Sunteți pe pagina 1din 8

543

A niche for adult neural stem cells


Fiona Doetsch
The adult mammalian brain harbors multipotent stem cells,
which reside and participate in specialized niches that support
self-renewal and differentiation. The first cellular and molecular
elements of the stem cell niche in the adult brain have been
identified and include cellcell interactions and somatic cell
signaling, the vasculature, the extracellular matrix and basal
lamina. Furthermore, regulation at the epigenetic level via
chromatin modification and remodeling is an integral aspect of
stem cell biology. Understanding the in vivo stem cell niche will
provide a framework for the elucidation of stem cell function in
the adult brain.
Addresses
Harvard University, Department of Molecular and Cellular Biology,
16 Divinity Avenue, Cambridge, Massachusetts 02138, USA
e-mail: fkd2101@columbia.edu

Current Opinion in Genetics & Development 2003, 13:543550


This review comes from a themed issue on
Differentiation and gene regulation
Edited by Azim Surani and Austin Smith
0959-437X/$ see front matter
2003 Elsevier Ltd. All rights reserved.
DOI 10.1016/j.gde.2003.08.012

Abbreviations
BDNF
brain-derived neurotrophic factor
BMP
bone morphogenetic protein
ECM
extracellular matrix
EGF
epidermal growth factor
bFGF
basic fibroblast growth factor
GFAP
glial fibrillary acidic protein
HAT
histone acetyltransferase
HDAC
histone deacetylase
IGF-1
insulin-like growth factor-1
LIF
leukemia inhibitory factor
PDGF
platelet derived growth factor
SDF-1
stromal cell-derived factor-1
SGZ
subgranular zone
Shh
Sonic hedgehog
SSEA-1 stage specific embryonic antigen-1
SVZ
subventricular zone
TGF-a
transforming growth factor-a
VEGF
vascular endothelial growth factor

Introduction
Stem cells are remarkable, having the capacity to selfrenew and differentiate along multiple lineages and to
contribute to ongoing tissue maintenance and regeneration after injury in the adult. During brain development,
neuroepithelial stem cells located in the ventricular zone
next to the ventricles give rise to neurons and glia [1].
Glia, largely considered support cells, comprise astrowww.current-opinion.com

cytes, star-shaped cells with diverse functions, and oligodendrocytes, which myelinate the axons of neurons.
Although neurogenesis is mostly complete by birth, gliogenesis continues throughout life. However, two germinal regions persist in the adult mammalian brain that
generate large numbers of neurons: the subventricular
zone (SVZ) of the lateral ventricle, and the subgranular
zone (SGZ) of the hippocampal formation [2]. This
unexpected form of brain plasticity led to a search for
the underlying stem cells. Strikingly, adult neural stem
cells are not amorphous undifferentiated cells, but exhibit
features of differentiated astrocytes at the ultrastructural
and molecular level including expression of glial intermediate filaments, such as glial fibrillary acidic protein
(GFAP) [2].
A specialized milieu therefore supports neurogenesis and
regeneration after injury in restricted brain regions. Stem
cells frequently reside in niches that regulate their selfrenewal, activation and differentiation [3]. Common components of stem cell niches are signaling from somatic
cells, a basement membrane for cell anchoring, and
extracellular matrices (ECMs), which modulate the adhesiveness and activity of signaling molecules. Understanding the in vivo adult neural stem cell niche is crucial to
delineating the function of neural stem cells and their
progeny and ultimately their therapeutic potential. In this
review, I outline the architecture, cell types and lineages
of adult mouse brain germinal regions, and important
aspects of the neural stem cell niche and integrate them
with relevant recent findings from other stem cell fields.

Anatomy of adult neurogenic regions


The subventricular zone

The SVZ is a layer of dividing cells extending along the


lateral wall of the lateral ventricle (Figure 1a). New
neurons are born throughout the SVZ and join a network
of chains of migrating neurons that coalesce to form the
rostral migratory stream leading to the olfactory bulb,
where they differentiate into granule and periglomerular
neurons [2]. The adult SVZ may also be a site of oligodendrocyte generation [4]. The SVZ therefore not only
supports cell genesis, but is a site of robust directional
migration. The cell types and architecture of the SVZ
have been characterized at the ultrastructural level [5].
There are four main cell types in the SVZ: neuroblasts
(Type A cells), SVZ astrocytes (Type B cells), immature
precursors (Type C cells) and ependymal cells [5]. A
single layer of multi-ciliated ependymal cells separates
the SVZ from the lateral ventricle (Figure 1c). The
neuroblasts divide as they migrate as chains through glial
tunnels formed by the processes of slowly dividing SVZ
Current Opinion in Genetics & Development 2003, 13:543550

544 Differentiation and gene regulation

Figure 1

The SVZ niche, cell types and stem cell lineage. (a) Frontal schematic of the adult mouse brain showing the location of the SVZ in orange between the
lateral ventricle (LV) and the striatum. The corpus callosum is depicted in dark gray. The box in (a) is expanded in (b). (b) Blood vessels (BV) are
common in the SVZ and endothelial cells lining the blood vessels are likely a source of signals for adult neurogenesis. A specialized basal lamina (BL)
extends from blood vessels into the SVZ and terminates in small bulbs adjacent to the ependymal cells (E), which line the lateral ventricle. (c) Crosssectional schematic showing the cell types and their organization in the SVZ. Multi-cilitated ependymal cells (E, gray) line the lateral ventricle. Chains of
neuroblasts (A, red) travel through tunnels formed by the processes of SVZ astrocytes (B, blue). Focal clusters of rapidly dividing Type C cells
(C, green) are scattered along the network of chains of neuroblasts. SVZ astrocytes occasionally extend a process to contact the lateral ventricle and
exhibit a short single cilium. An ECM-rich basal lamina (BL, black) makes extensive contact with all SVZ cell types, terminating in bulbs adjacent
to ependymal cells and forms an essential part of the SVZ stem cell niche. SVZ astrocytes (GFAP) act as stem cells in this region and divide to
generate transit-amplifying Type C cells (GFAP/Dlx2), which in turn divide to generate the neuroblasts (GFAPDlx2PSANCAM) that migrate to
the olfactory bulb.

astrocytes. Focal clusters of rapidly dividing precursors


(Type C cells) are scattered along the SVZ network of
chains. Occasionally, an SVZ astrocyte extends a process
between ependymal cells to contact the lateral ventricle
and exhibits a short, single cilium characteristic of neuroepithelial stem cells in the embryo [6]. Another noteworthy feature of this region is a specialized basal lamina,
which extends from blood vessels in the SVZ and terminates in small bulbs adjacent to ependymal cells, and
contacts all SVZ cell types [7] (Figure 1b). SVZ astrocytes are stem cells in this region and divide to generate
the neuroblasts via GFAP/Dlx2 transit-amplifying C
cells [8].
The subgranular zone

In contrast to the extensive migration undertaken by


neurons destined for the olfactory bulb, dentate gyrus
granule neurons are born locally in the SGZ, a germinal
layer between the dentate gyrus and the hilus (Figure 2).
Neurogenesis in the SGZ occurs in foci closely associated
Current Opinion in Genetics & Development 2003, 13:543550

with blood vessels [9]. These foci contain SGZ astrocytes,


which extend basal processes under the blades of the
dentate gyrus and an apical process into the granule cell
layer, dividing immature Type D cells, which have begun
to express markers of neuronal differentiation, newly generated neurons, and endothelial cells [9,10] (Figure 2).
As in the SVZ, SGZ astrocytes are the primary precursors
of neurons and divide to generate granule neurons via
Type D cells [10]. Interestingly, Type D cells divide less
frequently and are more differentiated than the transitamplifying C cells in the SVZ.
The anatomy described above reveals that several
architectural elements contribute to the stem cell niche
supporting adult neurogenesis: first, extensive cellcell
interactions; second (in the case of the SVZ) proximity
to the cerebrospinal fluid of the lateral ventricle; third,
close association with blood vessels; and fourth, a rich ECM
and specialized basal lamina. In addition, the epigenetic
state of cells determines their ability to respond to signals.
www.current-opinion.com

A niche for adult neural stem cells Doetsch 545

Figure 2

D
B

BV
GFAP+

PSANCAM+ PSANCAM+
Current Opinion in Genetics & Development

The SGZ neurogenic niche, cell types and lineage. Frontal schema shows the location of the SGZ in the hippocampal formation. The SGZ lies between
the granule cell layer and the hilus. The region in the box is expanded to the right and shows the cell types and organization of the SGZ. SGZ astrocytes
(B, dark blue) are in close proximity to blood vessels (BV, light gray). Both endothelial cells and neural precursors are found in foci of dividing
cells, frequently at the junction of blood vessels, suggesting mutual co-regulation. Endothelial cells are likely an important source of signals for
neurogenesis. Small clusters of immature dividing D cells (orange) are adjacent to SGZ astrocytes. SGZ astrocytes (GFAP) divide to give rise to D
cells (PSANCAM), which generate granule neurons (PSANCAM)(G, red).

Cellcell interactions and signaling


The abundant cellcell interactions in adult neurogenic
regions [5,7,9,10] can influence the maintenance, activation and differentiation of stem cells via feedback, signaling and the mode of division (asymmetric or symmetric).
Astrocytes are uniquely poised to act as sensors and
regulators in the stem cell niche. Their meandering
processes contact all cell types and their end feet terminate on the basal lamina of blood vessels. They are thus
primed to detect alterations in neuronal and precursor
number and to translate signals from the vasculature and
other cells in germinal regions. Astrocytes are extensively
coupled by gap junctions. As such, they may provide rapid
propagation of signaling within the stem cell niche.
Furthermore, astrocytes themselves secrete factors that
support neurogenesis in vitro [11,12]. How astrocytes
integrate their dual roles as support cells and stem cells
and whether all astrocytes have stem cell potential are
fascinating issues.
Ependymal cells are important regulators in the SVZ and
promote a neurogenic niche, although they are not stem
cells [8,13,14,15,16] as has been proposed [17].
Indeed, ependymal cells produce noggin, an antagonist
of BMP signaling, which prevents glial differentiation of
SVZ cells induced by BMPs [18]. Their proximity to the
lateral ventricle also leaves them poised to capture factors
secreted by the choroid plexus into the cerebrospinal
fluid, as are SVZ astrocytes contacting the lateral ventricle. In the adult, ependymal cells and SGZ precursors
express CXCR4, the receptor for the chemokine stromal
cell-derived factor-1 (SDF-1), required by haematopowww.current-opinion.com

eitic stem cells and for cerebellar and hippocampal development [19,20]. During development, SDF-1, secreted
by the meninges, acts as an attractant to hold cerebellar
precursors in the external granule layer and cooperates
with the mitogen Sonic hedgehog (Shh) to increase their
proliferation [21]. Reverse signaling through ephrinB
inhibits the attraction to SDF-1, allowing the inward
migration of granule cells [22]. The potential interaction
of CXCR4 and SDF-1 with EphB/ephrinB [23] and Shh
[24] signaling, both active in adult brain germinal regions,
merits further study.
Cellcell contact via adherens junctions regulates selfrenewal in the Drosophila germline stem cell niche [3].
Cadherins and b-catenin are integral components of
adherens junctions (reviewed in [25]). b-catenin is also
a downstream component of the Wnt signaling pathway
and Wnt signaling promotes the self-renewal of haematopoetic stem cells [26]. As such, b-catenin may act
through several pathways to affect stem cell self-renewal.
Cadherins are expressed in adult neurosphere cultures
[27] (see below), suggesting that b-catenin signaling is
involved in the adult neural stem cell niche, although
in vivo analysis is still lacking. In the embryo, b-catenin is
expressed in adherens junctions between neuroepithelial
stem cells/precursors in the ventricular zone [28]. Overexpression of constitutively active b-catenin in these
neuroepithelial precursors expands their pool size as a
result of increased cell cycle re-entry rather than exit
[28], perhaps due to increased symmetric self-renewing
divisions instead of asymmetric differentiative divisions.
Adherens junctions may also coordinate the plane of
division of a cell by orienting the mitotic spindle. As
Current Opinion in Genetics & Development 2003, 13:543550

546 Differentiation and gene regulation

such, the differential targeting of developmentally important mRNAs to one daughter centrosome [29] could
provide a mechanism for asymmetric division and inheritance of cell fate.

Growth factor signaling


Growth factor signaling acts at different stages of the stem
cell lineage, reflecting the need for tight control over cell
division within adult neurogenic regions. Although EGFresponsive cells in the SVZ are believed to be relatively
quiescent stem cells in vivo [30], the majority of EGFresponsive cells are the transit-amplifying C type [16].
In vivo stimulation of C cells via EGF infusion causes
them to arrest neuroblast production, divide rapidly and
become highly migratory along blood vessels and white
matter fiber tracts. Selective killing of dividing Dlx2 C
cells in vivo using genetic approaches dramatically
reduces this response [16]. Signaling through the
EGF-receptor may therefore provide a mechanism to
selectively amplify activated stem cells and transit-amplifying C cells without depleting the more quiescent stem
cells in vivo. Other factors affecting proliferation in adult
brain germinal regions are bFGF, IGF1, TGF-a, VEGF,
Eph/ephrin signaling, Shh, prolactin, adrenal hormones
and exercise, although the stages of the lineage each acts
on is unknown ([23,24,31,32]; reviewed in [33]). Different
cell-cycle inhibitors are also active in distinct progenitor
populations: p27 null mice have an increased number of
transit-amplifying C cells [34], whereas p19 acts primarily
in the neuroblasts [35].
Neural stem cells can be greatly expanded in vitro with
growth factors including EGF and bFGF, either as neurospheres (floating cell clusters) or as flat adherent cultures
[1]. Although beyond the scope of this review, in vitro
analysis of self-renewal in neurospheres has implicated
the Notch signaling pathway [36,37], Emx2 [38], ciliary
neurotrophic factor/gp130 signaling [36,39] and Pten [40].
However, it is important to note that the majority of EGFresponsive neurospheres arise from the transit-amplifying
C cells rather than the in vivo stem cells [16]. Thus,
although transit-amplifying cells have stem cell potential
in vitro, the in vivo niche exerts a key influence in
regulating the behavior of stem cells and their progeny.
The in vivo identity of FGF-responsive cells in the adult
brain remains unknown but they may correspond to
relatively quiescent stem cell astrocytes. This potential
can be harnessed for brain repair. The in vivo stimulation
of endogenous progenitors with both EGF and bFGF
after ischemia results in cell replacement and recovery of
brain function in the hippocampus [41].

The vascular niche of adult germinal


regions
The vasculature is an integral component of the stem cell
niche. Endothelial cells and pericytes surround the lumen
of blood vessels and are separated from the brain by a
Current Opinion in Genetics & Development 2003, 13:543550

basal lamina. Neurogenesis occurs in foci closely associated with blood vessels [9]. In fact, angiogenesis and
neurogenesis may be co-regulated and reciprocally signal.
Both are stimulated by the same factors, including bFGF,
VEGF, IGF-1 and TGF-a and endothelial cells secrete
known mitogens and differentiation and survival factors
of neurons (bFGF, IGF-1, VEGF, PDGF, IL8 and
BDNF) [9,31,42]. Furthermore, when the intimate association of endothelial cells and SGZ precursors is disrupted by irradiation, accompanied by microglial
activation, the neurogenic milieu is lost: even precursors
from a non-irradiated brain transplanted into an irradiated
host adopt a glial fate [43]. A further link between
angiogenesis and neurogenesis is found in the adult songbird brain during testosterone-induced angiogenesis [42].
Exogenous testosterone induces an expanded vasculature
through VEGF signaling and the localized endothelial
cell secretion of BDNF, a survival factor of new neurons,
at their integration site [42].

Extracellular matrix and basal lamina


The basal lamina tethers factors, anchors cells and provides spatial cues within the stem cell niche. Carbohydrates attached to the basal lamina and ECM can
potentiate ligand activity or sequester ligands as bound
stores. Moreover, ECM-bound or cell-surface-bound
molecules can be cleaved to release active ligand or
soluble inhibitors. For example, matrix metalloproteinase-9 regulates haematopoeitic stem cell and endothelial progenitor recruitment in the bone marrow, by
cleaving membrane-bound Kit ligand and sequestered
VEGF to release soluble Kit ligand and VEGF, respectively [44,45]. The specialized basal lamina and ECM
components in the adult SVZ [7] likely form an integral
part of the stem cell niche. The basal lamina bulbs
adjacent to ependymal cells that envelope SVZ cells may
correspond to concentrated foci of signaling molecules.
The adult SVZ is rich in tenascin-C, collagen-1, heparan
sulfate proteoglycans and chondroitin sulfate proteoglycans, as well as integrins [7,4649], which are implicated
in both proliferation and migration. Heparan sulfate
proteoglycans bind many factors active in adult neurogenesis, including morphogens and mitogens (BMP-2,-4,
Shh and Wnts), components of the ECM (collagens,
laminins and tenascin), growth factors (EGFs, FGFs,
IGF-II, PDGF-AA and VEGF), chemokines and cytokines [7,18,24,46,47,50]. Interestingly, the cell-surface
carbohydrate LeX/SSEA-1 (stage specific embryonic
antigen-1) is highly enriched around blood vessels in
adult germinal regions and in brain astrocytes [13] and
may be an important component of the stem cell niche.
In vitro, Lex carbohydrate is shed as an inhibitory ectodomain but it can also oligomerize bFGF, a mitogen of
neural stem cells [13]. Thus, factors from endothelial
cells and other cells in germinal regions can be regulated
in the stem cell niche by complexing with the ECM and/
or basal lamina.
www.current-opinion.com

A niche for adult neural stem cells Doetsch 547

The chromatin state and stem cell fate

Figure 3

(a)

Stem cell

Transit
amplifying

Differentiated

(b)

Ac
'Open'

HDAC

HAT

Neutral

Me
Repressed

The stages in the lineage from stem cell to fully differentiated cell are accompanied by sequential waves of expression of transcriptional regulators [51,52] (Figure 3a).
Indeed, the ectopic expression of master transcription
factors can induce the differentiation of a particular lineage. Chromatin modification and remodeling also play a
crucial role in stem cell biology and differentiation by
regulating the intrinsic state of responsiveness of a cell.
Epigenetic changes regulate gene activity and silencing
through chromatin alteration. DNA is complexed with
core histones to form chromatin, and can exist in an open
actively transcribed state, a repressed state, or a silent
state, mediated by the specific modification of histone
tails (Figure 3b). In an open state, chromatin is made
accessible through the acetylation of histone residues by
histone acetyl transferases (HATs). The open state can be
further stabilized by histone methylation and by the
interaction of trithorax proteins. Locus silencing is a
sequential multi-step process. First, acetyl groups are
removed by histone deacetylases (HDACs), followed
by methylation of histone residues. Methyl-binding proteins, including Polycomb group proteins, are recruited to
these sites and stabilize the condensed chromatin. A locus
is silenced by the recruitment of further Polycomb group
members, by CpG DNA methylation, and by repositioning of the locus within the nucleus (reviewed in [53]).
How might chromatin modification and remodeling be
involved in stem cell biology? Epigenetic changes are an
important intrinsic mechanism underlying cell identity
and cell fate decisions. DNA methylation of promoter
regions in stem cells can make them refractory to extracellular differentiation signals. LIF (leukemia inhibitory
factor) is a potent inducer of astrocyte differentiation via
STAT3 activation, but early embryonic neuroepithelial
stem cells do not differentiate in response to LIF. This
results from CpG methylation of a STAT3-binding element in the GFAP promoter [54], which prevents
STAT3 binding. At later developmental stages, the
STAT3 element is demethylated and the cells can differentiate into astrocytes in response to LIF [54].

Pc
'Silent'

Distinct loci are likely active in stem cells, transit-amplifying cells and differentiated cells, with differentiation

Current Opinion in Genetics & Development

A classic stem cell lineage in which a stem cell undergoes a selfrenewing division to generate another stem cell and a transit-amplifying
cell, which divides rapidly but is depleted after several divisions and
gives rise to differentiated cells. Two mechanisms act in concert in stem
cell lineages. (a) Transcription factors are sequentially activated at each
stage of differentiation and ultimately specify the phenotype of the
differentiated cells. Transcription factors may be deactivated at
sequential steps or maintained along the lineage. They may lie adjacent
www.current-opinion.com

to one another or may be dispersed over the genome. (b) The role of
chromatin remodeling in cell fate. Chromatin can be in an open actively
transcribed state or in a silent state associated with acetylation of
histone residues (yellow) by histone acetyltransferase (HAT). Histone
deacetylase (HDAC) removes the acetyl (Ac) residues (orange
triangles) as a first step in repressing chromatin. Conserved histone
residues are then methylated (Me, blue squares) rendering the
chromatin repressed. Other proteins bind to these methyl groups,
including HP-1 and Polycomb (Pc) group proteins (purple) and result in
complete silencing of a locus with CpG methylation. Contiguous
clusters of genes can be repressed through spreading of the
silencing machinery.
Current Opinion in Genetics & Development 2003, 13:543550

548 Differentiation and gene regulation

accompanied by switches between open and repressed


states of chromatin. Importantly, contiguous clusters of
genes can either be coordinately activated or inactivated as
a result of spreading of chromatin modification. For example, in non-neuronal cells, neuron-specific genes such as
the Type II sodium channel are repressed by binding of
REST/NRSF and CoREST to RE-1 response elements
[55] as a complex with HDACs. Contiguous genes without RE-1 response elements are also silenced due to the
spreading of molecular silencing machinery including
methyl-binding proteins along adjacent chromatin [56].
The clustering of stem cell genes/expressed sequence
tags on chromosomes [57] supports the idea that coordinate
regulation of genes will be central to stem cell biology.
Furthermore, Polycomb group genes, transcriptional repressors, have recently been implicated in haematopoeitic
stem cell self-renewal. In Bmi1 null mice, haematopoeitic
stem cells undergo reduced stem cell self-renewal as a
result of relieved gene suppression, including that of cellcycle inhibitors [58,59]. The use of distinct complements
of cell-cycle regulators in different progenitors [2] and
switches from one stage to another may be, in part, regulated by changes in chromatin accessibility.

Conclusions
Understanding the in vivo adult neural stem cell niche
and the cellular responses in it will lead to insights into
brain repair and the origin of pathological processes,
including cancer. The advent of novel technologies such
as RNA interference and conditional genetic approaches
in combination with genomic and proteomic analyses of
the in vivo stem cells and their progeny will elucidate
further their biology and function, and help realize the
tantalizing potential for therapy.

Acknowledgements
I apologize to all those whose work could not be cited due to space
limitations. Many thanks to J Rihel and L Zakhary for comments on the
manuscript. F Doetsch was supported by the Harvard University Society of
Fellows, the Radcliffe Institute for Advanced Study (Burroughs Wellcome
Fellow) and a WF Milton Fund.

subventricular germinal zone in the adult mammalian brain.


J Neurosci 1997, 17:5046-5061.
6.

Doetsch F, Garcia-Verdugo JM, Alvarez-Buylla A: Regeneration of


a germinal layer in the adult mammalian brain. Proc Natl Acad
Sci USA 1999, 96:11619-11624.

7.


Mercier F, Kitasako JT, Hatton GI: Anatomy of the brain


neurogenic zones revisited: fractones and the fibroblast/
macrophage network. J Comp Neurol 2002, 451:170-188.
Basal lamina are important components of stem cell niches. This paper
describes the presence of a specialized basal lamina in the SVZ based
on immunostaining for laminin at the light microscope and on ultrastructural analysis. The basal lamina contacts all SVZ cell types and
may play an important role in cell tethering and ligand binding and
regulation.
8.

Doetsch F, Caille I, Lim DA, Garca-Verdugo JM, Alvarez-Buylla A:


Subventricular zone astrocytes are neural stem cells in the
adult mammalian brain. Cell 1999, 97:703-716.

9.

Palmer TD, Willhoite AR, Gage FH: Vascular niche for adult
hippocampal neurogenesis. J Comp Neurol 2000, 425:479-494.

10. Seri B, Garcia-Verdugo JM, McEwen BS, Alvarez-Buylla A:


Astrocytes give rise to new neurons in the adult mammalian
hippocampus. J Neurosci 2001, 21:7153-7160.
11. Lim DA, Alvarez-Buylla A: Interaction between astrocytes and
adult subventricular zone precursors stimulates neurogenesis.
Proc Natl Acad Sci USA 1999, 96:7526-7531.
12. Song H, Stevens CF, Gage FH: Astroglia induce neurogenesis
from adult neural stem cells. Nature 2002, 417:39-44.
13. Capela A, Temple S: LeX/ssea-1 is expressed by adult mouse
 CNS stem cells, identifying them as non-ependymal.
Neuron 2002, 35:865-875.
This paper shows that the cell-surface carbohydrate LeX/SSEA-1,
expressed by pluripotent stem cells, is also expressed in adult germinal
regions. LeX is likely an important player in the stem cell niche, regulating
the accessibility and influence of ligands. It is the first cell-surface marker
identified that encompasses all neurosphere-forming cells in the adult
SVZ. The authors also show that ependymal cells are LeX negative and
purified ependymal cells do not make neurospheres.
14. Laywell ED, Rakic P, Kukekov VG, Holland EC, Steindler DA:
Identification of a multipotent astrocytic stem cell in the
immature and adult mouse brain. Proc Natl Acad Sci USA 2000,
97:13883-13888.
15. Chiasson BJ, Tropepe V, Morshead CM, van der Kooy D: Adult
mammalian forebrain ependymal and subependymal cells
demonstrate proliferative potential, but only subependymal
cells have neural stem cell characteristics. J Neurosci 1999,
19:4462-4471.

1.

Temple S: The development of neural stem cells. Nature 2001,


414:112-117.

16. Doetsch F, Petreanu L, Caille I, Garcia-Verdugo JM,


 Alvarez-Buylla A: EGF converts transit-amplifying neurogenic
precursors in the adult brain into multipotent stem cells.
Neuron 2002, 36:1021-1034.
In this paper, we used a combination of approaches to show that the
transit-amplifying C cells are the majority of EGF responsive cells both
in vivo and in vitro. C cells express the EGF-R and selective killing of C
cells in mice expressing herpes simplex virus thymidine kinase under the
Dlx2 promoter results in a dramatic reduction in the in vivo proliferative
response to EGF and loss of 70% of neurospheres in vitro. FACS
(fluorescence activated cell sorting) purified C cells are self-renewing
and multipotent, revealing that stem cell potential is not only restricted to
the primary stem cells.

2.

Doetsch F, Scharff C: Challenges for brain repair: insights from


adult neurogenesis in birds and mammals. Brain Behav Evol
2001, 58:306-322.

17. Johansson CB, Momma S, Clarke DL, Risling M, Lendahl U,


Frisen J: Identification of a neural stem cell in the adult
mammalian central nervous system. Cell 1999, 96:25-34.

3.

Spradling A, Drummond-Barbosa D, Kai T: Stem cells find their


niche. Nature 2001, 414:98-104.

4.

Nait-Oumesmar B, Decker L, Lachapelle F, Avellana-Adalid V,


Bachelin C, Van Evercooren AB: Progenitor cells of the adult
mouse subventricular zone proliferate, migrate and
differentiate into oligodendrocytes after demyelination.
Eur J Neurosci 1999, 11:4357-4366.

18. Lim DA, Tramontin AD, Trevejo JM, Herrera DG,


Garcia-Verdugo JM, Alvarez-Buylla A: Noggin antagonizes BMP
signaling to create a niche for adult neurogenesis.
Neuron 2000, 28:713-726.

References and recommended reading


Papers of particular interest, published within the annual period of
review, have been highlighted as:
 of special interest
 of outstanding interest

5.

Doetsch F, Garcia-Verdugo JM, Alvarez-Buylla A: Cellular


composition and three-dimensional organization of the

Current Opinion in Genetics & Development 2003, 13:543550

19. Stumm RK, Rummel J, Junker V, Culmsee C, Pfeiffer M,


Krieglstein J, Hollt V, Schulz S: A dual role for the SDF-1/CXCR4
chemokine receptor system in adult brain: isoform-selective
regulation of SDF-1 expression modulates CXCR4-dependent
neuronal plasticity and cerebral leukocyte recruitment after
focal ischemia. J Neurosci 2002, 22:5865-5878.
www.current-opinion.com

A niche for adult neural stem cells Doetsch 549

20. Lu M, Grove EA, Miller RJ: Abnormal development of the


hippocampal dentate gyrus in mice lacking the CXCR4
chemokine receptor. Proc Natl Acad Sci USA 2002,
99:7090-7095.

38. Galli R, Fiocco R, De Filippis L, Muzio L, Gritti A, Mercurio S,


Broccoli V, Pellegrini M, Mallamaci A, Vescovi AL: Emx2 regulates
the proliferation of stem cells of the adult mammalian central
nervous system. Development 2002, 129:1633-1644.

21. Klein RS, Rubin JB, Gibson HD, DeHaan EN, Alvarez-Hernandez X,
Segal RA, Luster AD: SDF-1 alpha induces chemotaxis and
enhances Sonic hedgehog-induced proliferation of cerebellar
granule cells. Development 2001, 128:1971-1981.

39. Shimazaki T, Shingo T, Weiss S: The ciliary neurotrophic factor/


leukemia inhibitory factor/gp130 receptor complex operates in
the maintenance of mammalian forebrain neural stem cells.
J Neurosci 2001, 21:7642-7653.

22. Lu Q, Sun EE, Klein RS, Flanagan JG: Ephrin-B reverse signaling
is mediated by a novel PDZ-RGS protein and selectively inhibits
G protein-coupled chemoattraction. Cell 2001, 105:69-79.

40. Groszer M, Erickson R, Scripture-Adams DD, Lesche R, Trumpp A,


Zack JA, Kornblum HI, Liu X, Wu H: Negative regulation of neural
stem/progenitor cell proliferation by the Pten tumor
suppressor gene in vivo. Science 2001, 294:2186-2189.

23. Conover JC, Doetsch F, Garcia-Verdugo JM, Gale NW,


Yancopoulos GD, Alvarez-Buylla A: Disruption of Eph/ephrin
signaling affects migration and proliferation in the adult
subventricular zone. Nat Neurosci 2000, 3:1091-1097.
24. Lai K, Kaspar BK, Gage FH, Schaffer DV: Sonic hedgehog
regulates adult neural progenitor proliferation in vitro and
in vivo. Nat Neurosci 2003, 6:21-27.
25. Perez-Moreno M, Jamora C, Fuchs E: Sticky business:
orchestrating cellular signals at adherens junctions. Cell 2003,
112:535-548.
26. Reya T, Duncan AW, Ailles L, Domen J, Scherer DC, Willert K,
Hintz L, Nusse R, Weissman IL: A role for Wnt signalling in selfrenewal of haematopoietic stem cells. Nature 2003,
423:409-414.
27. Lobo MV, Alonso FJ, Redondo C, Lopez-Toledano MA, Caso E,
Herranz AS, Paino CL, Reimers D, Bazan E: Cellular
characterization of epidermal growth factor-expanded freefloating neurospheres. J Histochem Cytochem 2003, 51:89-103.
28. Chenn A, Walsh CA: Regulation of cerebral cortical size by

control of cell cycle exit in neural precursors. Science 2002,
297:365-369.
Overexpression of constitutively active b-catenin in neuroepithelial precursors results in the expansion of the precursor pool due to increased
cell cycle re-entry, perhaps as a result of switching to symmetric division,
and is accompanied by the appearance of sulci and gyri-like folds in
the brain.
29. Lambert JD, Nagy LM: Asymmetric inheritance of centrosomally
localized mRNAs during embryonic cleavages. Nature 2002,
420:682-686.
30. Morshead CM, Reynolds BA, Craig CG, McBurney MW,
Staines WA, Morassutti D, Weiss S, Van der Kooy D: Neural stem
cells in the adult mammalian forebrain: a relatively quiescent
subpopulation of subependymal cells. Neuron 1994,
13:1071-1082.
31. Jin K, Zhu Y, Sun Y, Mao XO, Xie L, Greenberg DA: Vascular
endothelial growth factor (VEGF) stimulates neurogenesis in
vitro and in vivo. Proc Natl Acad Sci USA 2002, 99:11946-11950.
32. Shingo T, Gregg C, Enwere E, Fujikawa H, Hassam R, Geary C,
Cross JC, Weiss S: Pregnancy-stimulated neurogenesis in the
adult female forebrain mediated by prolactin. Science 2003,
299:117-120.

41. Nakatomi H, Kuriu T, Okabe S, Yamamoto S, Hatano O,


 Kawahara N, Tamura A, Kirino T, Nakafuku M: Regeneration of
hippocampal pyramidal neurons after ischemic brain injury by
recruitment of endogenous neural progenitors. Cell 2002,
110:429-441.
Infusion of bFGF and EGF after ischemia stimulates the regeneration of
pyramidal neurons in the hippocampus. This is accompanied by some
recovery of lost brain function in spatial cognitive tasks, as assayed by the
Morris water maze. The functional recovery of a damaged non-neurogenic brain region after stimulation of endogenous progenitors highlights
the potential for brain repair.
42. Louissaint A Jr, Rao S, Leventhal C, Goldman SA: Coordinated
interaction of neurogenesis and angiogenesis in the adult
songbird brain. Neuron 2002, 34:945-960.
43. Monje ML, Mizumatsu S, Fike JR, Palmer TD: Irradiation induces
neural precursor-cell dysfunction. Nat Med 2002, 8:955-962.
44. Heissig B, Hattori K, Dias S, Friedrich M, Ferris B, Hackett NR,
Crystal RG, Besmer P, Lyden D, Moore MA et al.: Recruitment of
stem and progenitor cells from the bone marrow niche requires
MMP-9 mediated release of kit-ligand. Cell 2002, 109:625-637.
45. Bergers G, Brekken R, McMahon G, Vu TH, Itoh T, Tamaki K,
Tanzawa K, Thorpe P, Itohara S, Werb Z et al.: Matrix
metalloproteinase-9 triggers the angiogenic switch during
carcinogenesis. Nat Cell Biol 2000, 2:737-744.
46. Gates MA, Thomas LB, Howard EM, Laywell ED, Sajin B, Faissner A,
Gotz B, Silver J, Steindler DA: Cell and molecular analysis of the
developing and adult mouse subventricular zone of the cerebral
hemispheres. J Comp Neurol 1995, 361:249-266.
47. Jankovski A, Sotelo C: Subventricular zone-olfactory bulb
migratory pathway in the adult mouse: cellular composition
and specificity as determined by heterochronic and
heterotopic transplantation. J Comp Neurol 1996, 371:376-396.
48. Jacques TS, Relvas JB, Nishimura S, Pytela R, Edwards GM,
Streuli CH, ffrench-Constant C: neural precursor cell chain
migration and division are regulated through different B1
integrins. Development 1998, 125:3167-3177.
49. Murase S, Horwitz AF: Deleted in colorectal carcinoma and
differentially expressed integrins mediate the directional
migration of neural precursors in the rostral migratory stream.
J Neurosci 2002, 22:3568-3579.

33. Peterson DA: Stem cells in brain plasticity and repair. Curr Opin
Pharmacol 2002, 2:34-42.

50. Bernfield M, Gotte M, Park PW, Reizes O, Fitzgerald ML,


Lincecum J, Zako M: Functions of cell surface heparan sulfate
proteoglycans. Annu Rev Biochem 1999, 68:729-777.

34. Doetsch F, Garcia-Verdugo JM, Caille I, Alvarez-Buylla A, Chao MV,


Casaccia-Bonnefil P: Lack of the cell cycle inhibitor p27Kip1
results in selective increase of transit amplifying cells for adult
neurogenesis. J Neurosci 2002, 22:2246-2254.

51. Isshiki T, Pearson B, Holbrook S, Doe CQ: Drosophila neuroblasts


sequentially express transcription factors which specify the
temporal identity of their neuronal progeny. Cell 2001,
106:511-521.

35. Coskun V, Luskin MB: The expression pattern of the cell cycle
inhibitor p19(Ink4d) by progenitor cells of the rat embryonic
telencephalon and neonatal anterior subventricular zone.
J Neurosci 2001, 21:3092-3103.

52. Bertrand N, Castro DS, Guillemot F: Proneural genes and the


specification of neural cell types. Nat Rev Neurosci 2002,
3:517-530.

36. Chojnacki A, Shimazaki T, Gregg C, Weinmaster G, Weiss S:


Glycoprotein 130 signaling regulates Notch1 expression and
activation in the self-renewal of mammalian forebrain neural
stem cells. J Neurosci 2003, 23:1730-1741.
37. Nakamura Y, Sakakibara S, Miyata T, Ogawa M, Shimazaki T,
Weiss S, Kageyama R, Okano H: The bHLH gene hes1 as a
repressor of the neuronal commitment of CNS stem cells.
J Neurosci 2000, 20:283-293.
www.current-opinion.com

53. Fisher AG, Merkenschlager M: Gene silencing, cell fate and


nuclear organisation. Curr Opin Genet Dev 2002, 12:193-197.
54. Takizawa T, Nakashima K, Namihira M, Ochiai W, Uemura A,
 Yanagisawa M, Fujita N, Nakao M, Taga T: DNA methylation is a
critical cell-intrinsic determinant of astrocyte differentiation in
the fetal brain. Dev Cell 2001, 1:749-758.
This paper highlights the importance of the epigenetic state of a cell in
regulating its responsiveness to extrinsic signals. DNA methylation of a
STAT3 binding element in the GFAP promoter of embryonic neuroepitheCurrent Opinion in Genetics & Development 2003, 13:543550

550 Differentiation and gene regulation

lial cells inhibits their differentiation into astrocytes by preventing STAT3


binding and transcriptional activation. Forced demethylation results in the
precocious acquisition of responsiveness to gliogenic signals.

HDACs. Contiguous genes, without RE-1 sites, are simultaneously


silenced by the recruitment and spreading of silencing machinery along
adjacent chromatin.

55. Ballas N, Battaglioli E, Atouf F, Andres ME, Chenoweth J,



Anderson ME, Burger C, Moniwa M, Davie JR, Bowers WJ et al.:
Regulation of neuronal traits by a novel transcriptional
complex. Neuron 2001, 31:353-365.
See annotation [56].

57. Ramalho-Santos M, Yoon S, Matsuzaki Y, Mulligan RC, Melton DA:


Stemness: transcriptional profiling of embryonic and adult
stem cells. Science 2002, 298:597-600.

56. Lunyak VV, Burgess R, Prefontaine GG, Nelson C, Sze SH,


 Chenoweth J, Schwartz P, Pevzner PA, Glass C, Mandel G et al.:
Corepressor-dependent silencing of chromosomal regions
encoding neuronal genes. Science 2002, 298:1747-1752.
These papers [55,56] demonstrate the coordinate regulation of genes
via repression. REST and Co-REST bind to RE-1 sites as a complex with

Current Opinion in Genetics & Development 2003, 13:543550

58. Lessard J, Sauvageau G: Bmi-1 determines the proliferative


capacity of normal and leukaemic stem cells. Nature 2003,
423:255-260.
59. Park IK, Qian D, Kiel M, Becker MW, Pihalja M, Weissman IL,
Morrison SJ, Clarke MF: Bmi-1 is required for maintenance of
adult self-renewing haematopoietic stem cells. Nature 2003,
423:302-305.

www.current-opinion.com

S-ar putea să vă placă și