Neural Stem Cells: Developmental Mechanisms and Disease Modeling

Download as pdf or txt
Download as pdf or txt
You are on page 1of 6

Cell and Tissue Research (2018) 371:1–6

https://doi.org/10.1007/s00441-017-2738-1

EDITORIAL

Neural stem cells: developmental mechanisms and disease modeling


Xinyu Zhao 1,2 & Darcie L. Moore 1

Published online: 2 December 2017


# Springer-Verlag GmbH Germany, part of Springer Nature 2017

Summary Stem cells

The astonishing progress in the field of stem cell biology The term Bstem cells^ first appeared in the scientific literature
during the past 40 years has transformed both science and in 1868 by the German biologist Ernst Haeckel (Haeckel
medicine. Neural stem cells (NSCs) are the stem cells of 1868). In his writings (Haeckel 1868), Bstem cells^ had two
the nervous system. During development they give rise to distinct meanings: one is the unicellular evolutionary origin of
the entire nervous system. In adults, a small number of all multicellular organisms and the other is the fertilized egg
NSCs remain and are mostly quiescent; however, ample giving rise to all other cell types of the body. The latter defi-
evidence supports their important roles in plasticity, ag- nition has evolved into the modern definition of stem cells—
ing, disease and regeneration of the nervous system. cells that can divide to self-renew and to differentiate into
Because NSCs are regulated by both intrinsic genetic other cell types in tissues and organs (Li and Zhao 2008;
and epigenetic programs and extrinsic stimuli transduced Ramalho-Santos and Willenbring 2007).
through the stem cell niche, dysregulation of NSCs due to The behavior and fate of stem cells are strongly influenced
either genetic causes or environmental impacts may lead by their specific anatomical locations and surrounding cell
to disease. Therefore, extensive investigations in the past types, called “the stem cell niche.” The niche provides phys-
decades have been devoted to understanding how NSCs ical support to host or anchor stem cells and supplies factors to
are regulated. On the other hand, ever since their discov- maintain and regulate them (Li and Zhao 2008). Stem cells are
ery, NSCs have been a focal point for cell-based therapeu- also regulated by intrinsic signaling cascades and transcrip-
tic strategies in the brain and spinal cord. The limited tional mechanisms, some of which are common among all
number of NSCs residing in the tissue has been a limiting stem cells and others that are unique to specific types. Some
factor for their clinical applications. Although recent ad- of the best known regulators include TGF-β, BMP, Smad,
vancements in embryonic and induced pluripotent stem Wnt, Notch and EGF fibroblast growth factors (Jobe et al.
cells have provided novel sources for NSCs, several chal- 2012; Li and Zhao 2008). Therefore, stem cells are regulated
lenges remain. In this special issue, leaders and experts in by complex mechanisms in both temporal- and context-
NSCs summarize our current understanding of NSC mo- specific manners to maintain their unique characteristics.
lecular regulation and the importance of NSCs for disease Understanding stem cell regulation gives us the opportunity
modeling and translational applications. to explore mechanisms of development, as well as disorders
resulting from their dysfunction.

* Xinyu Zhao NSCs in development


[email protected]
During development, the central nervous system (CNS) is
Darcie L. Moore generated from a small number of neural stem cells (NSCs)
[email protected]
lining the neural tube (Kriegstein and Alvarez-Buylla 2009).
1
Department of Neuroscience, Waisman Center and University of A great deal of experimental evidence has demonstrated that
Wisconsin-Madison, Madison, WI 53705, USA radial glia, the NSCs during mammalian CNS development,
2
Waisman Center and University of Wisconsin-Madison, undergo both symmetric divisions to expand the NSC pool
Madison, WI 53705, USA and asymmetric divisions to give rise to intermediate
2 Cell Tissue Res (2018) 371:1–6

progenitors (IPCs) and the differentiated cell types. The three behaviors such as pattern separation (Aimone et al. 2011)
major cell types in the CNS arise from NSCs in a temporally and spatial learning (Dupret et al. 2008), as well as
defined sequence, with neurons appearing first, followed by hippocampus-associated learning, memory and executive
astrocytes and then oligodendrocytes (Okano and Temple functions (Kempermann et al. 2015).
2009). The technical advancement of live imaging and geno- Significant effort has been devoted into understanding the
mic tools have allowed for the identification of human- regulation of adult neurogenesis. As a result, we now know
specific NSC populations (e.g., outer radial glia, or oRG) lo- that many extrinsic stimuli and intrinsic mechanisms can af-
cated at the outer subventricular zone (SVZ) (Gertz et al. fect this process. Mouse genetic studies have clearly demon-
2014). These oRG are essential for cortical expansion to strated the important role of transcriptional regulation of
achieve the large size of the human cortex. Single-cell geno- NSCs through intrinsic genetic mechanisms (Hsieh and
mic technologies have identified specific oRG markers that Zhao 2016). Some examples include SOXC family proteins
might be used for further characterization of these cells (Liu (Kavyanifar et al. 2018, in this issue), Bmi-1 (Molofsky et al.
et al. 2016, Pollen et al. 2014). Investigating the regulatory 2003), Sox2 (Ferri et al. 2004; Graham et al. 1999), PTEN
mechanisms governing the self-renewal and fate specification (Bonaguidi et al. 2011) and Notch (Zhang et al. 2018, in this
of NSCs, especially human-specific developmental features, issue). In addition, epigenetic regulation by DNA methylation
has significantly contributed to our understanding of human pathways (e.g., Mbd1, Mecp2, Dnmt, Tet) (Noguchi et al.
brain development and developmental diseases. In addition, 2015; Smrt et al. 2007; Tsujimura et al. 2009; Zhang et al.
this knowledge has also helped scientists refine protocols for 2013; Zhao et al. 2003), chromatin remodeling (e.g., BAF,
pluripotent stem cell differentiation into specific nervous sys- BRG1) (Ninkovic et al. 2013; Petrik et al. 2015; Tuoc et al.
tem cell types for both therapeutic goals and disease 2017) and noncoding RNAs (Liu et al. 2010; Anderson and
modeling. Lim 2018, in this issue) play important roles. Many growth
factors, signaling molecules and neurotransmitters have been
shown to regulate neurogenesis (Kempermann et al. 2015).
NSCs in the adult brain Catavero et al. (2018, in this issue) review the role of
GABA circuits, signaling and receptors in regulating develop-
In adult brains, NSCs are reduced and become restricted to ment of adult-born cells, as well as the molecular players that
specific brain regions. In rodents, both NSCs and ongoing modulate GABA signaling. Because progenitors with
neurogenesis have been widely documented in the SVZ of multipotent differentiation potentials have been found in brain
the lateral ventricles and the subgranular zone (SGZ) of the regions without active neurogenesis (Palmer et al. 1997), it is
dentate gyrus (DG) of the hippocampus (Kempermann et al. hypothesized that these progenitors might be manipulated to
2015). In humans, experimental evidence has supported on- become neuron-competent in vivo so that they can contribute
going neurogenesis in the hippocampus (Eriksson et al. 1998; to brain generation (Wang and Zhang 2018, in this issue).
Spalding et al. 2013). The confirmation of mammalian adult A great amount of literature has documented how physio-
neurogenesis in the 1990s was one of the most exciting mo- logical activities and an enriched environment influence adult
ments in science in the 21st century. Not only did it overthrow neurogenesis (Kempermann et al. 2015). However, as sum-
the prevailing dogma suggesting no neurons were made in the marized by Eisinger and Zhao (2018, in this issue), the genes
adult brain but it also hinted that these adult NSCs could be and gene network involved in these changes within NSCs
utilized for neural repair in disease and following injury. Forty have not been systematically analyzed at genome wide levels.
years later, we have learned a lot about NSCs. In the adult Adult neurogenesis is also influenced by diseases including
rodent SVZ, neurogenesis has been shown to be important epilepsy (Parent and Lowenstein 1997), stroke (Zhang and
for olfactory function and olfactory learning (Alonso et al. Chopp 2016), depression (Dranovsky and Hen 2006;
2006). During development, a subset of slowly-dividing Kempermann et al. 2003) and injury (Morshead and Ruddy
NSCs are set aside to be the NSCs of the SVZ in the postnatal 2018, in this issue). Thodeson et al. (2018, in this issue) fur-
and adult brain (Fuentealba et al. 2015; Furutachi et al. 2015). ther summarize the contribution and dysregulation of adult
The majority of neurogenic radial glia, however, become as- neurogenesis in epilepsy and discuss how we can translate
trocytes and ependymal cells at the end of embryonic these findings to human therapeutics by using patient-
neurogenesis (Noctor et al. 2004). A subset of these astrocytes derived neurons to study monogenic epilepsy-in-a-dish.
persist as NSCs in specialized niches in the adult brain and Aging affects every individual and is a major risk factor for
continuously generate neurons that functionally integrate into many diseases. One of the strongest negative regulators of
restricted brain regions (Doetsch 2003). In the hippocampus, adult neurogenesis is aging. Both intrinsic and extrinsic com-
radial glia-like stem cells of the SGZ make newborn neurons ponents regulate the limitations of NSC proliferation and
throughout life (Goritz and Frisen 2012). These newborn neu- function (Moore and Jessberger 2017; Seib and Martin-
rons integrate into the circuity of the DG, contributing to Villalba 2015). In this issue, Mosher and Schaffer (2018)
Cell Tissue Res (2018) 371:1–6 3

and Morshead and Ruddy (2018) examine factors such as neural differentiation protocol, most neural progenitors ob-
secreted signals, cell contact-dependent signals and extracel- tained are forebrain excitatory progenitors that produce mostly
lular matrix cues that control neurogenesis in an age- forebrain glutamatergic excitatory neurons. However, the pu-
dependent manner and define these signals by the extrinsic rity and layer-specific composition of these progenitors, as
mechanism through which they are presented to the NSCs. well as neurons, vary significantly from experiment to exper-
Smith et al. (2018, in this issue) discuss how age-related iment, cell line to cell line and lab to lab. In addition, differ-
changes in the blood, such as blood-borne factors and periph- entiation into specific types of neurons with high purity has
eral immune cells, contribute to the age-related decline in always been a challenging goal. Much effort has been devoted
adult neurogenesis in the mammalian brain. into improving the efficiency of dopaminergic neuron and
Despite the extensive knowledge we have gained regarding GABAergic neuron differentiation with great success (Hu
adult neurogenesis, critical questions remain. For example, the et al. 2010). However, the brain has many other types of neu-
control of the functional integration of new neurons remains a rons. Vadodaria et al. (2018, in this issue) discuss how to
mystery. It has been shown that adult NSC-differentiated new- generate serotonergic neurons, a type of neuron highly rele-
born neurons exhibit a critical period for sensitivity to external vant to psychiatric disorders. To better understand the molec-
stimuli (Bergami et al. 2015) and a heightened sensitivity to ular control of human PSC and NSC differentiation, where
seizures (Kron et al. 2010). It remains unclear how new neu- protocols result in a large amount of cellular heterogeneity in
rons choose their connections. Jahn and Bergami (2018, in identity and response, analysis must be done at the level of
this issue) further discuss the critical period and its regulators single cells. Harbom et al. (2018, in this issue) summarize how
during adult-newborn neuron development. new state-of-the-art single-cell analysis methods may help to
Understanding the extrinsic and intrinsic regulation of define differentiation from pluripotent cells.
adult NSCs and their newborn progeny and their response to The advancement in iPSC and gene editing technology has
both positive and negative stimuli will further illuminate their transformed the field of human disease modeling. As in many
role in disease, injury, stress and brain function. human disorders, especially neuropsychiatric disorders,
mouse models have been useful. Yet there are several critical
reasons why it is necessary to use human cells to define the
Pluripotent stem cell-derived NSCs underlying mechanisms that lead to human patient character-
istics, particularly those affecting the nervous system. For ex-
Human pluripotent stem cells (PSCs), including human em- ample, in fragile X syndrome (FXS), the epigenetic silencing
bryonic stem cells (ESCs) and induced PSCs (iPSCs), offer a of the Fragile X Mental Retardation Gene 1 (FMR1) gene that
model system to reveal cellular and molecular events causes FXS occurs only in humans. Mice engineered to mimic
underlying normal and abnormal neural development in the human mutation in the FMR1 gene do not show the same
humans. ESCs are pluripotent cells derived from the inner methylation and silencing characteristics of the gene as in
cell mass of blastocyst stage preimplantation embryos, humans (Brouwer et al. 2007). These results indicate that
which were first isolated in 1981 from mouse by Evans and some epigenetic mechanisms in human and mice are different
Kaufman (1981) and later, in 1998, from humans by Thomson and preclude the ability to study epigenetic mechanisms of
et al. (1998). Human ESCs are invaluable in the study of early FMR1 silencing in mouse models of FXS (Bhattacharyya
embryonic development, allowing us to identify critical regu- and Zhao 2016). In this issue, Li and Shi discuss disease
lators of cell commitment, differentiation and adult cell modeling using human PSC-differentiated neural progenitors
reprogramming (Dvash et al. 2006; Ren et al. 2009). iPSCs (Li et al. 2018), and Brito et al. specifically focus on modeling
are reprogrammed from somatic cells by forced expression of autism spectrum disorder (Brito et al. 2018).
stem cell genes and have the characteristics of ESCs (Okita
et al. 2007; Yu et al. 2007). The development of iPSC tech-
nology has allowed us access to cells of the human nervous Use of NSCs as therapy
system through reprogramming of patient-derived cells, revo-
lutionizing our ability to study human development and The use of NSCs as a treatment strategy in CNS disease and
diseases. injury has been tested for decades. Parkinsons’ disease specif-
To generate neural cells from either ESCs or iPSCs, the first ically has gained the most momentum for potential therapeutic
step is neural induction. Through actions of a number of acti- benefits (Studer 2017); however, similar work has been per-
vators and inhibitors of cell signaling pathways, this process formed in Huntington’s disease, stroke and following spinal
yields neural epithelial cell-like NSCs and then intermediate cord injury (for a review on this topic, see Vishwakarma et al.
neural progenitors, resembling embryonic development. 2014). In some of these paradigms, NSCs are expected to
Despite many advances, a major hurdle of neural differentia- differentiate into a specific cell type in the local CNS environ-
tion is lineage control. Using a Bstandard^ dorsal forebrain ment; in other cases, they are in a supportive role. In this issue,
4 Cell Tissue Res (2018) 371:1–6

Kameda et al. explore progress in using NSCs as a therapy Acknowledgements We thank Klaus Unsicker for his encouragement
and support and Jutta Jäger for her help with invitations and communi-
following spinal cord injury (Kameda et al. 2018).
cations with authors and reviewers. This work was supported by grants
from the US National Institutes of Health (R01MH078972,
R56MH113146, R21NS098767 and R21NS095632 to X.Z,
U54HD090256 to the Waisman Center), University of Wisconsin
Bypassing NSCs? (UW)-Madison Vilas Trust (Kellett Mid-Career Award to X.Z.) and
UW-Madison and Wisconsin Alumni Research Foundation (WARF to
While the development of PSC technologies has been a sci- X.Z.), Jenni and Kyle Professorship (to X.Zhao), a Sloan Research
Fellowship (to D.L.M.), a Junior Faculty Grant from the American
entific breakthrough for future studies, there are limitations Federation for Aging Research (to D.L.M.) and startup funds from UW-
and risks that may be associated with their use. ESCs, iPSCs Madison School of Medicine and Public Health, WARF and the
and their differentiated NSCs are dividing cells. Either trans- Neuroscience Department (to D.L.M.).
plantation of NSCs or in vivo reprogramming of endogenous
cells into NSCs could lead to tumorigenesis. In addition,
reprogramming somatic cells into iPSCs results in a loss of References
some epigenetic signatures of disease and aging, which are
critical for disease modeling (Mertens et al. 2015; Miller Aimone JB, Deng W, Gage FH (2011) Resolving new memories: a crit-
et al. 2013; Ocampo et al. 2016). In recent years, direct ical look at the dentate gyrus, adult neurogenesis, and pattern sepa-
ration. Neuron 70:589–596
reprogramming of fibroblasts or other cell types into induced Alonso M, Viollet C, Gabellec MM, Meas-Yedid V, Olivo-Marin JC,
neurons (iN) has been developed (for review, see Mertens Lledo PM (2006) Olfactory discrimination learning increases the
et al. 2016). Remarkably, a growing number of studies have survival of adult-born neurons in the olfactory bulb. J Neurosci 26:
demonstrated that such direct reprogramming also can be ef- 10508–10513
Anderson RA, Lim DA (2018) Forging our understanding of lncRNAs in
fective in vivo. Wang and Zhang (2018, in this issue) summa-
the brain. Cell Tissue Res. https://doi.org/10.1007/s00441-017-
rize recent progress of in vivo reprogramming into new neu- 2711-z
rons and present how this method can be used for spinal cord Bergami M, Masserdotti G, Temprana SG, Motori E, Eriksson TM,
injury. Gobel J, Yang SM, Conzelmann KK, Schinder AF, Gotz M,
In cellular reprogramming, the cells targeted and the genet- Berninger B (2015) A critical period for experience-dependent re-
modeling of adult-born neuron connectivity. Neuron 85:710–717
ic factors used vary; however, the biggest difference is that Bhattacharyya A, Zhao X (2016) Human pluripotent stem cell models of
some protocols push cells through a NSC stage, whereas fragile X syndrome. Mol Cell Neurosci 73:43–51
others skip these stages (Gascon et al. 2017; Guo et al. Bonaguidi MA, Wheeler MA, Shapiro JS, Stadel RP, Sun GJ, Ming GL,
2014; Wang et al. 2016). Bypassing this developmental stage Song H (2011) In vivo clonal analysis reveals self-renewing and
multipotent adult neural stem cell characteristics. Cell 145:1142–
has both pros and cons and may lead to a completely novel 1155
path towards lineage commitment (discussed by Falk and Brito A, Russo FB, Muotri AR, Beltrão-Braga PCB (2018) Autism spec-
Karow 2018 in this issue). trum disorders and disease modeling using stem cells. Cell Tissue
Res. https://doi.org/10.1007/s00441-017-2685-x
Brouwer JR, Mientjes EJ, Bakker CE, Nieuwenhuizen IM, Severijnen
LA, Van der Linde HC, Nelson DL, Oostra BA, Willemsen R
Perspective (2007) Elevated Fmr1 mRNA levels and reduced protein expression
in a mouse model with an unmethylated fragile X full mutation. Exp
Cell Res 313:244–253
NSCs are fascinating and promising cells because of their Catavero C, Bao H, Song J (2018) Neural mechanisms underlying
capability, flexibility and multiplicity. Understanding how GABAergic regulation of adult hippocampal neurogenesis. Cell
NSCs function provides important knowledge in the develop- Tissue Res. https://doi.org/10.1007/s00441-017-2668-y
ment, adaptation, disease, regeneration and rehabilitation of Doetsch F (2003) A niche for adult neural stem cells. Curr Opin Genet
Dev 13:543–550
the nervous system. The studies of cortical development and Dranovsky A, Hen R (2006) Hippocampal neurogenesis: regulation by
adult neurogenesis using rodent models have contributed sig- stress and antidepressants. Biol Psychiatry 59:1136–1143
nificantly to our knowledge about NSCs and will continually Dupret D, Revest JM, Koehl M, Ichas F, De Giorgi F, Costet P, Abrous
yield important new information, taking advantage of novel DN, Piazza PV (2008) Spatial relational memory requires hippo-
campal adult neurogenesis. PLoS ONE 3:e1959
genetic and imaging technologies. However, using human
Dvash T, Ben-Yosef D, Eiges R (2006) Human embryonic stem cells as a
NSCs provides us with a window to investigate human- powerful tool for studying human embryogenesis. Pediatr Res 60:
specific aspects of development and disease mechanisms, 111–117
which is potentiated by the fast advancement of stem cell Eisinger BE, Zhao X (2017) Identifying Molecular Mediators of
and gene editing technologies. Challenges still remain regard- Environmentally-Enhanced Neurogenesis. Cell Tissue Res. https://
doi.org/10.1007/s00441-017-2718-5
ing cell lineage control, in vivo NSC behavior, three- Eriksson PS, Perfilieva E, Bjork-Eriksson T, Alborn AM, Nordborg C,
dimensional cellular interactions and preservation of epigenet- Peterson DA, Gage FH (1998) Neurogenesis in the adult human
ic and aging marks. hippocampus. Nat Med 4:1313–1317
Cell Tissue Res (2018) 371:1–6 5

Evans MJ, Kaufman MH (1981) Establishment in culture of Li L, Chao J, Shi Y (2018) Modeling neurological diseases using iPSC-
pluripotential cells from mouse embryos. Nature 292:154–156 derived neural cells. Cell Tissue Res. https://doi.org/10.1007/
Falk S, Karow M (2018) Natural and forced neurogenesis: similar and yet s00441-017-2713-x
different. Cell Tissue Res. https://doi.org/10.1007/s00441-017- Li X, Zhao X (2008) Epigenetic regulation of mammalian stem cells.
2690-0 Stem Cells Dev 17:1043–1052
Ferri AL, Cavallaro M, Braida D, Di Cristofano A, Canta A, Vezzani A, Liu C, Teng ZQ, Santistevan NJ, Szulwach KE, Guo W, Jin P, Zhao X
Ottolenghi S, Pandolfi PP, Sala M, DeBiasi S, Nicolis SK (2004) (2010) Epigenetic regulation of miR-184 by MBD1 governs neural
Sox2 deficiency causes neurodegeneration and impaired stem cell proliferation and differentiation. Cell Stem Cell 6:433–444
neurogenesis in the adult mouse brain. Development 131:3805– Liu SJ, Nowakowski TJ, Pollen AA, Lui JH, Horlbeck MA, Attenello FJ,
3819 He D, Weissman JS, Kriegstein AR, Diaz AA, Lim DA (2016)
Fuentealba LC, Rompani SB, Parraguez JI, Obernier K, Romero R, Single-cell analysis of long non-coding RNAs in the developing
Cepko CL, Alvarez-Buylla A (2015) Embryonic origin of postnatal human neocortex. Genome Biol 17:67
neural stem cells. Cell 161:1644–1655 Mertens J, Marchetto MC, Bardy C, Gage FH (2016) Evaluating cell
Furutachi S, Miya H, Watanabe T, Kawai H, Yamasaki N, Harada Y, reprogramming, differentiation and conversion technologies in neu-
Imayoshi I, Nelson M, Nakayama KI, Hirabayashi Y, Gotoh Y roscience. Nat Rev Neurosci 17:424–437
(2015) Slowly dividing neural progenitors are an embryonic origin Mertens J, Paquola AC, Ku M, Hatch E, Bohnke L, Ladjevardi S,
of adult neural stem cells. Nat Neurosci 18:657–665 McGrath S, Campbell B, Lee H, Herdy JR, Goncalves JT, Toda T,
Gascon S, Masserdotti G, Russo GL, Gotz M (2017) Direct neuronal Kim Y, Winkler J, Yao J, Hetzer MW, Gage FH (2015) Directly
reprogramming: achievements, hurdles, and new roads to success. reprogrammed human neurons retain aging-associated
Cell Stem Cell 21:18–34 Transcriptomic signature s and r eve al a ge- re la t ed
Gertz CC, Lui JH, LaMonica BE, Wang X, Kriegstein AR (2014) Diverse Nucleocytoplasmic defects. Cell Stem Cell 17:705–718
behaviors of outer radial glia in developing ferret and human cortex. Miller JD, Ganat YM, Kishinevsky S, Bowman RL, Liu B, Tu EY,
J Neurosci 34:2559–2570 Mandal PK, Vera E, Shim JW, Kriks S, Taldone T, Fusaki N,
Goritz C, Frisen J (2012) Neural stem cells and neurogenesis in the adult. Tomishima MJ, Krainc D, Milner TA, Rossi DJ, Studer L (2013)
Cell Stem Cell 10:657–659 Human iPSC-based modeling of late-onset disease via progerin-
Graham JD, Hunt SM, Tran N, Clarke CL (1999) Regulation of the induced aging. Cell Stem Cell 13:691–705
expression and activity by progestins of a member of the SOX gene Molofsky AV, Pardal R, Iwashita T, Park IK, Clarke MF, Morrison SJ
family of transcriptional modulators. J Mol Endocrinol 22:295–304 (2003) Bmi-1 dependence distinguishes neural stem cell self-
renewal from progenitor proliferation. Nature 425:962–967
Guo Z, Zhang L, Wu Z, Chen Y, Wang F, Chen G (2014) In vivo direct
reprogramming of reactive glial cells into functional neurons after Moore DL, Jessberger S (2017) Creating age asymmetry: consequences
brain injury and in an Alzheimer's disease model. Cell Stem Cell 14: of inheriting damaged goods in mammalian cells. Trends Cell Biol
27:82–92
188–202
Morshead CM, Ruddy RM (2018) Home sweet home: the neural stem
Haeckel E (1868) Natürliche Schöpfungsgeschichte. Georg Reimer,
cell niche throughout development and after injury. Cell Tissue Res.
Berlin
https://doi.org/10.1007/s00441-017-2658-0
Harbom LJ, Michel N, McConnell MJ (2017) Single cell analysis of
Mosher KI, Schafer DV (2018) Influence of Hippocampal niche signals
diversity in human stem cell-derived neurons. Cell Tissue Res.
on neural stem cell functions during aging. Cell Tissue Res. https://
https://doi.org/10.1007/s00441-017-2728-3
doi.org/10.1007/s00441-017-2709-6
Hsieh J, Zhao X (2016) Genetics and Epigenetics in adult Neurogenesis.
Ninkovic J, Steiner-Mezzadri A, Jawerka M, Akinci U, Masserdotti G,
Cold Spring Harb Perspect Biol 8
Petricca S, Fischer J, von Holst A, Beckers J, Lie CD, Petrik D,
Hu BY, Weick JP, Yu J, Ma LX, Zhang XQ, Thomson JA, Zhang SC Miller E, Tang J, Wu J, Lefebvre V, Demmers J, Eisch A, Metzger
(2010) Neural differentiation of human induced pluripotent stem D, Crabtree G, Irmler M, Poot R, Gotz M (2013) The BAF complex
cells follows developmental principles but with variable potency. interacts with Pax6 in adult neural progenitors to establish a neuro-
Proc Natl Acad Sci U S A 107:4335–4340 genic cross-regulatory transcriptional network. Cell Stem Cell 13:
Jahn HM, Bergami M (2018) Critical periods regulating the circuit inte- 403–418
gration of adult-born hippocampal neurons. Cell Tissue Res. https:// Noctor SC, Martinez-Cerdeno V, Ivic L, Kriegstein AR (2004) Cortical
doi.org/10.1007/s00441-017-2677-x neurons arise in symmetric and asymmetric division zones and mi-
Jobe EM, McQuate AL, Zhao X (2012) Crosstalk among epigenetic grate through specific phases. Nat Neurosci 7:136–144
pathways regulates Neurogenesis. Front Neurosci 6:59 Noguchi H, Kimura A, Murao N, Matsuda T, Namihira M, Nakashima K
Kameda T, Imamura T, Nakashima K (2018) Epigenetic regulation of (2015) Expression of DNMT1 in neural stem/precursor cells is crit-
neural stem cell differentiation towards spinal cord regeneration. ical for survival of newly generated neurons in the adult hippocam-
Cell Tissue Res DOI. https://doi.org/10.1007/s00441-017-2656-2 pus. Neurosci Res 95:1–11
Kavyanifar A, Turan S, Lie DC (2018) SoxC transcription factors – mul- Ocampo A, Reddy P, Martinez-Redondo P, Platero-Luengo A, Hatanaka
tifunctional regulators of neurodevelopment. Cell Tissue Res. F, Hishida T, Li M, Lam D, Kurita M, Beyret E, Araoka T, Vazquez-
https://doi.org/10.1007/s00441-017-2708-7 Ferrer E, Donoso D, Roman JL, Xu J, Rodriguez Esteban C, Nunez
Kempermann G, Gast D, Kronenberg G, Yamaguchi M, Gage FH (2003) G, Nunez Delicado E, Campistol JM, Guillen I, Guillen P, Izpisua
Early determination and long-term persistence of adult-generated Belmonte JC (2016) In vivo amelioration of age-associated hall-
new neurons in the hippocampus of mice. Development 130:391– marks by partial reprogramming. Cell 167:1719–1733 e1712
399 Okano H, Temple S (2009) Cell types to order: temporal specification of
Kempermann G, Song H, Gage FH (2015) Neurogenesis in the adult CNS stem cells. Curr Opin Neurobiol 19:112–119
hippocampus. Cold Spring Harb Perspect Biol 7:a018812 Okita K, Ichisaka T, Yamanaka S (2007) Generation of germline-
Kriegstein A, Alvarez-Buylla A (2009) The glial nature of embryonic and competent induced pluripotent stem cells. Nature 448:313–317
adult neural stem cells. Annu Rev Neurosci 32:149–184 Palmer TD, Takahashi J, Gage FH (1997) The adult rat hippocampus
Kron MM, Zhang H, Parent JM (2010) The developmental stage of den- contains primordial neural stem cells. Mol Cell Neurosci 8:389–404
tate granule cells dictates their contribution to seizure-induced plas- Parent JM, Lowenstein DH (1997) Mossy fiber reorganization in the
ticity. J Neurosci 30:2051–2059 epileptic hippocampus. Curr Opin Neurol 10:103–109
6 Cell Tissue Res (2018) 371:1–6

Petrik D, Latchney SE, Masiulis I, Yun S, Zhang Z, Wu JI, Eisch AJ by the methyl-CpG-binding protein MeCP2. Exp Neurol 219:104–
(2015) Chromatin Remodeling factor Brg1 supports the early main- 111
tenance and late responsiveness of nestin-lineage adult neural stem Tuoc T, Dere E, Radyushkin K, Pham L, Nguyen H, Tonchev AB, Sun G,
and progenitor cells. Stem Cells 33:3655–3665 Ronnenberg A, Shi Y, Staiger JF, Ehrenreich H, Stoykova A (2017)
Pollen AA, Nowakowski TJ, Shuga J, Wang X, Leyrat AA, Lui JH, Li N, Ablation of BAF170 in developing and postnatal dentate Gyrus
Szpankowski L, Fowler B, Chen P, Ramalingam N, Sun G, Thu M, affects neural stem cell proliferation, differentiation, and learning.
Norris M, Lebofsky R, Toppani D, Kemp DW 2nd, Wong M, Mol Neurobiol 54:4618–4635
Clerkson B, Jones BN, Wu S, Knutsson L, Alvarado B, Wang J, Vadodaria KC, Stern ST, Marchetto MC, Gage FH (2018) Serotonin in
Weaver LS, May AP, Jones RC, Unger MA, Kriegstein AR, West JA psychiatry: in vitro disease modeling using patient-derived neurons.
(2014) Low-coverage single-cell mRNA sequencing reveals cellular Cell Tissue Res. https://doi.org/10.1007/s00441-017-2670-4
heterogeneity and activated signaling pathways in developing cere- Vishwakarma SK, Bardia A, Tiwari SK, Paspala SA, Khan AA (2014)
bral cortex. Nat Biotechnol 32:1053–1058 Current concept in neural regeneration research: NSCs isolation,
Ramalho-Santos M, Willenbring H (2007) On the origin of the term "stem characterization and transplantation in various neurodegenerative
cell". Cell Stem Cell 1:35–38 diseases and stroke: a review. J Adv Res 5:277–294
Ren JQ, Jin P, Wang E, Marincola FM, Stroncek DF (2009) MicroRNA Wang L, Zhang C-L (2017) Engineering new neurons: In vivo
and gene expression patterns in the differentiation of human embry- reprogramming in the mammalian brain and spinal cord. Cell
onic stem cells. J Transl Med 7 Tissue Res. https://doi.org/10.1007/s00441-017-2729-2
Seib DR, Martin-Villalba A (2015) Neurogenesis in the normal ageing
Wang LL, Su Z, Tai W, Zou Y, Xu XM, Zhang CL (2016) The p53
hippocampus: a mini-review. Gerontology 61:327–335
pathway controls SOX2-mediated reprogramming in the adult
Smith LK, White CWI, Villeda SA (2017) The systemic environment: at
mouse spinal cord. Cell Rep 17:891–903
the interface of aging and adult neurogenesis. Cell Tissue Res.
https://doi.org/10.1007/s00441-017-2715-8 Yu JY, Vodyanik MA, Smuga-Otto K, Antosiewicz-Bourget J, Frane JL,
Smrt RD, Eaves-Egenes J, Barkho BZ, Santistevan NJ, Zhao C, Aimone Tian S, Nie J, Jonsdottir GA, Ruotti V, Stewart R, Slukvin II,
JB, Gage FH, Zhao X (2007) Mecp2 deficiency leads to delayed Thomson JA (2007) Induced pluripotent stem cell lines derived
maturation and altered gene expression in hippocampal neurons. from human somatic cells. Science 318:1917–1920
Neurobiol Dis 27:77–89 Zhang R, Engler A, Taylor V (2018) Notch: an interactive player in
Spalding KL, Bergmann O, Alkass K, Bernard S, Salehpour M, Huttner neurogenesis and disease. Cell Tissue Res. https://doi.org/10.1007/
HB, Bostrom E, Westerlund I, Vial C, Buchholz BA, Possnert G, s00441-017-2641-9
Mash DC, Druid H, Frisen J (2013) Dynamics of hippocampal Zhang RR, Cui QY, Murai K, Lim YC, Smith ZD, Jin S, Ye P, Rosa L,
neurogenesis in adult humans. Cell 153:1219–1227 Lee YK, Wu HP, Liu W, Xu ZM, Yang L, Ding YQ, Tang F,
Studer L (2017) Strategies for bringing stem cell-derived dopamine neu- Meissner A, Ding C, Shi Y, Xu GL (2013) Tet1 regulates adult
rons to the clinic-the NYSTEM trial. Prog Brain Res 230:191–212 hippocampal neurogenesis and cognition. Cell Stem Cell 13:237–
Thodeson DM, Brulet R, Hsieh J (2018) Neural stem cells and epilepsy: 245
functional roles and disease-in-a-dish models. Cell Tissue Res. Zhang Z, Chopp M (2016) Neural stem cells and ischemic brain. J Stroke
https://doi.org/10.1007/s00441-017-2675-z 18:267–272
Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, Swiergiel JJ, Zhao X, Ueba T, Christie BR, Barkho B, McConnell MJ, Nakashima K,
Marshall VS, Jones JM (1998) Embryonic stem cell lines derived Lein ES, Eadie BD, Willhoite AR, Muotri AR, Summers RG, Chun
from human blastocysts. Science 282:1145–1147 J, Lee KF, Gage FH (2003) Mice lacking methyl-CpG binding pro-
Tsujimura K, Abematsu M, Kohyama J, Namihira M, Nakashima K tein 1 have deficits in adult neurogenesis and hippocampal function.
(2009) Neuronal differentiation of neural precursor cells is promoted Proc Natl Acad Sci U S A 100:6777–6782

You might also like