pluripotent stem cells can not only divide to give adult sources make these cells uniquely suited for
embryonic (trophectodermal) rise to daughter pluripotent stem cells — so called regenerative medicine, tissue repair and gene therapy
tissues. self-renewing cell divisions — but they can also dif- applications. An increasing number of in vitro and
ferentiate to give rise to all the cells of the mesoderm, in vivo studies have shown that stem cells can reca-
INNER CELL MASS
A mass of pluripotent cells in
endoderm and ectoderm as well as germ cells (FIG. 1). pitulate embryonic and adult tissue development, and
the interior of the developing During development, pluripotent stem cells from the can therefore repair injured or congenitally defective
blastocyst that give rise to all ICM become increasingly restricted in their lineage tissues. However, the mechanisms that govern the self-
embryonic tissues. The potential and generate tissue-specific, MULTIPOTENT stem renewal and multi-lineage differentiation potential of
blastocyst is part of the pre-
cells. These multipotent stem cells give rise to progeny stem cells remain largely unknown. Although stem-
implantation-stage embryo and
consists of a hollow sphere of that comprise specific, mature tissue. Although this cell fate decisions might have a stochastic component7,
cells with a distinct outer hierarchical archetypal model of stem-cell biology is increasing evidence indicates that extrinsic signals from
trophectoderm layer and an generally the rule, recent reports have described the the stem-cell microenvironment, or niche, can con-
inner cell mass. persistence of stem cells with less lineage-restricted verge on intrinsic cellular signals to regulate stem-cell
differentiation potential into post-natal life1 BOX 1. proliferation and cell-fate decisions. Various molecu-
Department of Medicine When isolated from the blastocyst in vitro, the lar techniques have been used to reveal the molecular
and Stem Cell Institute, pluripotent stem cells of the ICM can be maintained regulation of stem-cell fate decisions BOX 2.
University of Minnesota,
in culture as embryonic stem cell (ESC) lines. Murine It has been postulated that, although each unique
Minneapolis, Minnesota,
USA. (m)ESCs were first isolated in 1981 REF. 2 and are the stem cell can be characterized by the expression of
*These authors contributed most extensively characterized pluripotent cell type, a specific set of genes, the defining self-renewal and
equally to this work. and human (h)ESCs were isolated and characterized multi-lineage differentiation characteristics of stem
Correspondence to C.M.V. in the late 1990s3. Primordial germ cells (PGCs), which cells are encoded by a shared set of genes that are
e-mail: verfa001@umn.edu
doi:10.1038/nrm1713
are derived from the GENITAL RIDGE of early embryos, are expressed by all distinct stem-cell populations, and
Published online another type of pluripotent cell4. There are also ESC that therefore represents a conserved stem-cell molec-
15th August 2005 lines that have been derived from chicken and rhesus ular signature8–10. Several studies have examined the

2 | ADVANCE ONLINE PUBLICATION www.nature.com/reviews/molcellbio


REVIEWS

Molecular signature of embryonic stem cells


Transcription profiling has revealed that most dif-
ferentiated cell types express only 10–20% of their
genes, consisting of a mix of ubiquitous housekeep-
ing genes and tissue-specific genes11. By contrast,
30–60% of genes are expressed in ESCs11–14. These
data might be consistent with the theory of stem-cell
‘priming’ — the hypothesis that stem cells express many
different lineage-specific genes at low levels15–18. There
is evidence to indicate that much of the chromatin of
embryonic and adult stem cells is in an open, accessible
state, which might allow the promiscuous expression of
lineage-specific genes, and that epigenetic modifications
of chromatin structure and/or methylation of DNA leads
to a more restricted gene-expression pattern concomitant
with lineage differentiation19. It is possible that such low-
level transcription enables the rapid regulation of genes
that is required for differentiation during development
or following injury, by maintaining a transcriptionally
permissive chromatin structure.
Furthermore, the low-level expression of multiple
lineage-specific surface receptors in stem cells might
allow the cell to detect a wide range of extracellular sig-
nals to respond to complex microenvironmental cues.
However, for many lineage-specific transcripts found in
stem cells, no corresponding protein can be detected,
which indicates that gene expression is either post-
transcriptionally repressed or that transcript levels
do not reach a critical threshold until the cell receives
intrinsic and/or extrinsic cues to differentiate. Therefore,
the theory that low-level transcription allows the cells to
sample the microenvironment might not be correct.
) d)
Murine embryonic stem cells. Several studies have
Figure 1 | The stem-cell hierarchy. The totipotent zygote formed by the fusion of egg and
sperm divides to form the inner cell mass (ICM) and the extra-embryonic (EE) tissue of the
evaluated the TRANSCRIPTOME of mESC lines. Most of
blastocyst. When isolated from the blastocyst in vitro, the cells of the ICM can be maintained in these have compared the gene-expression profiles of
culture as pluripotent embryonic stem cell (ESC) lines. During the development of the embryo, single mESC lines with that of adult stem cells, mature
the pluripotent stem cells in the ICM become increasingly restricted in their lineage potential differentiated cells and TROPHECTODERM. For instance,
and generate tissue-specific, multipotent stem cells. These include epidermal stem cells (bulge comparison of the mESC line CCE with adult haemat-
cells) that form skin and hair, haematopoietic stem cells in the bone marrow that give rise to all opoietic stem cells (HSCs), neural stem cells (NSCs)
haematopoietic cells, neural stem cells in the subventricular zone of the brain, gastrointestinal
and mature blood cells identified several transcripts,
stem cells that are located in the crypt of the small intestine, oval cells that give rise to liver (not
shown), and mesenchymal stem cells that reside in the bone marrow and can form bone, including the HOMEODOMAIN TRANSCRIPTION FACTOR Oct4
stromal cells and adipocytes (not shown)88,115. (also known as Pou5F1 or Oct3/4)8, that were enriched
in mESCs, whereas a study that compared the gene-
expression profiles of the mESC line R1 with trophec-
toderm and fibroblasts identified 124 ESC-enriched
gene-expression pattern of both embryonic and adult genes, which also included Oct4 REF. 20. A large scale
stem-cell populations. Although these analyses have EST SEQUENCING project that analysed 19 different tissues
provided some insights into the genetic mechanisms including early mouse embryos, mESCs, newborn
MULTIPOTENT that are responsible for the stem-cell phenotype, there organs and adult stem-cell populations identified 75
The ability to give rise to the are inconsistencies in the resultant lists of ‘stemness’ genes that were expressed specifically by mESCs21.
diverse cell types of one or a few genes. So far, it is not known whether these inconsist- Oct4 and another homeodomain transcription factor,
tissues.
encies are due to technical disparities, or whether these Nanog, are among the functionally characterized genes
GENITAL RIDGE results signify that ‘stemness’ is not defined by a unique that are crucial to the mESC molecular signature22–24. In
The bilateral structures in the set of genes. In this review, we focus on the molecu- the absence of Oct4, mESCs TRANSDIFFERENTIATE into tro-
developing embryo that give lar signature of embryonic and adult stem cells and phectodermal cells22, whereas loss of Nanog results in an
rise to the gonads. their microenvironmental niches as a means of better increase in extra-embryonic endodermal transcripts24.
understanding the stem-cell phenotype. We explain Overexpression of Nanog allows mESC growth in the
TRANSCRIPTOME
The entire transcriptional
how these datasets can be used to further unravel absence of leukaemia inhibitory factor (LIF)23 — a
repertoire of a cell or cell the molecular regulation of stem cells, which will be factor that is required for the maintenance of mESC
population. required to exploit their full therapeutic potential. lines — whereas overexpression of Oct4 induces the

NATURE REVIEWS | MOLECULAR CELL BIOLOGY ADVANCE ONLINE PUBLICATION | 3


REVIEWS

Box 1 | Adult stem-cell plasticity* Human embryonic stem cells. Several studies have
compared the transcriptome of hESC lines with differ-
Adult stem cells are thought to be multipotent, but not pluripotent like embryonic ent populations of more mature cells and mESCs12,13,28.
stem cells (ESCs). However, in the past few years, more than 300 reports have In the most extensive study, the transcriptomes of six
indicated that adult stem cells might possess developmental capabilities that different hESC lines were compared with universal
resemble those of more immature, pluripotent cells, similar to ESCs. RNA29. This study identified 92 genes that showed
The main criticism regarding the claims of adult STEMCELL PLASTICITY is that most
increased levels of expression in all 6 hESC lines,
studies that describe such plasticity do not fulfill the criteria commonly used to
including OCT4, NANOG and TDGF1. Only 15 of the
describe stem cells: For instance, most studies published so far have not
92 genes were classified as ‘unknown’, which is much
definitively proved that the greater potency of adult stem cells can be ascribed to a
less than other studies12,13,28, perhaps owing to an
single cell that can differentiate into the tissue of origin and one or more
additional tissues. Furthermore, most studies have equated differentiation with under-representation of unknown genes on the arrays
the acquisition of morphological and phenotypic characteristics of a novel cell used. By comparing the gene-expression profile of
type, but have not proven the functionality of the resulting cells. Similarly, few, if three independent hESC lines, Abeyta et al. found that
any, studies have shown that the adult stem cell can robustly repopulate not only 52% of all genes examined were expressed in all three
the tissue from which it originates but also another tissue. cell lines11. Most of these 7,385 genes are ubiquitously
expressed, but tissue-specific genes that have been
There are four plausible explanations for the observed plasticity of adult implicated in stem-cell self-renewal and pluripotency,
stem cells: such as OCT4, SOX2 and TDGF1, were also expressed
• The apparent differentiation of an adult stem cell to a cell lineage other than the
in all three hESC lines11. A comparison of all the gene-
tissue of origin could be due to contamination of the population by a stem cell or
expression profiles of hESC lines described so far iden-
progenitor cell from the second tissue of origin.
tify LIN28, OCT4, NANOG, DNMT3B, TGIF, TDGF1,
• Fusion between donor and recipient cells, as occurs in heterokaryons, with CHEK2, GDF3, GJA1 and FLJ21837 among others, as
silencing of the genetic programme of one of the two cells. There is evidence that being expressed in all these lines11,13,14,28–30.
fusion can occur in vitro and in vivo.
• Stem cells might dedifferentiate and then redifferentiate, or might be Cross-species embryonic stem-cell comparisons.
reprogrammed, in a manner similar to that found in other species (that is, Although human and mouse ESCs show many simi-
blastema formation in amphibians), during metaplasia, or as occurs in somatic larities, these two cell types also have several differ-
cell nuclear transplantation. ences. For instance, both hESCs and mESCs are
• Pluripotent stem cells generated before or after gastrulation might persist typically cultured in the presence of mouse embryonic
during development into adulthood. fibroblasts (MEFs), but hESCs, unlike mESCs, do not
require exogenous LIF-mediated JAK–STAT3 activa-
*REFS 110–114 specifically address adult stem-cell plasticity.
tion to maintain their pluripotency in culture, although
STAT3 might be activated by other means in hESCs25,27.
Maintenance of the pluripotent state of mESC lines also
TROPHECTODERM differentiation of mESCs into endoderm and meso- requires bone morphogenetic protein-4 (BMP4)31; by
The outer portion of the
derm. Nanog, Oct4 and LIF-mediated JAK–STAT3 contrast, BMP4 induces trophoblast differentiation
blastocyst that gives rise to the
embryonic portion of the activation therefore represent independent pathways in hESCs32. Unlike mESCs, hESCs can be maintained
placenta. that maintain mESC pluripotency and self renewal25. MEF-free using fibroblast growth factor-2 (FGF2)33.
Mitsui et al. performed an IN SILICO DIFFERENTIAL DISPLAY This indicates that some of the important signal path-
HOMEODOMAIN
and identified 20 genes that were specifically expressed ways that are required for pluripotency differ between
TRANSCRIPTION FACTOR
A transcription factor that
in mESCs and in mouse pre-implantation embryos24. human and mouse ESCs. These differences might
contains a homeodomain DNA- These included Oct4 and Nanog; as well as zinc finger explain the discordance between human and mouse
binding domain. protein-42 (Zfp42, also known as Rex1), the transcrip- ESC expression profiles — comparisons of several
tion factor Utf1, the constitutively active Ras protein ES- important studies show that only 13–55% of transcripts
EST SEQUENCING
cell-expressed Ras (Eras), and the growth factor Tdgf1 that are enriched in mESC lines are also enriched in
The sequencing of short
segments of expressed genes (also known as Cripto); all these genes were previously hESC lines but, by contrast, comparisons between
(expressed sequence tags or known to be important for ESC self-renewal24. When different hESC lines are 85–99% concordant11,12,25,29.
ESTs) present in cDNA libraries OCT4 is found in a complex with the transcription fac- Furthermore, some mESC-enriched transcripts are
that can be used for gene tor SOX2, it upregulates its own expression as well as not expressed in all of the hESC lines examined by
cloning or to show which genes
are present in a cell population.
the expression of Fgf4, Zfp42 REFS 24,26 and Nanog (S. expression profiling so far, despite being implicated
Yamanaka, personal communication), which identi- in self-renewal and pluripotency. For example, ZFP42
TRANSDIFFERENTIATE fies Oct4 as a key regulator of ESC genes (reviewed in is not expressed in the hESC line HES4, STAT3 is
Differentiation of one cell type REFS 26,27). The downstream targets of Nanog and Zfp42 not expressed in H9 cells, and SOX2 and ESG1 are not
directly to another cell type
have yet to be reported. Importantly, none of the genes expressed in other hESC lines analysed in these experi-
without dedifferentiation to a
more primitive intermediate. mentioned above were identified through global gene- ments11,29. Therefore, the expression of Zfp42, Oct4 and
expression profiling. Instead, most were identified as Sox2, which form a transcriptional feedback loop, is
STEMCELL PLASTICITY downstream targets of Oct4. Ongoing studies are now upregulated in mESCs, whereas only OCT4 is consist-
The apparent ability of a stem/ using candidate-gene approaches to evaluate the role of ently expressed in hESCs. This might indicate that, in
progenitor cell fated to a
particular tissue to acquire a
these and other genes and pathways that are involved hESCs, OCT4 does not require the presence of SOX2
differentiated phenotype of a in the maintenance, cell signalling, metabolism and cell to activate transcription. Therefore, the differences in
different tissue. cycle of ESCs25. gene expression that have been noted between mESCs

4 | ADVANCE ONLINE PUBLICATION www.nature.com/reviews/molcellbio


REVIEWS

diverse array of all mature tissues during embryonic


Box 2 | Common techniques for gene-expression analysis
development. A subset of their progeny, so-called
Northern blotting, RPA, RT-PCR adult or tissue-specific stem cells, retains self-renewal
(Individual mRNA transcripts) and multi-lineage differentiation potential into adult-
hood. Such adult stem cells, however, have much less
self-renewal capacity compared with ESCs and are
Subtractive hybridization Population A Population B Gene-expression arrays
not pluripotent. Adult stem cells have been identified
mRNA mRNA in many tissues, where they continuously generate
Population B Population A
cDNA cDNA Labelled and regenerate mature tissues either as part of nor-
cDNA or mal physiology or in response to injury TABLE 1.
Population A Population B cRNA
mRNA mRNA Haematopoietic, neural, epidermal and gastrointesti-
nal stem cells have been phenotypically characterized,
and this has facilitated the analysis of their gene-
Macroarray Microarray
Serial analysis of
expression patterns. However, complex organs with
cDNA: gene expression numerous cell types such as the lung or kidney might
mRNA not be maintained by a single type of stem cell, but by
hybrids
several stem-cell types that have yet to be identified.
Nylon Glass slide
membrane In general, the lack of phenotypic markers that can
Gene-specific 14mers be used to purify most tissue-specific stem cells and
the lack of appropriate assays to assess the function of
mRNA mRNA Ligation into sequencing Measurement of labelled cDNA many adult stem cells have impeded gene-expression
expressed expressed vectors to determine the or cRNA intensity at a specific
in A > B in B > A analysis.
relative abundance of location on an array to determine
expressed sequences abundance of mRNA transcripts
Haematopoietic stem cells. The HSCs that reside in the
Traditionally, individual transcripts were identified by hybridizing radio-labelled bone marrow microenvironment during adult life are
complementary nucleic-acid probes in Northern blot or RNase protection assays the best-characterized adult stem cells, and therefore
(RPAs). Alternatively, a complementary DNA (cDNA) copy of the entire serve as the model for stem-cell biology (FIG. 2). HSCs
transcriptome can be generated, followed by the identification of specific genes by are also at the forefront of our understanding of the
PCR or direct sequencing of cDNA. These analyses represent a gold standard for gene- molecular regulation of the adult stem-cell phenotype.
expression analysis, but are difficult to perform on a large scale. Investigation of the function and molecular regulation
Subtractive hybridization (SH) (see figure, left) is used to characterize transcripts of HSCs and their progeny, haematopoietic progenitor
that are differentially expressed between two cell populations. The cDNA synthesized cells (HPCs), has been facilitated by the development
from one population is mixed with mRNA from another population, which results in of monoclonal antibodies to cell-surface antigens and
the formation of cDNA–mRNA hybrids of transcripts that are expressed in both
FLUORESCENCE ACTIVATED CELL SORTING, which allow purifi-
populations. More abundant mRNA remains unhybridized, and can then be isolated
cation of murine (m)HSCs to near homogeneity34. The
and characterized. SH does not require specific sequence information and generates a
advent of molecular techniques that make it possible
global picture of differential gene expression, but provides little information
regarding the relative abundance of transcripts.
to perform gene-expression analysis using a few or
Serial analysis of gene expression (SAGE; see figure, centre) involves digesting cDNA even single cells has enabled a detailed analysis of the
to generate short (~14 base pair), gene-specific sequence tags that represent the extrinsic and intrinsic signals that govern HSC-fate
transcriptome of a cell population, followed by sequencing and quantitation of these decisions.
tags. SAGE does not require prior knowledge of target sequences and provides a Most cell-intrinsic genes that define HSCs were
quantitative and global analysis of gene expression. identified before the advent of genome-wide micro-
cDNA macroarray analysis (see figure, right) is performed by hybridizing cDNA array analysis, owing to their involvement in clonal
from a cell population with gene-specific cDNA probes that are immobilized onto genetic aberrations in haematopoietic malignancies.
nylon membranes. cDNA and oligonucleotide microarray analyses are conceptually For instance, recurrent chromosomal translocations
similar to macroarrays, but are typically performed by synthesis and hybridization of in human acute T-cell leukaemias that deregulate
complementary (c)RNA to gene-specific oligonucleotides (25–60mers) that are the expression or function of genes such as NOTCH1
immobilized on glass slides. For each assay, gene-expression data are extracted by REF. 35, TAL1 (also known as SCL)36,37 and LMO2
normalization and quantitation of radioactive or fluorescent tags that have been REF. 38 result in proliferation of primitive haemat-
incorporated into the test cDNA or cRNA to determine the abundance of specific opoietic progenitor cells in the absence of normal
sequences. Global gene-expression profiling using array technology allows assessment differentiation. These observations led to the subse-
of the differential expression of tens of thousands of transcripts from a small number quent characterization of these genes as important
of input cells, but it is limited by the requirement of prior sequence information for transcriptional regulators involved in the initial stages
probe design. of non-malignant haematopoietic development in ani-
mal models39–41. Notch1 is a transmembrane surface
and hESCs might identify differences in the signal receptor that functions as a developmental regulator
pathways that are required for pluripotency in differ- of cell-fate decisions through interactions with sur-
cRNA
ent species. face-expressed Notch ligands on neighbouring cells.
A complementary RNA
molecule that hybridizes with a
On ligand binding, Notch1 is proteolytically processed
specific messenger RNA Molecular signature of adult stem cells thereby liberating its intracellular domain that subse-
sequence. Pluripotent stem cells are crucial for generating the quently translocates to the nucleus and functions as a

NATURE REVIEWS | MOLECULAR CELL BIOLOGY ADVANCE ONLINE PUBLICATION | 5


REVIEWS

Table 1 | Mammalian adult stem cells


Tissue Stem cell Niche Progeny References
Blood Haematopoietic Endosteal surface of All myeloid and lymphoid 7–10,16–18,34–63,
stem cell bone marrow blood lineages 86,90,91,99,100
Mesenchyme Mesenchymal stem Within bone marrow Bone, cartilage, tendon, 116
cell cavity smooth muscle, adipose
tissue and stroma
Brain Neural stem cell/ Subventricular zone Neurons, glial cells and 8–10,57,64–70,88
neurosphere and hippocampus oligodendrocytes
Gut Crypt cell/gut Gut crypt Enterocytes, 74,75
epithelial progenitor enteroendocrine cells,
goblet cells and Paneth cells
Heart Cardiac progenitor Not determined Cardiac myocytes 117
Liver Oval cell Terminal biliary ductule Hepatocytes and 118–120
cholangiocytes
Pancreas Pancreas-derived Not determined Pancreatic endocrine and 121,122
multipotent acinar cells
precursors
Skeletal muscle Satellite cell Between sarcolemma Myocytes/myofibrils 123
and basil lamina
Skin/hair Bulge cell Bulge in hair follicle Epidermis, hair follicles and 71–73
sebaceous glands
Male germ cells As spermatogonia Basement membrane Sperm 124
of seminiferous tubule

transcription factor. Overexpression of a constitutively genes have been identified on the basis of phenotypic
active form of Notch1 in murine HSCs creates clonal observations such as quiescence; for example, Cdkn1a
IN SILICO DIFFERENTIAL multipotent haematopoietic cell lines that can recon- (also known as p21), which was found to be selectively
DISPLAY
stitute the haematopoietic system without evidence of expressed in the most primitive haematopoietic cells,
The use of computer algorithms
to determine differential
leukaemic transformation39. Likewise, stimulation of and was subsequently shown to be crucial for mainte-
expression of transcripts from HSCs through the Notch ligands, Delta and Jagged, nance of the mHSC pool55.
gene-expression databases. results in an expansion of primitive murine and human These studies provide insight into the molecular
in vivo repopulating cells consistent with a role for regulation of the HSC phenotype. Specifically, long-
FLUORESCENCE ACTIVATED
Notch signalling in stem-cell self-renewal42–45. Further term maintenance of the stem-cell pool requires qui-
CELL SORTING
Automated, high-speed sorting insights into the genetic control of HSC function come escence of HSCs, and therefore Bmi1, a transcriptional
of cell populations based on the from reverse-transcriptase polymerase chain reaction repressor, and Cdkn1a, a cell-cycle inhibitor, probably
presence of intrinsic fluorescent (RT-PCR)-based screening methods that are designed regulate this aspect of the HSC phenotype. Expansion
labels such as GFP expression, to determine the expression patterns of specific genes of the HSC pool requires symmetric self-renewing cell
or extrinsic fluorescent labels
such as monoclonal antibodies
and gene families in subsets of human and murine divisions that give rise to two daughter cells that retain
conjugated to fluorochromes. haematopoietic stem and progenitor cells. Several HSC function. Hoxb4, Wnt signalling and Notch sig-
RT-PCR-based approaches have focused on crucial nalling represent the best-characterized mechanisms
HOMEOBOX HOX GENE regulators of embryonic development such as the thought to drive symmetric self-renewal of HSCs.
FAMILY
HOMEOBOX HOX GENE FAMILY, POLYCOMB PCG GENE FAMILY and Early strategies to determine a more global genetic
A family of transcriptional 46–48
regulators that share a WNT GENE FAMILY . This approach has been effective programme of HSCs used subtractive hybridiza-
conserved homeobox DNA- in identifying genes that are selectively expressed in tion of highly purified murine fetal liver and murine
binding domain, and that are the most primitive subsets of haematopoietic cells and adult bone marrow HSCs in combination with high-
involved in the regulation of that functionally regulate HSC-fate decisions. Hoxb4 density cDNA macroarrays to delineate genes that
embryonic and adult
developmental fates.
is one of the most well-characterized genes involved are expressed in HSCs56. The presence of previously
in the self-renewal of human and murine long-term, characterized HSC-associated genes among those
POLYCOMB PCG GENE FAMILY repopulating HSCs49–51. Identified in a similar man- identified, such as the genes that encode cell-surface
Genes encoding a family of ner, both the polycomb gene Bmi1, as well as frizzled molecules FLT3 and CD34 as well as the transcrip-
proteins that form complexes
receptors that bind secreted Wnt ligands, were found tional regulator Runx1 gene, confirmed the validity of
that modify chromatin
structure and selectively repress to be expressed in primitive haematopoietic cells47,48 this experimental approach and indicated that genes
gene transcription. and were subsequently proven to have crucial roles in and signalling pathways that had not previously been
HSC self-renewal52,53. Other genes that are implicated implicated in haematopoietic development might be
WNT GENE FAMILY
in the HSC phenotype have been identified on the important in HSC behaviour56. To distribute the data
A family of genes that mediate
intercellular signalling through
basis of their roles in the development of mesoderm on the HSC-specific transcriptome and to foster the
secreted glycoprotein Wnt and its derivatives; for example, the morphogens sonic collaborations that are essential to approach a problem
ligands. hedgehog (SHH) and BMP4 REF. 54. And more HSC as complex as the genetic regulation of stem cells, these

6 | ADVANCE ONLINE PUBLICATION www.nature.com/reviews/molcellbio


REVIEWS

primitive cobblestone formation by mHSCs in vitro


(a surrogate readout for HSC-like cells) and in the
maintenance of mHSCs in vivo has recently been
Hoxb4
BMP4 Notch pathway demonstrated58, which supports the validity of this
Bmi1
Cdkn1a
SHH Wnt pathway analysis. The expression of this ligand–receptor pair
in HSCs indicates that HSC-fate decisions might
Genes/pathways associated with self-renewal be regulated in an autocrine or paracrine fashion.
Self-renewal and multi-lineage differentiation potential
Promiscuous gene expression of multiple lineage-specific transcripts Although comparisons of adult and fetal HSCs have
identified shared patterns of gene expression, there
is only partial concordance between gene expression
in adult and fetal HSCs (reviewed in REF. 59), which
Stem cell Progenitor cells Mature cells
might reflect fundamental differences between
B cell ontogenically distinct HSCs57,60.
CLP
A comparison between mHSCs and murine
T cell
multipotent progenitors (MPPs), which have only
NK cell limited self-renewal potential, found that genes that
MEP are implicated in the development and self-renewal
Erythrocyte
of mHSCs, such as Bmp4, Bmi1 and Notch1, were
HSC CMP Megakaryocyte expressed at higher levels in the HSCs61. Almost half
GMP
of the genes analysed on a 1,200 probe high-density
Granulocyte
cDNA macroarray were expressed at detectable levels
Monocyte in mHSCs, and differentiation to mMPPs and com-
mitted cells resulted in the upregulation of specific
lineage markers with concomitant loss of global gene
expression16. Similar results were seen in a more exten-
Genes/pathways associated with haematopoietic cell function sive analysis of HSCs, MPPs, common myeloid pro-
Gene expression of individual lineage-specific transcripts
genitors (CMPs) and common lymphoid progenitors
Globin genes
Gata1 (CLPs) — more than 40% of haematopoiesis-related
Epor
Mpo
Mpl
genes analysed, including HSC-specific as well as other
lineage-specific transcripts, were expressed in HSCs,
and this transcriptional promiscuity was lost during
lineage commitment17. Therefore, similar to ESCs,
HSCs have a transcriptionally accessible genome that
Figure 2 | The haematopoietic stem cell. Haematopoietic stem cells (HSCs) supply the becomes progressively more restricted coincident with
entire repertoire of mature blood cells for the lifetime of an organism. To accomplish this task, lineage differentiation (FIG. 2).
HSCs are endowed with self-renewal capacity that maintains and expands the stem-cell pool, In contrast to mHSCs and murine haematopoetic
and multi-lineage differentiation potential that produces the diverse components of the mature progenitor cells (HPCs), hHSCs have not yet been
haematopoietic system. As HSCs differentiate, they lose the ability to self-renew and become
purified to homogeneity on the basis of their surface
increasingly restricted in their lineage potential. Genes and proteins associated with the self-
renewal of HSCs (for example, Hoxb4 REFS 4951, Bmi1 REF. 53, Cdkn1a55, BMP4 REF. 54, phenotype. Therefore, defining the transcriptome of
SHH54, the Notch pathway39,42–45 and the Wnt pathway52) are highly expressed in HSCs, but hHSCs has been complicated by the greater heteroge-
are downregulated in committed progenitor and mature haematopoietic cells. HSCs also neity of hHSC and hHPC populations. Nevertheless,
promiscuously express multiple lineage-specific transcripts at low levels, however, comparison of the transcriptomes of highly puri-
differentiation is associated with the loss of this promiscuous transcript expression and fied mHSCs and the more heterogeneous human
selective expression of individual lineage-specific genes17.CLP, common lymphoid progenitor;
CD34+CD38– cells has identified approximately 40%
CMP, common myeloid progenitor; GMP, granulocyte monocyte progenitor; MEP,
megakaryocyte erythrocyte progenitor; NK, natural killer.
congruence. Considering the challenges of hetero-
geneity and comparative genomics8, this is probably
a conservative figure. It is probable that advances in
the ability to prospectively isolate human HSCs using
results were compiled in an open access, searchable strategies that enrich for HSCs, for example, the exclu-
stem-cell database (SCDb; see the Online links box)56. sion of rhodamine 123 REF. 62, or aldehyde dehydro-
Not long after the establishment of the SCDb, genase activity63, will show greater conservation of the
further expression-profiling studies were performed molecular signature for murine and human HSCs.
using different combinations of phenotypically and
ontogenically distinct murine HSCs and HPCs. Neural stem cells. Until recently, it was thought that
One analysis of transcripts enriched in mHSCs that few, if any, stem cells were present in the adult central
were derived from adult bone marrow identified nervous system (CNS). However, several studies have
several genes that were classified in the SCDb as now shown that NSCs persist into adulthood in the
genes expressed in HSCs derived from murine fetal subventricular zone of the lateral ventricles and the
liver 56,57. These included angiopoietin-1 (Angpt1) dentate gyrus of the hippocampus. These NSCs con-
and the angiopoietin receptor, Tek (also called Tie2). tinuously give rise to new neurons to replace those that
A functional role for Tek and Angpt1 in enhancing die in the normal aging process or in neurodegenerative

NATURE REVIEWS | MOLECULAR CELL BIOLOGY ADVANCE ONLINE PUBLICATION | 7


REVIEWS

disorders64. Although some phenotypic markers have cells and acid-secreting parietal cells74, and the expres-
been identified that allow selection of NSCs65, NSCs are sion profiles of small intestinal epithelial progenitor
most commonly isolated by culture as neurospheres cells (SiEPs) have been compared with Paneth cells75
— a heterogeneous population that also contains — each cell type was identified on the basis of its
more mature cells. Various studies have sought to precise location in the crypts of the gastric and small
define the expression profile of NSCs by comparing intestinal mucosa. Of the 147 genes thought to define
the gene-expression profiles of human and murine GEPs, 11 were also upregulated in SiEPs, and another
neurospheres66–68, murine retinal progenitor cells69, or 22 transcripts that were upregulated in SiEPs encode
NSC-containing neuroepithelial cells70 with those of functionally related family members or additional
more mature neural progenitors or mature neurons. subunits of multi-subunit complexes of transcripts
Confirming the validity of these studies, several genes enriched in GEPs75. This surprising degree of overlap
with known roles during CNS development, such as between the GEP and SiEP datasets indicates that
the transcriptional regulators Sox2, Pax6 and Lhx2, shared mechanisms probably regulate both SiEP
and the cell-cycle regulator Ccnd1 (also known as and GEP function. With advances in the phenotypic
Cyclin D1; REFS 6670) were shown to be enriched in characterization of adult stem cells, it is probable that
NSC-containing populations in these studies. And, more homogeneous stem and progenitor cells from
consistent with other stem-cell gene-profiling studies, the gastrointestinal tract will be isolated, and that
many genes that have no known role in neural develop- fewer background transcripts will obscure potentially
ment, and transcripts without any known function are relevant stem-cell-specific transcripts.
expressed in NSC-enriched populations9,66–70. Further
characterization of the function of these genes might Cross-tissue-specific stem-cell comparisons. Despite the
provide novel insights into the molecular regulation shortcomings in gene-expression evaluation for many
of NSCs and their progeny. However, because of the adult stem-cell populations, comparison between the
considerable heterogeneity of isolated NSC-containing molecular signatures of HSCs, NSCs, bulge stem cells
cell populations, it is unlikely that the true transcrip- and gastrointestinal stem cells have identified several
tome of NSCs is reflected in these studies. Advances classes of genes that might be expressed in all tissue-
in the methods used to isolate pure populations of specific stem cells. For instance, 15–30% of genes that
NSCs from the brain or from ESCs will lead to a bet- are expressed specifically in HSCs, but not in HPCs,
ter characterization of the NSC molecular signature. are also specifically expressed in NSC-enriched
Towards this goal, human NSCs have been purified to populations8,9,57; and 14% of genes that are expressed
near homogeneity65, but the transcriptional profile of in bulge stem cells are also expressed in HSCs and
such cells has yet to be determined. NSCs71. Among these are genes involved in Wnt sig-
nalling (for example, Tcfs and Fzd7), Notch signalling
Epidermal stem cells. The stem cells that generate skin (for example, Hes1), adhesion (for example, Itga6) and
epithelium and skin appendages (known as bulge cells) transcriptional regulation (for example, Bmi1).
are found in a niche in the bulge of the root sheath of
hairs (FIG. 1). Bulge cells give rise to both epidermis and The stem-cell niche
hair follicles in transplant experiments, and develop Stem cells exist in vivo in a complex microenviron-
into colonies of undifferentiated cells in vitro71. Murine ment, or niche, which is composed of differentiated
bulge cells can be isolated using a GFP label that is somatic cells and extracellular matrix, as well as stem
expressed under the control of the ‘bulge-preferred’ cells and their progeny. The niche provides factors
keratin-15 promoter in combination with the expres- that maintain the self-renewal ability of stem cells and
sion of CD34 and integrin-α6 cell surface antigens, or prevent their differentiation. Most insights into the role
by the retention of bromodeoxyuridine in these qui- of stem-cell niches come from studies on Drosophila
escent cells (label retaining cells)71–73. Whether these melanogaster germ stem cells (GSCs; reviewed in
approaches purify epidermal stem cells to homogene- REFS 76,77), which interact with specialized somatic cells
ity, or whether there are several distinct stem cells in in the niche — known as cap cells (during oogenesis
the bulge is not yet clear71. Expression profiling of bulge in the ovary)78 or hub cells (during spermatogenesis in
stem cells by two independent laboratories has iden- the testes)79–81 (FIG. 3). This physical interaction main-
tified 97–157 genes that are differentially expressed tains the undifferentiated state of the stem cell and is
in bulge cells when compared with differentiated mediated through a cadherin–catenin pathway82. The
keratinocytes, with 80–90% concordance between the GSC-cap/hub-cell interaction also regulates symmet-
different studies71,72. These genes include FGF1 and its ric versus asymmetric GSC divisions by polarizing the
receptor, transforming growth factor-β (TGFβ) activa- stem cell and affecting the orientation of the mitotic
tors, BMP and Wnt pathway inhibitors71,73, which are spindle and the segregation of differentiation and
all known to be involved in the regulation of epidermal stem-cell determinants in daughter cells83–85.
stem-cell proliferation and differentiation. Similar to GSCs, mammalian adult stem cells reside
in niches that protect the ability of the stem cell to self-
Gastrointestinal stem cells. The gene-expression pro- renew and that prevent differentiation. Niches have been
files of gastric epithelial progenitor (GEP) cells have identified for epidermal stem cells, gastrointestinal stem
been compared with those of more mature zymogen cells, HSCsand NSCs. In common with GSC niches,

8 | ADVANCE ONLINE PUBLICATION www.nature.com/reviews/molcellbio


REVIEWS

a Germ stem-cell niche b Haematopoietic stem-cell niche instance, subtractive hybridization was used to com-
pare the transcriptome of a fetal-liver stromal cell
CXCL12 line (known as AFT024) that supports HSC main-
(secreted by tenance in vitro, with fetal-liver-derived stromal cell
osteoblasts) CXCL12/CXCR4 Wnts/Frizzled
Terminal
filament
lines that do not support the maintenance of HSCs.
PTH (from Wnts Genes that were expressed in the supportive AFT024
outside niche) (secreted
Cadherin– HSC by osteoblasts) cell line were compiled in an internet-based stromal
catenin
junctions cell database (StroCDb; see the Online links box).
PTH/PTHR1 Kit Notch1
Cap Integrins
Among the genes that were preferentially expressed
Jag1
KitI in AFT024 were Kitl, Bmp2, Wnt5a and Dll1, as well
ECM
as many secreted and surface molecules with no
GSC GSC
BMSC known haematopoietic function91.
Although adult stem cells can undergo asymmetric
and symmetric divisions, how physical interactions
between somatic stem cells and their niches regulate
Endosteal bone surface these cell divisions, as has been described for GSCs92,
Figure 3 | Molecular regulation of stem-cell niches. a | Components of the stem-cell niche
remains largely unknown. As other roles of the
provide cell–cell and cell–matrix interactions that are crucial for protecting the integrity and D. melanogaster GSC niche seem to be conserved in
function of the resident stem cells. Cadherin–catenin-mediated interactions between somatic mammalian adult stem cells, the mechanisms that
cap cells (cap) and the Drosophila melanogaster germ stem cells (GSCs) in the ovary maintain underly asymmetric versus symmetric cell divisions
the stem cells in an undifferentiated state. They also regulate symmetric versus asymmetric cell will probably also be conserved from fly to man and
divisions by polarizing the mitotic spindle and facilitating the partitioning of the cellular contents from germ stem cell to adult stem cell.
into the daughter cells. b | The functional interactions between stem cells and their niches have
been conserved from flies to mammals. Within the extracellular matrix (ECM) at the endosteal
surface of the bone marrow cavity, osteoblasts and other bone-marrow stromal cells (BMSCs) A conserved stem-cell molecular signature?
regulate haematopoietic stem cells (HSCs) through secreted signals as well as by cell–cell and It has been suggested that the self-renewal and multi-
cell–matrix interactions. Chemokines such as CXCL12 provide a signal to recruit CXCR4- lineage differentiation characteristics of stem cells is
expressing HSCs to the appropriate niche, whereas ECM components interact with HSC- the result of a genetic programme that is common to
expressed integrins to retain the stem cells. Niche cells also provide haematopoietic cytokines stem cells of all origins, and that stem cells therefore
such as Kitl. Further regulation of HSCs by the HSC niche depends on activation of Notch
share a conserved molecular signature. Two seminal
signalling by Jagged ligands (Jag1), and Wnt signalling by secreted Wnt ligands45,48. The HSC
niche itself is regulated by paracrine factors such as parathyroid hormone (PTH) and its
studies have compared the gene-expression profiles of
receptor (PTHR1) that can alter the cellular composition and size of the niche, and thereby murine ESCs, HSCs and neurospheres8,9. In one study,
regulate HSC numbers45. Part a of the figure was modified with permission from REF. 78 © 283 genes were expressed by all three stem-cell popu-
(2000) American Association for the Advancement of Science. lations8 and in the second, 216 genes were expressed
by all three stem-cell sources9. These genes comprised
both known genes as well as genes without an anno-
adult stem-cell niches contain extracellular matrix tated function. However, a comparison of these two
components and cell-surface ligands that ‘bind’ the stem independent studies, which used seemingly compara-
cell to the niche by cadherin and integrin interactions71. ble input cell populations and methodologies, revealed
Moreover, niche-derived factors that regulate stem-cell only six common stem-cell genes that were identified
fate are conserved from GSC niches to most adult stem- in both studies10 TABLE 2. In a third study, investigators
cell niches. For instance, in the HSC niche, osteoblasts compared the gene-expression profiles of murine ESCs,
express Notch ligands that support HSC self-renewal, as neurospheres and retinal progenitor cells, and found
shown by the promotion of the self-renewal of HSCs45,86 that 385 genes were expressed in all three cell types10.
owing to overexpression of activated parathyroid hor- When compared with the first two studies however,
mone receptors (which upregulates Notch ligand expres- only one gene, integrin-α6 (Itga6) was expressed in all
sion). A similar mechanism operates in NSC niches87. stem-cell populations TABLE 2.
Other mechanisms through which niches regulate Possible explanations for the discrepancies observed
the fate of adult stem cells is by the secretion of Wnts include technical and methodological differences among
and soluble Frizzled receptors48,71,88, by the production the three analyses, such as differences in the stem-cell
of members of the TGFβ/BMP family71,86 and by the isolation methods, the type of gene arrays used and the
production of Kitl (also known as stem cell factor)71,89,90 type of computational analyses used to identify shared
(FIG. 3). stem-cell genes. In fact, the number of shared ‘stemness’
Identification of the precise location of epidermal genes increased substantially when the data were com-
and gastrointestinal stem cells has facilitated analy- pared using the same algorithms to define differential
sis of the gene-expression profile of the surrounding gene expression59. However, as a much higher degree
niche cells. With the identification of the precise of congruence was seen when gene expression in ESCs
location of HSCs in the bone marrow, such studies (n = 332) or NSCs (n = 236) was compared between
are now also possible for HSC-niche cells. Another the studies (p < 10–8), methodological differences
method to identify niche-derived extrinsic regula- alone might not fully explain the failure to find more
tors of stem-cell fate is by the creation of stromal cell shared stem-cell genes10. Another explanation might be
lines that support stem-cell self-renewal in vitro. For that genes that confer stem-cell activity might not be

NATURE REVIEWS | MOLECULAR CELL BIOLOGY ADVANCE ONLINE PUBLICATION | 9


REVIEWS

Table 2 | Overlap of stem-cell-specific genes


Gene symbol* Gene description Functional annotation
Fortunel and Ramalho-Santos datasets
1110068L01Rik RIKEN cDNA 1110068L01 gene Not determined
1810009A15Rik RIKEN cDNA 1810009A15 gene Not determined
2410015N17Rik RIKEN cDNA 2410015N17 gene Not determined
2410080P20Rik RIKEN cDNA 2410080P20 gene Not determined
2410091N08Rik RIKEN cDNA 2410091N08 gene Not determined
2700084L22Rik RIKEN cDNA 2700084L22 gene Protein modification
2810436B06Rik RIKEN cDNA 2810436B06 gene Chloride transport
AA407558 Expressed sequence AA407558 Not determined
AA420417 Expressed sequence AA420417 Not determined
C81206 Expressed sequence C81206 Not determined
Cdkap1 CDK2-associated protein-1 Cell growth and/or maintenance
Chd1 Chromodomain helicase DNA binding protein-1 Chromatin assembly/disassembly
eIF4Ebp1 eIF4E binding protein-1 Insulin receptor signalling pathway
Etl1 Enhancer trap locus-1 Transcriptional regulation
Gfer Growth factor, Erv1 (S. cerevisiae)-like Not determined
Nol5 Nucleolar protein-5 Not determined
Rnf4 Ring finger protein-4 Transcriptional regulation
Sfrs3 Splicing factor, arginine/serine-rich 3 (SRp20) mRNA splice-site selection
Tead2 TEA domain family member-2 Transcriptional regulation
Tmk Thymidylate kinase Nucleotide biosynthesis
Trif-pending TRIF gene Not determined
Zfx Zinc finger protein X-linked Transcriptional regulation
Ivanova and Ramalho-Santos datasets
AI643885 Expressed sequence AI643885 Not determined
Cpx1-pending Metallocarboxypeptidase-1 Not determined
Laptm4b Lysosomal-associated protein transmembrane-4B Not determined
Lce-pending Long chain fatty acyl elongase Fatty acid elongation
Pkd2 Polycystic kidney disease-2 Cation transport
Fortunel and Ivanova datasets
Edr1 Early development regulator-1 Development
Tcf3 Transcription factor-3 Transcriptional regulation
Fortunel, Ramalho-Santos and Ivanova datasets
Itga6 Integrin-α6 Integrin signalling pathway
*Transcripts for the corresponding gene symbols were identified as more highly expressed in multiple adult and embryonic stem-cell
populations compared with their progeny using gene-expression microarrays, and they therefore represent conserved stem-cell-
specific genes. Each subheading lists the studies that were included for each section. The stem-cell populations analysed in these
studies were murine haematopoietic stem cells (mHSCs), murine embryonic stem cells (mESCs) and murine neural progenitor cells
(mNPCs) by Ivanova et al.8; mHSCs, mESCs and mNPCs by Ramalho-Santos et al.9; and mHSCs, mESCs and murine retinal
progenitor cells by Fortunel et al.10 This table is adapted from Fortunel et al.10 CDK, cyclin-dependent kinase; elF, eukaryotic
translation-initiation factor; S. cerevisiae, Saccharomyces cerevisiae.

represented on the oligonucleotide-based microarrays that shared stem-cell genes might be expressed only
that were used for gene-expression analysis10, therefore transiently and would therefore be difficult to detect
precluding the identification of important genes that in a homeostatic stem-cell population10. Furthermore,
are expressed in all stem cells. The use of subtractive stem-cell genes might not be expressed exclusively in
hybridization and serial analysis of gene expression stem cells, but might also be expressed — albeit at lower
(SAGE) that do not share these limitations would or higher levels — in differentiated cells, and therefore
circumvent this problem. It has also been suggested would be more difficult to identify by differential

10 | ADVANCE ONLINE PUBLICATION www.nature.com/reviews/molcellbio


REVIEWS

expression analysis. Consistent with this notion, only of expressed genes in stem-cell function. For instance,
4 of the 216 genes identified by Ramalho-Santos et al. highly purified mHSCs express low levels of mRNA for
as ‘stemness genes’, were not expressed in the differenti- lineage-specific genes before lineage commitment99, and
ated cell populations analysed by differential display9. fewer than 25% of genes that have been analysed have
Therefore, stem-cell function might not be imparted on consistent levels of both mRNA and protein expression
cells by a defined and limited set of master stem-cell- during myeloid cell differentiation100. Although tech-
specific genes or by a small number of pathways, but niques to define the proteome are less sensitive than
by the combined effect of the complex upregulation or large-scale transcriptional profiling, it is possible that a
downregulation of many different genes and gene path- proportion of differentially expressed genes identified
ways. These studies might also indicate that a conserved in any large-scale transcriptional screen encode proteins
stem-cell molecular signature does not exist. that are subject to post-translational modifications that
Although no definitive set of genes that defines all alter protein stability, structure or localization. Therefore,
stem cells has been identified, several signalling path- the abundance of mRNA transcripts does not infer
ways have been identified that seem to regulate dif- functionality. Furthermore, studies in Caenorhabditis
ferent types of stem cell. For instance, canonical Wnt elegans have shown that microRNAS, small non-coding
signalling through β-catenin has been implicated in RNA sequences, can modify mRNA translation and
the maintenance and self-renewal of ESCs as well as therefore have implications for protein synthesis and
adult stem cells, such as epidermal, gastrointestinal, gene function101. A large number of microRNAs are
haematopoietic and neural stem cells93. Although acti- also found in mammalian cells, including stem cells102,103
vation of β-catenin induces the self-renewal of various — such microRNAs could modulate expressed genes to
stem cells, this is not the only mechanism that supports refine complex cellular functions such as self-renewal
self-renewal, as HSCs that lack β-catenin maintain and lineage commitment.
their self-renewal capacity94 and Wnt activators are Therefore, a crucial step for determining the implica-
downregulated in skin stem cells compared with their tions of gene-expression profiles is to design gene-target-
progeny72,73. Notch signalling is another developmen- ing experiments that functionally characterize the genes
tal regulatory pathway that can mediate self-renewal and gene pathways of interest. Currently, several groups
of stem cells — Notch signals promote the mainte- are exploring approaches to functionally validate stem-
nance and self-renewal of neural, haematopoietic, cell gene-expression data, which include the creation of
gastrointestinal and epidermal stem cells by inhibiting knockout mice or the use of RNAi and gene overexpres-
differentiation95. Moreover, interaction between the sion in cell-culture models. An alternative approach is
Wnt and Notch signalling pathways might provide the the use of model organisms to define stem-cell-specific
cellular signals that are needed to drive proliferation genes, for example, RNAi in C. elegans and MORPHOLINO
(Wnt signals) in the absence of differentiation (Notch ANTISENSE OLIGONUCLEOTIDES (MOs) in Xenopus laevis and
signals), leading to symmetric self-renewing cell divi- zebrafish have been used to evaluate loss-of-function
sions required for stem-cell expansion96. Regulation phenotypes in a high-throughput manner104–108. This
of stem-cell fate in vivo is undoubtedly more complex, comparative genomics approach might increase the
microRNA
A family of short, non-coding involving polycomb genes, such as Bmi1, and devel- likelihood of identifying pathways that are important
RNA molecules opmental morphogens, such as Hedgehog, that have for stem-cell function owing to evolutionary conserva-
(~22 nucleotides) that both been shown to regulate fate decisions for several tion. In a recent study, the functional roles of genes that
post-transcriptionally regulate stem-cell types95. It should again be noted that few, if were differentially expressed between human HSCs and
target-gene expression
primarily by inhibiting protein
any, of these shared stem-cell regulators were identi- HPCs were evaluated using a zebrafish model of hae-
translation. fied by global gene-expression analysis. matopoiesis109. In this system, MO-based knockdown
of 14 out of 61 transcripts (23%) that were differentially
RNAi Implications and future directions expressed in HSCs compared with HPCs and that had
A functional tool that use small
The inability to identify a consensus stem-cell gene- no previously known roles in early haematopoiesis
interfering RNAs (siRNAs) to
knock down gene expression expression signature has led some researchers to ques- resulted in defective haematopoietic-cell development in
through sequence-specific tion the validity of such a strategy97. Despite some of MO-injected zebrafish embryos, showing the usefulness
decay of target mRNA the caveats regarding the different studies that have of model organisms for large-scale functional validation
molecules. characterized the transcriptome of defined stem cells of transcriptional profiles.
MORPHOLINO ANTISENSE
discussed above, it is clear that progress is being made As complex transcriptional networks that confer spe-
OLIGONUCLEOTIDES towards defining gene-expression patterns associated cialized cellular functions often result from the relatively
Chemically synthesized with stem-cell behaviour. The ultimate goal of defining simple actions of important transcription factors, studies
oligonucleotide analogues used a molecular signature of stem cells will be advanced will also be needed to determine the protein–DNA inter-
to knock down gene expression
further when it becomes possible to purify stem cells actions that regulate the entire cellular transcriptome. For
by specifically binding to target
transcripts to inhibit RNA to homogeneity. instance, chromatin immunoprecipitation CHIP analysis
splicing or translation. However, expression profiles do not necessarily dic- can be used to gain insight into the proteins that mediate
tate which of the genes or gene pathways are functionally complex cellular gene-expression patterns. When com-
CHIP
involved in self-renewal and pluripotency/multipotency. bined with PCR for focused analysis of transcriptional
Technique used to
immunoprecipitate complexes
There is increasing evidence that the proteome and regulation or with large promoter arrays, such analyses
of DNA with associated transcriptome of cells are only partially overlapping98, might begin to identify the gene networks that regulate
proteins. and this complicates elucidation of the functional roles stem-cell behaviour.

NATURE REVIEWS | MOLECULAR CELL BIOLOGY ADVANCE ONLINE PUBLICATION | 11


REVIEWS

Insights into the genes and gene pathways that regu- extrinsic growth factors and small molecules that allow
late stem-cell function will advance not only our basic the expansion of adult stem cells in vitro or in vivo, or
understanding of stem cells but also the entire field of that enhance the differentiation of embryonic or adult
regenerative medicine, with important implications stem cells to functional mature progeny, might ultimately
for the development of clinically applicable stem-cell lead to the development of novel therapies for a host of
therapies. Defining the combination of cellular signals, currently incurable genetic and degenerative disorders.

1. Jiang, Y. et al. Pluripotency of mesenchymal stem cells 22. Nichols, J. et al. Formation of pluripotent stem cells in the 44. Ohishi, K., Varnum-Finney, B. & Bernstein, I. D. Delta-1
derived from adult marrow. Nature 418, 41–49 (2002). mammalian embryo depends on the POU transcription enhances marrow and thymus repopulating ability of
2. Evans, M. J. & Kaufman, M. H. Establishment in culture of factor Oct4. Cell 95, 379–391 (1998). human CD34+CD38– cord blood cells. J. Clin. Invest. 110,
pluripotential cells from mouse embryos. Nature 292, 23. Chambers, I. et al. Functional expression cloning of 1165–1174 (2002).
154–156 (1981). Nanog, a pluripotency sustaining factor in embryonic stem 45. Calvi, L. M. et al. Osteoblastic cells regulate the
3. Thomson, J. A. et al. Embryonic stem cell lines derived cells. Cell 113, 643–655 (2003). haematopoietic stem cell niche. Nature 425, 841–846
from human blastocysts. Science 282, 1145–1147 (1998). 24. Mitsui, K. et al. The homeoprotein Nanog is required for (2003).
4. Labosky, P. A., Barlow, D. P. & Hogan, B. L. Embryonic maintenance of pluripotency in mouse epiblast and ES 46. Sauvageau, G. et al. Differential expression of homeobox
germ cell lines and their derivation from mouse primordial cells. Cell 113, 631–642 (2003). genes in functionally distinct CD34+ subpopulations of
germ cells. Ciba. Found. Symp. 182, 157–168; discussion 25. Rao, M. Conserved and divergent paths that regulate self- human bone marrow cells. Proc. Natl Acad. Sci. USA 91,
168–178 (1994). renewal in mouse and human embryonic stem cells. Dev. 12223–12227 (1994).
5. Pain, B. et al. Long-term in vitro culture and Biol. 275, 269–286 (2004). 47. Lessard, J., Baban, S. & Sauvageau, G. Stage-specific
characterisation of avian embryonic stem cells with 26. Niwa, H. Molecular mechanism to maintain stem cell expression of polycomb group genes in human bone
multiple morphogenetic potentialities. Development 122, renewal of ES cells. Cell Struct. Funct. 26, 137–148 marrow cells. Blood 91, 1216–1224 (1998).
2339–2348 (1996). (2001). 48. Austin, T. W., Solar, G. P., Ziegler, F. C., Liem, L. &
6. Thomson, J. A. & Marshall, V. S. Primate embryonic stem An excellent review on OCT4 signalling in ESCs that Matthews, W. A role for the Wnt gene family in
cells. Curr. Top. Dev. Biol. 38, 133–165 (1998). describes the achievements and the difficulties of hematopoiesis: expansion of multilineage progenitor cells.
7. Ogawa, M. Differentiation and proliferation of confirming gene function in stem cells. Blood 89, 3624–3635 (1997).
hematopoietic stem cells. Blood 81, 2844–2853 (1993). 27. Chambers, I. The molecular basis of pluripotency in mouse 49. Antonchuk, J., Sauvageau, G. & Humphries, R. K. HOXB4
8. Ivanova, N. B. et al. A stem cell molecular signature. embryonic stem cells. Cloning Stem Cells 6, 386–391 overexpression mediates very rapid stem cell regeneration
Science 298, 601–604 (2002). (2004). and competitive hematopoietic repopulation. Exp.
9. Ramalho-Santos, M., Yoon, S., Matsuzaki, Y., Mulligan, R. C. 28. Richards, M., Tan, S. P., Tan, J. H., Chan, W. K. & Hematol. 29, 1125–1134 (2001).
& Melton, D. A. ‘Stemness’: transcriptional profiling of Bongso, A. The transcriptome profile of human embryonic 50. Antonchuk, J., Sauvageau, G. & Humphries, R. K.
embryonic and adult stem cells. Science 298, 597–600 stem cells as defined by SAGE. Stem Cells 22, 51–64 HOXB4-induced expansion of adult hematopoietic stem
(2002). (2004). cells ex vivo. Cell 109, 39–45 (2002).
References 8 and 9 are the first analyses of global 29. Bhattacharya, B. et al. Gene expression in human 51. Buske, C. et al. Deregulated expression of HOXB4
gene-expression profiling of multiple adult and embryonic stem cell lines: unique molecular signature. enhances the primitive growth activity of human
embryonic stem-cell populations to identify Blood 103, 2956–2964 (2004). hematopoietic cells. Blood 100, 862–868 (2002).
conserved stem-cell genes. 30. Brandenberger, R. et al. MPSS profiling of human 52. Reya, T. et al. A role for Wnt signalling in self-renewal of
10. Fortunel, N. O. et al. Comment on “‘Stemness’: embryonic stem cells. BMC Dev. Biol. 4, 10 (2004). haematopoietic stem cells. Nature 423, 409–414 (2003).
transcriptional profiling of embryonic and adult stem cells” 31. Ying, Q. L., Nichols, J., Chambers, I. & Smith, A. 53. Lessard, J. & Sauvageau, G. Bmi-1 determines the
and “A stem cell molecular signature”. Science 302, BMP induction of Id proteins suppresses differentiation proliferative capacity of normal and leukaemic stem cells.
393; author reply 393 (2003). and sustains embryonic stem cell self-renewal in Nature 423, 255–260 (2003).
Similar to references 8 and 9, reports another global collaboration with STAT3. Cell 115, 281–292 (2003). 54. Bhardwaj, G. et al. Sonic hedgehog induces the
gene-expression analysis of multiple adult and 32. Xu, R. H. et al. BMP4 initiates human embryonic stem cell proliferation of primitive human hematopoietic cells via
embryonic stem-cell populations, and compares the differentiation to trophoblast. Nature Biotechnol. 20, BMP regulation. Nature Immunol. 2, 172–180 (2001).
stem-cell genes identified in references 8–10 1261–1264 (2002). 55. Cheng, T. et al. Hematopoietic stem cell quiescence
pointing out the discordance of these studies. 33. Xu, R. H. et al. Basic FGF and supression of BMP signaling maintained by p21cip1/waf1. Science 287, 1804–1808 (2000).
11. Abeyta, M. J. et al. Unique gene expression signatures of sustain undifferentiated proliferation of human ES cells. 56. Phillips, R. L. et al. The genetic program of hematopoietic
independently-derived human embryonic stem cell lines. Nature Methods 2, 185–190 (2005). stem cells. Science 288, 1635–1640 (2000).
Hum. Mol. Genet. 13, 601–608 (2004). 34. Osawa, M., Hanada, K., Hamada, H. & Nakauchi, H. One of the first attempts to generate a global
This extensive work identifies genes expressed in Long-term lymphohematopoietic reconstitution by a single differential gene-expression profile of a stem-cell
multiple human ESC lines as well as comparing CD34-low/negative hematopoietic stem cell. Science 273, population compared to its progeny using
these genes to mouse ES and adult stem-cell 242–245 (1996). subtractive hybridization of cDNA libraries.
profiles. 35. Ellisen, L. W. et al. TAN-1, the human homolog of the 57. Terskikh, A. V. et al. From hematopoiesis to neuropoiesis:
12. Sato, N. et al. Molecular signature of human embryonic Drosophila Notch gene, is broken by chromosomal evidence of overlapping genetic programs. Proc. Natl
stem cells and its comparison with the mouse. Dev. Biol. translocations in T lymphoblastic neoplasms. Cell 66, Acad. Sci. USA 98, 7934–7939 (2001).
260, 404–413 (2003). 649–661 (1991). 58. Arai, F. et al. Tie2/angiopoietin-1 signaling regulates
13. Sperger, J. M. et al. Gene expression patterns in human 36. Begley, C. G. et al. Chromosomal translocation in a human hematopoietic stem cell quiescence in the bone marrow
embryonic stem cells and human pluripotent germ cell leukemic stem-cell line disrupts the T-cell antigen receptor niche. Cell 118, 149–161 (2004).
tumors. Proc. Natl Acad. Sci. USA 100, 13350–13355 δ-chain diversity region and results in a previously 59. Li, L. & Akashi, K. Unraveling the molecular components
(2003). unreported fusion transcript. Proc. Natl Acad. Sci. USA and genetic blueprints of stem cells. Biotechniques 35,
14. Brandenberger, R. et al. Transcriptome characterization 86, 2031–2035 (1989). 1233–1239 (2003).
elucidates signaling networks that control human ES cell 37. Finger, L. R. et al. Involvement of the TCL5 gene on human 60. Rebel, V. I., Miller, C. L., Eaves, C. J. & Lansdorp, P. M.
growth and differentiation. Nature Biotechnol. 22, 707–716 chromosome 1 in T-cell leukemia and melanoma. Proc. The repopulation potential of fetal liver hematopoietic stem
(2004). Natl Acad. Sci. USA 86, 5039–5043 (1989). cells in mice exceeds that of their liver adult bone marrow
15. Hu, M. et al. Multilineage gene expression precedes 38. Royer-Pokora, B., Loos, U. & Ludwig, W. D. TTG-2, a new counterparts. Blood 87, 3500–3507 (1996).
commitment in the hemopoietic system. Genes Dev. 11, gene encoding a cysteine-rich protein with the LIM motif, 61. Park, I. K. et al. Differential gene expression profiling of adult
774–785 (1997). is overexpressed in acute T-cell leukaemia with the murine hematopoietic stem cells. Blood 99, 488–498 (2002).
16. Terskikh, A. V., Miyamoto, T., Chang, C., Diatchenko, L. & t(11;14)(p13;q11). Oncogene 6, 1887–1893 (1991). 62. Liu, H. & Verfaillie, C. M. Myeloid-lymphoid initiating cells
Weissman, I. L. Gene expression analysis of purified 39. Varnum-Finney, B. et al. Pluripotent, cytokine-dependent, (ML-IC) are highly enriched in the rhodamine-c-kit+CD33–
hematopoietic stem cells and committed progenitors. hematopoietic stem cells are immortalized by constitutive CD38– fraction of umbilical cord CD34+ cells. Exp.
Blood 102, 94–101 (2003). Notch1 signaling. Nature Med. 6, 1278–1281 (2000). Hematol. 30, 582–589 (2002).
17. Akashi, K. et al. Transcriptional accessibility for genes of 40. Lecuyer, E. & Hoang, T. SCL: from the origin of 63. Hess, D. A. et al. Functional characterization of highly
multiple tissues and hematopoietic lineages is hematopoiesis to stem cells and leukemia. Exp. Hematol. purified human hematopoietic repopulating cells isolated
hierarchically controlled during early hematopoiesis. Blood 32, 11–24 (2004). according to aldehyde dehydrogenase activity. Blood 104,
101, 383–389 (2003). 41. Yamada, Y. et al. The T cell leukemia LIM protein Lmo2 is 1648–1655 (2004).
18. Ye, M. et al. Hematopoietic stem cells expressing the necessary for adult mouse hematopoiesis. Proc. Natl 64. Gage, F. H. Mammalian neural stem cells. Science 287,
myeloid lysozyme gene retain long-term, multilineage Acad. Sci. USA 95, 3890–3895 (1998). 1433–1438 (2000).
repopulation potential. Immunity 19, 689–699 (2003). 42. Varnum-Finney, B., Brashem-Stein, C. & Bernstein, I. D. 65. Uchida, N. et al. Direct isolation of human central nervous
19. Arney, K. L. & Fisher, A. G. Epigenetic aspects of Combined effects of Notch signaling and cytokines induce system stem cells. Proc. Natl Acad. Sci. USA 97,
differentiation. J. Cell Sci. 117, 4355–4363 (2004). a multiple log increase in precursors with lymphoid and 14720–14725 (2000).
20. Tanaka, T. S. et al. Gene expression profiling of embryo- myeloid reconstituting ability. Blood 101, 1784–1789 66. Geschwind, D. H. et al. A genetic analysis of neural
derived stem cells reveals candidate genes associated (2003). progenitor differentiation. Neuron 29, 325–339 (2001).
with pluripotency and lineage specificity. Genome Res. 12, 43. Karanu, F. N. et al. Human homologues of Delta-1 and 67. Karsten, S. L. et al. Global analysis of gene expression in
1921–1928 (2002). Delta-4 function as mitogenic regulators of primitive neural progenitors reveals specific cell-cycle, signaling,
21. Sharov, A. A. et al. Transcriptome analysis of mouse stem human hematopoietic cells. Blood 97, 1960–1967 and metabolic networks. Dev. Biol. 261, 165–182
cells and early embryos. PLoS Biol. 1, E74 (2003). (2001). (2003).

12 | ADVANCE ONLINE PUBLICATION www.nature.com/reviews/molcellbio


REVIEWS

68. Wright, L. S. et al. Gene expression in human neural stem 90. Driessen, R. L., Johnston, H. M. & Nilsson, S. K. 111. Grove, J. E., Bruscia, E. & Krause, D. S. Plasticity of bone
cells: effects of leukemia inhibitory factor. J. Neurochem. Membrane-bound stem cell factor is a key regulator in the marrow-derived stem cells. Stem Cells 22, 487–500
86, 179–195 (2003). initial lodgment of stem cells within the endosteal marrow (2004).
69. Livesey, F. J., Young, T. L. & Cepko, C. L. An analysis of region. Exp. Hematol. 31, 1284–1291 (2003). 112. Wagers, A. J. & Weissman, I. L. Plasticity of adult stem
the gene expression program of mammalian neural 91. Hackney, J. A. et al. A molecular profile of a hematopoietic cells. Cell 116, 639–648 (2004).
progenitor cells. Proc. Natl Acad. Sci. USA 101, stem cell niche. Proc. Natl Acad. Sci. USA 99, 113. Camargo, F. D., Chambers, S. M. & Goodell, M. A. Stem
1374–1379 (2004). 13061–13066 (2002). cell plasticity: from transdifferentiation to macrophage
70. Luo, Y. et al. Microarray analysis of selected genes in Provides a complementary analysis of gene fusion. Cell Prolif. 37, 55–65 (2004).
neural stem and progenitor cells. J. Neurochem. 83, expression in a stem-cell niche, specifically the 114. Verfaillie, C. M. Adult stem cells: assessing the case for
1481–1497 (2002). haematopoietic stem-cell niche, as a means to pluripotency. Trends Cell Biol. 12, 502–508 (2002).
71. Blanpain, C., Lowry, W. E., Geoghegan, A., Polak, L. & understand the extrinsic signals that regulate stem- 115. Khavari, P. A. Profiling epithelial stem cells. Nature
Fuchs, E. Self-renewal, multipotency, and the existence of cell function. Biotechnol. 22, 393–394 (2004).
two cell populations within an epithelial stem cell niche. 92. Yamashita, Y. M., Jones, D. L. & Fuller, M. T. Orientation of 116. Gronthos, S. et al. Molecular and cellular characterisation
Cell 118, 635–648 (2004). asymmetric stem cell division by the APC tumor of highly purified stromal stem cells derived from
Identifies two spatially distinct stem-cell populations suppressor and centrosome. Science 301, 1547–1550 human bone marrow. J. Cell Sci. 116, 1827–1835
in the bulge-cell pool and characterizes their (2003). (2003).
expression profile, in vivo and in vitro potential and 93. Reya, T. & Clevers, H. Wnt signalling in stem cells and 117. Oh, H. et al. Cardiac progenitor cells from adult myocardium:
niche. cancer. Nature 434, 843–850 (2005). homing, differentiation, and fusion after infarction. Proc. Natl
72. Morris, R. J. et al. Capturing and profiling adult hair follicle 94. Cobas, M. et al. β-catenin is dispensable for Acad. Sci. USA 100, 12313–12318 (2003).
stem cells. Nature Biotechnol. 22, 411–417 (2004). hematopoiesis and lymphopoiesis. J. Exp. Med. 199, 118. Paku, S., Schnur, J., Nagy, P. & Thorgeirsson, S. S.
73. Tumbar, T. et al. Defining the epithelial stem cell niche in 221–229 (2004). Origin and structural evolution of the early proliferating
skin. Science 303, 359–363 (2004). 95. Molofsky, A. V., Pardal, R. & Morrison, S. J. Diverse oval cells in rat liver. Am. J. Pathol. 158, 1313–1323
74. Mills, J. C., Andersson, N., Hong, C. V., Stappenbeck, T. S. mechanisms regulate stem cell self-renewal. Curr. Opin. (2001).
& Gordon, J. I. Molecular characterization of mouse gastric Cell Biol. 16, 700–707 (2004). 119. Petkov, P. M. et al. Gene expression pattern in hepatic
epithelial progenitor cells. Proc. Natl Acad. Sci. USA 99, 96. Espinosa, L., Ingles-Esteve, J., Aguilera, C. & Bigas, A. stem/progenitor cells during rat fetal development using
14819–14824 (2002). Phosphorylation by glycogen synthase kinase-3β down- complementary DNA microarrays. Hepatology 39,
75. Stappenbeck, T. S., Mills, J. C. & Gordon, J. I. Molecular regulates Notch activity, a link for Notch and Wnt 617–627 (2004).
features of adult mouse small intestinal epithelial pathways. J. Biol. Chem. 278, 32227–32235 (2003). 120. Arai, M. et al. Gene expression profiles in liver regeneration
progenitors. Proc. Natl Acad. Sci. USA 100, 1004–1009 97. Zipori, D. The nature of stem cells: state rather than entity. with oval cell induction. Biochem. Biophys. Res. Comm.
(2003). Nature Rev. Genet. 5, 873–878 (2004). 317, 370–376 (2004).
76. Gilboa, L. & Lehmann, R. How different is Venus from A critical analysis of reported stem-cell gene- 121. Seaberg, R. M. et al. Clonal identification of multipotent
Mars? The genetics of germ-line stem cells in Drosophila expression data that raises questions regarding the precursors from adult mouse pancreas that generate
females and males. Development 131, 4895–4905 validity of the concept of conserved stem-cell genes. neural and pancreatic lineages. Nature Biotechnol. 22,
(2004). 98. Gygi, S. P., Rochon, Y., Franza, B. R. & Aebersold, R. 1115–1124 (2004).
77. Yamashita, Y. M., Fuller, M. T. & Jones, D. L. Signaling in Correlation between protein and mRNA abundance in 122. Bonner-Weir, S. et al. The pancreatic ductal epithelium
stem cell niches: lessons from the Drosophila germline. yeast. Mol. Cell. Biol. 19, 1720–1730 (1999). serves as a potential pool of progenitor cells. Pediatr.
J. Cell Sci. 118, 665–672 (2005). 99. Miyamoto, T. et al. Myeloid or lymphoid promiscuity as a Diabetes 5, 16–22 (2004).
78. Xie, T. & Spradling, A. C. A niche maintaining germ line stem critical step in hematopoietic lineage commitment. Dev. 123. Hawke, T. J. & Garry, D. J. Myogenic satellite cells:
cells in the Drosophila ovary. Science 290, 328–330 (2000). Cell 3, 137–147 (2002). physiology to molecular biology. J. Appl. Physiol. 91,
79. Tulina, N. & Matunis, E. Control of stem cell self-renewal in 100. Lian, Z. et al. Genomic and proteomic analysis of the 534–551 (2001).
Drosophila spermatogenesis by JAK–STAT signaling. myeloid differentiation program. Blood 98, 513–524 124. de Rooij, D. G. & Grootegoed, J. A. Spermatogonial stem
Science 294, 2546–2549 (2001). (2001). cells. Curr. Opin. Cell Biol. 10, 694–701 (1998).
80. Kiger, A. A., Jones, D. L., Schulz, C., Rogers, M. B. & 101. He, L. & Hannon, G. J. MicroRNAs: small RNAs with a big
Fuller, M. T. Stem cell self-renewal specified by JAK–STAT role in gene regulation. Nature Rev. Genet. 5, 522–531 Acknowledgements
activation in response to a support cell cue. Science 294, (2004). The authors would like to acknowledge the work of our colleagues
2542–2545 (2001). 102. Suh, M. R. et al. Human embryonic stem cells express a that we have discussed, and apologize to our colleagues whose
81. Gonczy, P. & DiNardo, S. The germ line regulates somatic unique set of microRNAs. Dev. Biol. 270, 488–498 (2004). work was not discussed due to space limitations.
cyst cell proliferation and fate during Drosophila 103. Chen, C. Z., Li, L., Lodish, H. F. & Bartel, D. P. MicroRNAs
spermatogenesis. Development 122, 2437–24347 (1996). modulate hematopoietic lineage differentiation. Science Competing interests statement
82. Song, X., Zhu, C. H., Doan, C. & Xie, T. Germline stem 303, 83–86 (2004). The authors declare no competing financial interests.
cells anchored by adherens junctions in the Drosophila 104. Pickart, M. A. et al. Functional genomics tools for the
ovary niches. Science 296, 1855–1857 (2002). analysis of zebrafish pigment. Pigment Cell Res. 17,
83. Tran, J., Brenner, T. J. & DiNardo, S. Somatic control over 461–470 (2004). Online links
the germline stem cell lineage during Drosophila 105. Nasevicius, A. & Ekker, S. C. Effective targeted gene
spermatogenesis. Nature 407, 754–757 (2000). ‘knockdown’ in zebrafish. Nature Genet. 26, 216–220 DATABASES
84. Deng, W. & Lin, H. Spectrosomes and fusomes anchor (2000). The following terms in this article are linked online to:
mitotic spindles during asymmetric germ cell divisions and 106. Ekker, S. C. Morphants: a new systematic vertebrate Entrez Gene: http://www.ncbi.nlm.nih.gov/entrez/query.
facilitate the formation of a polarized microtubule array for functional genomics approach. Yeast 17, 302–306 fcgi?db=gene
oocyte specification in Drosophila. Dev. Biol. 189, 79–94 (2000). Angpt1 | Bmi1 | BMP4 | Ccnd1 | Cdkn1a | CHEK2 | DNMT3B |
(1997). 107. Heasman, J. Morpholino oligos: making sense of Eras | FGF1 | GDF3 | GJA1 | Hoxb4 | Lhx2 | LIN28 | LMO2 |
85. Kiger, A. A., White-Cooper, H. & Fuller, M. T. Somatic antisense? Dev. Biol. 243, 209–214 (2002). Nanog | NOTCH1 | Oct4 | Pax6 | Runx1 | SHH |SOX2 | TAL1 |
support cells restrict germline stem cell self-renewal and 108. Bargmann, C. I. High-throughput reverse genetics: RNAi Tek | Tdgf1 | TDGF1 | TGIF | Utf1 | Zfp42
promote differentiation. Nature 407, 750–754 (2000). screens in Caenorhabditis elegans. Genome Biol. 2,
86. Zhang, J. et al. Identification of the haematopoietic stem REVIEWS1005 (2001). FURTHER INFORMATION
cell niche and control of the niche size. Nature 425, 109. Eckfeldt, C. E. et al. Functional analysis of hematopoietic Catherine Verfaillie’s Institute: http://www.stemcell.umn.edu
836–841 (2003). stem cell gene expressin using zebrafish. PLoS Biol. 3, National Institutes of Health Stem Cell Information:
87. Shen, Q. et al. Endothelial cells stimulate self-renewal and e254 (2005). http://stemcells.nih.gov
expand neurogenesis of neural stem cells. Science 304, One of the first descriptions of global gene- The Stem Cell Database (SCDB):
1338–1340 (2004). expression profiling of a human stem-cell population http://stemcell.princeton.edu
88. Doetsch, F. A niche for adult neural stem cells. Curr. Opin. coupled with high-throughput functional genomics The Stem Cell Genome Anatomy Project (SCGAP):
Genet. Dev. 13, 543–550 (2003). screening using zebrafish. http://www.scgap.org
89. Nishimura, E. K. et al. Dominant role of the niche in 110. Pomerantz, J. & Blau, H. M. Nuclear reprogramming: a key The Stromal Cell Database (StroCDB):
melanocyte stem-cell fate determination. Nature 416, to stem cell function in regenerative medicine. Nature Cell http://stromalcell.princeton.edu
854–860 (2002). Biol. 6, 810–816 (2004). Access to this interactive links box is free online.

NATURE REVIEWS | MOLECULAR CELL BIOLOGY ADVANCE ONLINE PUBLICATION | 13

También podría gustarte