Articles

  • KSBNS 2024

Article

Review Article

Exp Neurobiol 2016; 25(3): 103-112

Published online June 30, 2016

https://doi.org/10.5607/en.2016.25.3.103

© The Korean Society for Brain and Neural Sciences

Adult Neurogenesis and Gliogenesis: Possible Mechanisms for Neurorestoration

Zoltán Rusznák1†, Willem Henskens1,2, Emma Schofield1, Woojin S. Kim1,3# and YuHong Fu1,3*#

1Neuroscience Research Australia, Sydney, NSW 2031,2Prince of Wales Clinical School, UNSW Medicine, University of New South Wales,3School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia

Correspondence to: *To whom correspondence should be addressed.
TEL: 61 2 93991056, FAX: 61 2 9399 1005
e-mail: y.fu@neura.edu.au
#Joint senior authors
Present address: Australian Catholic University, Canberra Campus, ACT 2602, Australia

Received: April 27, 2016; Revised: June 8, 2016; Accepted: June 8, 2016

The subgranular zone (SGZ) and subventricular zone (SVZ) are developmental remnants of the germinal regions of the brain, hence they retain the ability to generate neuronal progenitor cells in adult life. Neurogenesis in adult brain has an adaptive function because newly produced neurons can integrate into and modify existing neuronal circuits. In contrast to the SGZ and SVZ, other brain regions have a lower capacity to produce new neurons, and this usually occurs via parenchymal and periventricular cell genesis. Compared to neurogenesis, gliogenesis occurs more prevalently in the adult mammalian brain. Under certain circumstances, interaction occurs between neurogenesis and gliogenesis, facilitating glial cells to transform into neuronal lineage. Therefore, modulating the balance between neurogenesis and gliogenesis may present a new perspective for neurorestoration, especially in diseases associated with altered neurogenesis and/or gliogenesis, cell loss, or disturbed homeostasis of cellular constitution. The present review discusses important neuroanatomical features of adult neurogenesis and gliogenesis, aiming to explore how these processes could be modulated toward functional repair of the adult brain.

Keywords: Neurogenesis, gliogenesis, aging, neurodegeneration, neurorestoration

The adult brain has some ability to adapt to changes in its environment. This ability is, in part, related to neurogenesis and gliogenesis. Neurogenesis modifies neuronal connectivity in specific brain areas, whereas gliogenesis ensures that myelination occurs and produces new supporting cells by generating oligodendrocytes and astrocytes [1]. Altered neurogenesis and gliogenesis have been revealed in a number of pathological conditions affecting the central nervous system (CNS); for instance, neuropsychiatric diseases [2], neurodegenerative diseases [3,4,5,6], and demyelinating diseases [7]. Understanding the extent to which adult neurogenesis and gliogenesis can be modulated to compensate for functional loss has been gaining attention [6,8]. Most of the information in this field was obtained from rodent studies. Although these findings are instrumental in our understanding, they are not directly applicable to the human brain, mainly due to differences in the neurorestoration capacities of different species [9,10,11,12]. The purpose of this review is to bring together available evidence on adult neurogenesis and gliogenesis, and to explore the potential application of new findings to human CNS diseases.

Recent studies detecting the concentration of 14C in the genomic DNA of the human brain demonstrated that the olfactory bulb, cerebellum, and cortex preserve only negligible levels of neurogenesis in adulthood, although substantial neurogenesis was reported in the hippocampus and striatum [13,14,15,16,17]. These results may dispute the hope that human adult neurogenesis can be modulated as a potential approach for neurorestoration. Here, we advise to carefully interrelate these data with the real importance of neurogenic processes for several reasons. Firstly, because the concentration of 14C reflects the average age of cells in a neuronal pool, existing methods may not be sensitive enough to detect subtle changes caused by the introduction of a small proportion of new neurons. Secondly, the selection of brain areas and sample sizes may not have been most appropriate due to the current lack of information about regional variations of the neurogenic capacity in human brains. Thirdly, the biological importance of low levels of neurogenic activity may have been underestimated because even a small number of new neurons in the adult human brain can evoke meaningful functional changes by integrating into existing neuronal circuits [18,19,20]. Finally, gliogenic activity and the presence of quiescent progenitor cells have been detected in numerous brain areas by other studies [1,21,22,23,24]. In particular, glial cells can transform into neuronal lineage under certain circumstances. This review introduces main concepts and important questions in the field, aiming to direct attention to understand the interaction between neurogenesis and gliogenesis, from which factors may be found to improve neurorestoration in the human brain.

Primary neurogenic sites in the brain

The adult subgranular zone (SGZ) and subventricular zone (SVZ) are developmental remnants of the germinal regions originating in the hippocampal sulcus and lateral ganglionic eminence, respectively [28]. In fact, the unique ontogenesis of the SGZ and SVZ allows these two areas to preserve their capacity to generate proliferating cells in the brain during the entire mammalian lifespan. Adult SVZ and SGZ are capable of both neurogenesis and gliogenesis, especially after injury or certain stimulation. In adults, the primary neurogenic sites produce mainly neurons along with some astrocytes and oligodendroglia.

The subgranular zone

The main function of the SGZ is to provide new granule cells for the dentate gyrus [29]. In the human brain, approximately 700 new neurons are generated in the hippocampus per day [13]. These newly generated neurons may have functional significance as demonstrated in rodent studies in which hippocampus-dependent behaviors activated newly generated granule cells several times more often than older granule cells [19,30,31]. Spalding and co-workers (2013) identified three features of human hippocampal neurogenesis that are different from that of rodents — (1) in humans, a much larger proportion of hippocampal neurons are replaced in adulthood; (2) the age-dependent decline in the rate of hippocampal neurogenesis is less pronounced in humans; and (3) hippocampal neurogenesis results in a net increase in the neuronal number of the rodent dentate gyrus, whereas the continuous generation of new neurons in the human hippocampus provides a pool of neurons with specific functional properties [13]. These findings indicate that human hippocampal neurogenesis is important in maintaining hippocampal functions [29,32].

Altered hippocampal neurogenesis may contribute to the severe pathology that occurs in this region in Alzheimer's disease (AD) [3,33,34]. Based on evidence for a link between hippocampal adult neurogenesis and AD, animal studies have revealed that interventions beneficial for hippocampal neurogenesis (e.g. anti-stress medications, physical activity, or administration of allopregnanolone) can improve cognition [35,36,37,38,39]. However, to be effective, these interventions must be introduced at an appropriate time and to an appropriate extent. This requires a better understanding of the variations in hippocampal neurogenesis that occur along with the progression of memory impairment in AD and other cognitive disorders.

The subventricular zone

In the adult rodent brain, neuronal progenitor cells generated in the SVZ travel through the rostral migration stream and settle down in the olfactory bulb, where they differentiate into local interneurons, granule cells, and periglomerular cells [40,41,42,43,44]. Adult neurogenesis in the rodent olfactory bulb is required for specific forms of olfactory behavior, and certain genetic manipulations result in conditional and selective enhancement of olfactory neurogenesis [45,46]. In addition, gliogenesis has been found in the rodent olfactory bulb and the SVZ of multiple sclerosis patients [7,47]. Olfactory neurogenesis is not the same in humans as in rodents. The difference may be partly due to the consequence of the varying importance of olfactory stimuli in ensuring the survival of the species or related to the different glomerular organization in the human and rodent olfactory systems [48].

Under specific circumstances, human olfactory neurogenesis can become noticeable, for example in depression and Parkinson's disease (PD) [49, 50]. Regardless, compared to the substantial amount of new neurons produced in the rodent olfactory bulb, the level of neurogenesis in the healthy adult human olfactory bulb is low [14]. Nevertheless, the adult human SVZ preserves active neurogenesis [11,51,52]. If the SVZ-generated neural progenitor cells do not migrate to the olfactory bulb, the alternative destination of these cells may be the neighboring striatum as suggested by the pronounced striatal neurogenesis that has been detected in the human brain [15]. Interestingly, when olfactory bulb/striatum volume ratios were compared in non-primates, non-human primates, and human, the greater the phylogenetic distance between animals and human the higher this ratio became [11]. This shift in the relative volumes is consistent with the markedly different reliance on these two brain structures. The pronounced neurogenesis in the human striatum may serve the structural refinement that is driven by cognition- and movement-related functional requirements [15].

The fate of NSCs is controlled by complex regulatory machinery, as witnessed by the findings that neurogenic activity of the SVZ can be influenced by GABAergic, dopaminergic, serotoninergic, cholinergic, and nitric oxide-releasing neurons [53,54,55,56]. For this reason, pharmacons affecting neurotransmitter levels (e.g. selective serotonin re-uptake inhibitors and reversible acetylcholinesterase inhibitors) may have therapeutic benefits in diseases where neurogenesis is impaired. On a similar note, there is hope that a better understanding of the anatomical features of striatal neurogenesis may identify factors that can facilitate rehabilitation in diseases that typically affect the striatum (e.g. stroke, Huntington's disease (HD), and PD).

Parenchymal cell genesis

In addition to the primary neurogenic sites, the parenchyma is also capable of producing new cells [9, 59, 61, 62, 63]. In most CNS regions, parenchymal progenitors maintain a slow rate of constitutional gliogenesis, which is important for the renewal of oligodendrocytes and, to a smaller extent, astrocytes [61, 64, 65]. Substantial numbers of oligodendrocyte precursor cells are distributed widely in the adult CNS [66]. They are relatively quiescent but retain regenerative capacity to maintain white matter homeostasis [67]. In a mouse model with damaged white matter, residual oligodendrocyte precursor cells were found to respond to signals released by astrocytes, suggesting that they serve as a 'back-up population' for generating mature oligodendrocytes [68].

Adult astrogenesis has been reported in the mouse cortex and is mainly due to the local division of mature astrocytes [69]. Unlike neurons and oligodendrocytes, astrocytes remain capable of undergoing mitosis even in adulthood — at least in the mouse cortex and spinal cord [70, 71]. In fact, astrocytes have the hallmarks of stem cells because they are capable of perpetuating themselves throughout the entire lifespan of the brain and exhibit multipotency [72, 73]. Astrocytes do not proliferate in the healthy brain parenchyma, but some of them resume proliferation after brain injury [70, 74, 75]. Some astrocytes that re-enter the proliferative cycle acquire the potential to form self-renewing, multipotent neurospheres, which can subsequently produce functional neurons in the mouse cortex [70]. Factors that may trigger astrocyte destabilization/activation include sonic hedgehog (Shh), epidermal growth factor (EGF), and fibroblast growth factor (FGF) [77, 78, 79, 80]. Notably, these growth factors are present in the parenchyma, and their production increases upon brain injury. Most reactive astrocytes, however, remain within their lineage and generate only astrocytes in vivo.

New neurons are also produced in the parenchyma, albeit not many [21]. In fact, in non-human mammals, parenchymal cell genesis has been identified as a potential mechanism for producing neuronal progenitors. The beneficial features of parenchymal progenitors are their abundance and widespread distribution [10, 81, 82]. Although newly produced cells demonstrate a propensity to differentiate, their functional integration may be restricted by the inherent features of the mature parenchyma [10, 81, 82]. The fate of the parenchymal progenitors depends on the environment of the specific brain regions in which they are generated [21] and consequently can be influenced by extrinsic factors. This creates an opportunity for intervention into the process of parenchymal neurogenesis and/or gliogenesis, e.g. by modifying the extrinsic environment in which the cells are located. Potentially, this could be used to counteract the specific pathological process in some CNS diseases.

Human fetal brain parenchymal cells initially express both glial and neuronal markers and are capable of differentiating into either neuronal or glial lineage [24]. However, more work is required to determine if new parenchymal cells can be generated in areas of the human CNS other than the primary neurogenic sites. Research so far suggests that new cortical neurons are not created, because cortical cells in stroke patients were found to be as old as the individual [17]. This suggests that parenchymal neurogenesis may not respond strongly to neuron loss in the human brain, but no further studies on cortical neurogenesis have been undertaken to confirm this.

Periventricular cell genesis

In the forebrain, uncommitted neural precursor cells initially reside in the luminal cell layer (ventricular zone) and later in the SVZ [83]. In addition to the uncommitted precursor cells, some ependymal cells are also able to act as NSCs in the adult brain [83, 84]. For instance, forebrain ventricular CD133-positive ependymal cells show NSCs characteristics in response to brain injury [85, 86]. Further, tanycytes in the adult hypothalamus and ependymal lining of the third ventricle function as multipotential progenitor cells [87, 88]. Finally, there is a consensus that the spinal cord central canal possesses ependymal cells with neural stem cell properties [89, 90].

Whether human adult ventricular ependymal cells have neurogenic potential or not is still unknown. More research is needed to assess the self-regenerative potential of brain regions that are not close to the SGZ and SVZ neurogenic regions but instead close to other parts of the ventricular system. One such region is the substantia nigra, which accommodates dopaminergic neurons and undergoes characteristic pathological changes in PD. Anatomically, the adult substantia nigra is situated in the proximity of the inferior horn of the lateral ventricle, the third ventricle, and the aqueduct; but it is distant from the SVZ (Fig. 1A). There is some evidence for neurogenesis in this region, such as a large number of polysialic acid-positive cells (indicative of immature neurons) are found in the substantia nigra pars reticulata of PD patients [91]. Further, neural progenitor cells could be isolated from the substantia nigra of PD patients [92]. However the source of these NSCs is still unknown. Furthermore, allopregnanolone and platelet-derived growth factor (PDGF)-BB were reported to increase the number of dopaminergic neurons and convey a trophic effect on the surviving dopaminergic neurons [8, 93, 94]. It is unknown if the substantia nigra recruits newly produced neuronal and/or glial progenitor cells from the nearby periventricular zone rather than from the remote forebrain SVZ. If this could be determined, it would open up possibilities as to the methods that could be used to facilitate the survival of new neurons.

Studies using rodent models and cell cultures suggest that the interrelation between adult neurogenesis and gliogenesis can be exploited. Such observations, as described below, suggest that various modulatory processes may be utilized for disease prevention/treatment.

Proliferation and differentiation of NSCs are effectively stimulated by brain lesions [95]. Rodent experiments provide evidence that the stimulatory effect of hypoxia is mediated via the hypoxia-inducible factor (HIF)-1α pathway [96, 97, 98, 99]. The balance between neurogenesis and gliogenesis is affected by numerous factors (Fig. 1B). Bone morphogenetic protein (BMP)-signaling, for example, has been found to oppose the function of oligodendrocyte transcription factor 2 (Olig2) by affecting its expression level [100]. In contrast, FGF and Shh are positive regulators of Olig2 [101]. Finally, infusion of EGF promotes oligodendrogenesis and reduces neurogenesis in the mouse SVZ [102].

In fact, astroglia from the mouse cerebral cortex may be directed toward neurogenesis by 'direct reprogramming'; i.e., by forced expression of paired box 6 (Pax6) [103, 104, 105, 106]. Astrocytes can also directly differentiate into neurons without reactivating progenitor hallmarks, such as cell division [103]. Further, single transcription factor SRY (sex determining region Y)-box 2 (Sox2) is able to convert astrocytes into proliferative neuroblasts in the adult mouse brain [60]. When supplied with brain-derived neurotrophic factor (BDNF) and noggin, or treated with valproic acid (VPA; a histone deacetylase inhibitor), these astrocyte-derived neuroblasts develop into neurons [60]. Finally, platelet-derived growth factor-α receptor (PDGFαR)-expressing and glial fibrillary acidic protein (GFAP)-positive neural stem cells isolated from the mouse SVZ can give rise to both neurons and oligodendrocytes [107] suggesting that PDGFαR signaling may regulate the balance between neurogenesis and oligodendrogenesis. Importantly, astrocytes have been generated from human pluripotent stem cells by exposing them to BMP, ciliary neutrotrophic factor (CNTF), and neuregulin [76].

Altered adult neurogenesis and gliogenesis appear to be associated with some of the neurological and neuropsychiatric diseases, indicating that modulation of the processes involved in adult neurogenesis and gliogenesis may provide a plausible strategy for treatment. However, still much of the specific information is missing, especially in the case of the human brain. It would be important to have more detailed information about (i) cell genesis in non-primary neurogenic sites, (ii) the migration routes of neural precursor cells, (iii) modulatory mechanisms that control the interplay between adult neurogenesis and gliogenesis, (iv) the pathogenic significance of altered neurogenesis and gliogenesis in brain diseases, and (v) effective ways to activate quiescent NSC without compromising the long-term potential of neurogenesis. Research into the relationship between neurogenesis and gliogenesis that is focused on these unknowns is likely to reveal valuable targets for interventions that may improve neurorestoration.

Fig. 1. Neurogenesis and gliogenesis in the adult brain. Potential neurogenic regions described in adult rodent (A1) and human (A2) brains. Solid pink lines indicate the rodent rostral migration stream and the known primary neurogenic regions in rodent and human brains. Pink dots in the rodent olfactory bulb and human striatum indicate the possible final destinations of SVZ-generated new neurons. Pink arrowheads mark some of the putative migration routes of the precursor cells before reaching their final destination (e.g. substantia nigra). Abbreviations: 3V, third ventricle; 4V, fourth ventricle; Aq, aqueduct; DG, dentate gyrus; Hip, hippocampus; LV, lateral ventricle; LVIH, inferior horn of the lateral ventricle; OB, olfactory bulb; RMS, rostral migration stream; SN, substantia nigra. (B) Potential factors reported to modulate the balance between neurogenesis and gliogenesis in rodents and cell cultures. Negative signs mark inhibitory effects. Production of different cell types is region-dependent (as indicated in the lower part of panel B), and this preference may be altered in response to certain diseases or other conditions. For details see the main text.
  1. Bergmann O, Frisén J. Neuroscience. Why adults need new brain cells. Science 2013;340:695-696.
    Pubmed
  2. Braun SM, Jessberger S. Adult neurogenesis: mechanisms and functional significance. Development 2014;141:1983-1986.
    Pubmed
  3. Winner B, Winkler J. Adult neurogenesis in neurodegenerative diseases. Cold Spring Harb Perspect Biol 2015;7:a021287.
    Pubmed
  4. Choi SS, Lee SR, Lee HJ. Neurorestorative role of stem cells in Alzheimer's disease: astrocyte involvement. Curr Alzheimer Res 2016;13:419-427.
    Pubmed
  5. Marxreiter F, Regensburger M, Winkler J. Adult neurogenesis in Parkinson's disease. Cell Mol Life Sci 2013;70:459-473.
    Pubmed
  6. Gil-Mohapel J, Simpson JM, Ghilan M, Christie BR. Neurogenesis in Huntington's disease: can studying adult neurogenesis lead to the development of new therapeutic strategies. Brain Res 2011;1406:84-105.
    Pubmed
  7. Nait-Oumesmar B, Picard-Riéra N, Kerninon C, Baron-Van Evercooren A. The role of SVZ-derived neural precursors in demyelinating diseases: from animal models to multiple sclerosis. J Neurol Sci 2008;265:26-31.
    Pubmed
  8. Foltynie T. Can Parkinson's disease be cured by stimulating neurogenesis?. J Clin Invest 2015;125:978-980.
    Pubmed
  9. Bonfanti L, Peretto P. Adult neurogenesis in mammals--a theme with many variations. Eur J Neurosci 2011;34:930-950.
    Pubmed
  10. Peretto P, Bonfanti L. Adult neurogenesis 20 years later: physiological function vs. brain repair. Front Neurosci 2015;9:71.
    Pubmed
  11. Ernst A, Frisén J. Adult neurogenesis in humans-common and unique traits in mammals. PLoS Biol 2015;13:e1002045.
    Pubmed
  12. Magnusson JP, Frisén J. Stars from the darkest night: unlocking the neurogenic potential of astrocytes in different brain regions. Development 2016;143:1075-1086.
    Pubmed
  13. Spalding KL, Bergmann O, Alkass K, Bernard S, Salehpour M, Huttner HB, Boström E, Westerlund I, Vial C, Buchholz BA, Possnert G, Mash DC, Druid H, Frisén J. Dynamics of hippocampal neurogenesis in adult humans. Cell 2013;153:1219-1227.
    Pubmed
  14. Bergmann O, Liebl J, Bernard S, Alkass K, Yeung MS, Steier P, Kutschera W, Johnson L, Landén M, Druid H, Spalding KL, Frisén J. The age of olfactory bulb neurons in humans. Neuron 2012;74:634-639.
    Pubmed
  15. Ernst A, Alkass K, Bernard S, Salehpour M, Perl S, Tisdale J, Possnert G, Druid H, Frisén J. Neurogenesis in the striatum of the adult human brain. Cell 2014;156:1072-1083.
    Pubmed
  16. Spalding KL, Bhardwaj RD, Buchholz BA, Druid H, Frisén J. Retrospective birth dating of cells in humans. Cell 2005;122:133-143.
    Pubmed
  17. Huttner HB, Bergmann O, Salehpour M, Rácz A, Tatarishvili J, Lindgren E, Csonka T, Csiba L, Hortobágyi T, Méhes G, Englund E, Solnestam BW, Zdunek S, Scharenberg C, Ström L, Ståhl P, Sigurgeirsson B, Dahl A, Schwab S, Possnert G, Bernard S, Kokaia Z, Lindvall O, Lundeberg J, Frisén J. The age and genomic integrity of neurons after cortical stroke in humans. Nat Neurosci 2014;17:801-803.
    Pubmed
  18. Houweling AR, Brecht M. Behavioural report of single neuron stimulation in somatosensory cortex. Nature 2008;451:65-68.
    Pubmed
  19. Snyder JS, Radik R, Wojtowicz JM, Cameron HA. Anatomical gradients of adult neurogenesis and activity: young neurons in the ventral dentate gyrus are activated by water maze training. Hippocampus 2009;19:360-370.
    Pubmed
  20. Kang HJ, Kawasawa YI, Cheng F, Zhu Y, Xu X, Li M, Sousa AM, Pletikos M, Meyer KA, Sedmak G, Guennel T, Shin Y, Johnson MB, Krsnik Z, Mayer S, Fertuzinhos S, Umlauf S, Lisgo SN, Vortmeyer A, Weinberger DR, Mane S, Hyde TM, Huttner A, Reimers M, Kleinman JE, Sestan N. Spatiotemporal transcriptome of the human brain. Nature 2011;478:483-489.
    Pubmed
  21. Bonfanti L. The (real) neurogenic/gliogenic potential of the postnatal and adult brain parenchyma. ISRN Neurosci 2013;2013:354136.
    Pubmed
  22. Belachew S, Chittajallu R, Aguirre AA, Yuan X, Kirby M, Anderson S, Gallo V. Postnatal NG2 proteoglycan-expressing progenitor cells are intrinsically multipotent and generate functional neurons. J Cell Biol 2003;161:169-186.
    Pubmed
  23. Yokoyama A, Yang L, Itoh S, Mori K, Tanaka J. Microglia, a potential source of neurons, astrocytes, and oligodendrocytes. Glia 2004;45:96-104.
    Pubmed
  24. Rieske P, Azizi SA, Augelli B, Gaughan J, Krynska B. A population of human brain parenchymal cells express markers of glial, neuronal and early neural cells and differentiate into cells of neuronal and glial lineages. Eur J Neurosci 2007;25:31-37.
    Pubmed
  25. Huttner WB. Stem cells: slow and steady wins the race. Nat Neurosci 2015;18:613-614.
    Pubmed
  26. Katsimpardi L, Litterman NK, Schein PA, Miller CM, Loffredo FS, Wojtkiewicz GR, Chen JW, Lee RT, Wagers AJ, Rubin LL. Vascular and neurogenic rejuvenation of the aging mouse brain by young systemic factors. Science 2014;344:630-634.
    Pubmed
  27. Yousef H, Conboy MJ, Morgenthaler A, Schlesinger C, Bugaj L, Paliwal P, Greer C, Conboy IM, Schaffer D. Systemic attenuation of the TGF-beta pathway by a single drug simultaneously rejuvenates hippocampal neurogenesis and myogenesis in the same old mammal. Oncotarget 2015;6:11959-11978.
    Pubmed
  28. Curtis MA, Low VF, Faull RL. Neurogenesis and progenitor cells in the adult human brain: a comparison between hippocampal and subventricular progenitor proliferation. Dev Neurobiol 2012;72:990-1005.
    Pubmed
  29. Lepousez G, Nissant A, Lledo PM. Adult neurogenesis and the future of the rejuvenating brain circuits. Neuron 2015;86:387-401.
    Pubmed
  30. Snyder JS, Choe JS, Clifford MA, Jeurling SI, Hurley P, Brown A, Kamhi JF, Cameron HA. Adult-born hippocampal neurons are more numerous, faster maturing, and more involved in behavior in rats than in mice. J Neurosci 2009;29:14484-14495.
    Pubmed
  31. Ramirez-Amaya V, Marrone DF, Gage FH, Worley PF, Barnes CA. Integration of new neurons into functional neural networks. J Neurosci 2006;26:12237-12241.
    Pubmed
  32. Cameron HA, Glover LR. Adult neurogenesis: beyond learning and memory. Annu Rev Psychol 2015;66:53-81.
    Pubmed
  33. Vivar C. Adult hippocampal neurogenesis, aging and neurodegenerative diseases: possible strategies to prevent cognitive impairment. Curr Top Med Chem 2015;15:2175-2192.
    Pubmed
  34. Martinez-Canabal A. Reconsidering hippocampal neurogenesis in Alzheimer's disease. Front Neurosci 2014;8:147.
    Pubmed
  35. Voss MW, Vivar C, Kramer AF, van Praag H. Bridging animal and human models of exercise-induced brain plasticity. Trends Cogn Sci 2013;17:525-544.
    Pubmed
  36. Singh C, Liu L, Wang JM, Irwin RW, Yao J, Chen S, Henry S, Thompson RF, Brinton RD. Allopregnanolone restores hippocampal-dependent learning and memory and neural progenitor survival in aging 3xTgAD and nonTg mice. Neurobiol Aging 2012;33:1493-1506.
    Pubmed
  37. Opendak M, Gould E. Adult neurogenesis: a substrate for experience-dependent change. Trends Cogn Sci 2015;19:151-161.
    Pubmed
  38. Bouchard J, Villeda SA. Aging and brain rejuvenation as systemic events. J Neurochem 2015;132:5-19.
    Pubmed
  39. Irwin RW, Solinsky CM, Loya CM, Salituro FG, Rodgers KE, Bauer G, Rogawski MA, Brinton RD. Allopregnanolone preclinical acute pharmacokinetic and pharmacodynamic studies to predict tolerability and efficacy for Alzheimer's disease. PLoS One 2015;10:e0128313.
    Pubmed
  40. Lazarini F, Gabellec MM, Moigneu C, de Chaumont F, Olivo-Marin JC, Lledo PM. Adult neurogenesis restores dopaminergic neuronal loss in the olfactory bulb. J Neurosci 2014;34:14430-14442.
    Pubmed
  41. Gheusi G, Lepousez G, Lledo PM. Adult-born neurons in the olfactory bulb: integration and functional consequences. Curr Top Behav Neurosci 2013;15:49-72.
    Pubmed
  42. Alvarez-Buylla A, Garcia-Verdugo JM. Neurogenesis in adult subventricular zone. J Neurosci 2002;22:629-634.
    Pubmed
  43. Sakamoto M, Kageyama R, Imayoshi I. The functional significance of newly born neurons integrated into olfactory bulb circuits. Front Neurosci 2014;8:121.
    Pubmed
  44. Merkle FT, Fuentealba LC, Sanders TA, Magno L, Kessaris N, Alvarez-Buylla A. Adult neural stem cells in distinct microdomains generate previously unknown interneuron types. Nat Neurosci 2014;17:207-214.
    Pubmed
  45. Wang W, Lu S, Li T, Pan YW, Zou J, Abel GM, Xu L, Storm DR, Xia Z. Inducible activation of ERK5 MAP kinase enhances adult neurogenesis in the olfactory bulb and improves olfactory function. J Neurosci 2015;35:7833-7849.
    Pubmed
  46. Persson L, Witt RM, Galligan M, Greer PL, Eisner A, Pazyra-Murphy MF, Datta SR, Segal RA. Shh-proteoglycan interactions regulate maturation of olfactory glomerular circuitry. Dev Neurobiol 2014;74:1255-1267.
    Pubmed
  47. Aguirre A, Gallo V. Postnatal neurogenesis and gliogenesis in the olfactory bulb from NG2-expressing progenitors of the subventricular zone. J Neurosci 2004;24:10530-10541.
    Pubmed
  48. Maresh A, Rodriguez Gil D, Whitman MC, Greer CA. Principles of glomerular organization in the human olfactory bulb--implications for odor processing. PLoS One 2008;3:e2640.
    Pubmed
  49. Huisman E, Uylings HB, Hoogland PV. A 100% increase of dopaminergic cells in the olfactory bulb may explain hyposmia in Parkinson's disease. Mov Disord 2004;19:687-692.
    Pubmed
  50. Maheu ME, Devorak J, Freibauer A, Davoli MA, Turecki G, Mechawar N. Increased doublecortin (DCX) expression and incidence of DCX-immunoreactive multipolar cells in the subventricular zone-olfactory bulb system of suicides. Front Neuroanat 2015;9:74.
    Pubmed
  51. Knoth R, Singec I, Ditter M, Pantazis G, Capetian P, Meyer RP, Horvat V, Volk B, Kempermann G. Murine features of neurogenesis in the human hippocampus across the lifespan from 0 to 100 years. PLoS One 2010;5:e8809.
    Pubmed
  52. Sanai N, Nguyen T, Ihrie RA, Mirzadeh Z, Tsai HH, Wong M, Gupta N, Berger MS, Huang E, Garcia-Verdugo JM, Rowitch DH, Alvarez-Buylla A. Corridors of migrating neurons in the human brain and their decline during infancy. Nature 2011;478:382-386.
    Pubmed
  53. Paez-Gonzalez P, Asrican B, Rodriguez E, Kuo CT. Identification of distinct ChAT(+) neurons and activity-dependent control of postnatal SVZ neurogenesis. Nat Neurosci 2014;17:934-942.
    Pubmed
  54. Young SZ, Taylor MM, Bordey A. Neurotransmitters couple brain activity to subventricular zone neurogenesis. Eur J Neurosci 2011;33:1123-1132.
    Pubmed
  55. Tong CK, Chen J, Cebrián-Silla A, Mirzadeh Z, Obernier K, Guinto CD, Tecott LH, García-Verdugo JM, Kriegstein A, Alvarez-Buylla A. Axonal control of the adult neural stem cell niche. Cell Stem Cell 2014;14:500-511.
    Pubmed
  56. Pilz GA, Jessberger S. ChAT me up: how neurons control stem cells. Nat Neurosci 2014;17:897-898.
    Pubmed
  57. Dawson MR, Polito A, Levine JM, Reynolds R. NG2-expressing glial progenitor cells: an abundant and widespread population of cycling cells in the adult rat CNS. Mol Cell Neurosci 2003;24:476-488.
    Pubmed
  58. Aguirre AA, Chittajallu R, Belachew S, Gallo V. NG2-expressing cells in the subventricular zone are type C-like cells and contribute to interneuron generation in the postnatal hippocampus. J Cell Biol 2004;165:575-589.
    Pubmed
  59. Butt AM, Hamilton N, Hubbard P, Pugh M, Ibrahim M. Synantocytes: the fifth element. J Anat 2005;207:695-706.
    Pubmed
  60. Niu W, Zang T, Zou Y, Fang S, Smith DK, Bachoo R, Zhang CL. In vivo reprogramming of astrocytes to neuroblasts in the adult brain. Nat Cell Biol 2013;15:1164-1175.
    Pubmed
  61. Horner PJ, Power AE, Kempermann G, Kuhn HG, Palmer TD, Winkler J, Thal LJ, Gage FH. Proliferation and differentiation of progenitor cells throughout the intact adult rat spinal cord. J Neurosci 2000;20:2218-2228.
    Pubmed
  62. Dayer AG, Cleaver KM, Abouantoun T, Cameron HA. New GABAergic interneurons in the adult neocortex and striatum are generated from different precursors. J Cell Biol 2005;168:415-427.
    Pubmed
  63. Kokoeva MV, Yin H, Flier JS. Neurogenesis in the hypothalamus of adult mice: potential role in energy balance. Science 2005;310:679-683.
    Pubmed
  64. Nishiyama A, Komitova M, Suzuki R, Zhu X. Polydendrocytes (NG2 cells): multifunctional cells with lineage plasticity. Nat Rev Neurosci 2009;10:9-22.
    Pubmed
  65. Boda E, Buffo A. Glial cells in non-germinal territories: insights into their stem/progenitor properties in the intact and injured nervous tissue. Arch Ital Biol 2010;148:119-136.
    Pubmed
  66. Guan T, Kong J. Functional regeneration of the brain: white matter matters. Neural Regen Res 2015;10:355-356.
    Pubmed
  67. Gibson EM, Purger D, Mount CW, Goldstein AK, Lin GL, Wood LS, Inema I, Miller SE, Bieri G, Zuchero JB, Barres BA, Woo PJ, Vogel H, Monje M. Neuronal activity promotes oligodendrogenesis and adaptive myelination in the mammalian brain. Science 2014;344:1252304.
    Pubmed
  68. Miyamoto N, Maki T, Shindo A, Liang AC, Maeda M, Egawa N, Itoh K, Lo EK, Lok J, Ihara M, Arai K. Astrocytes promote oligodendrogenesis after white matter damage via brain-derived neurotrophic factor. J Neurosci 2015;35:14002-14008.
    Pubmed
  69. Ge WP, Miyawaki A, Gage FH, Jan YN, Jan LY. Local generation of glia is a major astrocyte source in postnatal cortex. Nature 2012;484:376-380.
    Pubmed
  70. Buffo A, Rite I, Tripathi P, Lepier A, Colak D, Horn AP, Mori T, Götz M. Origin and progeny of reactive gliosis: a source of multipotent cells in the injured brain. Proc Natl Acad Sci U S A 2008;105:3581-3586.
    Pubmed
  71. Barnabé-Heider F, Göritz C, Sabelström H, Takebayashi H, Pfrieger FW, Meletis K, Frisén J. Origin of new glial cells in intact and injured adult spinal cord. Cell Stem Cell 2010;7:470-482.
    Pubmed
  72. Weissman IL. Stem cells: units of development, units of regeneration, and units in evolution. Cell 2000;100:157-168.
    Pubmed
  73. Adlaf EW, Mitchell-Dick A, Kuo CT. Discerning neurogenic vs. non-neurogenic postnatal lateral ventricular astrocytes via activity-dependent input. Front Neurosci 2016;10:111.
    Pubmed
  74. Buffo A, Vosko MR, Ertürk D, Hamann GF, Jucker M, Rowitch D, Götz M. Expression pattern of the transcription factor Olig2 in response to brain injuries: implications for neuronal repair. Proc Natl Acad Sci U S A 2005;102:18183-18188.
    Pubmed
  75. Robel S, Berninger B, Götz M. The stem cell potential of glia: lessons from reactive gliosis. Nat Rev Neurosci 2011;12:88-104.
    Pubmed
  76. Shaltouki A, Peng J, Liu Q, Rao MS, Zeng X. Efficient generation of astrocytes from human pluripotent stem cells in defined conditions. Stem Cells 2013;31:941-952.
    Pubmed
  77. Smith C, Berry M, Clarke WE, Logan A. Differential expression of fibroblast growth factor-2 and fibroblast growth factor receptor 1 in a scarring and nonscarring model of CNS injury in the rat. Eur J Neurosci 2001;13:443-456.
    Pubmed
  78. Smith GM, Strunz C. Growth factor and cytokine regulation of chondroitin sulfate proteoglycans by astrocytes. Glia 2005;52:209-218.
    Pubmed
  79. Liu B, Chen H, Johns TG, Neufeld AH. Epidermal growth factor receptor activation: an upstream signal for transition of quiescent astrocytes into reactive astrocytes after neural injury. J Neurosci 2006;26:7532-7540.
    Pubmed
  80. Amankulor NM, Hambardzumyan D, Pyonteck SM, Becher OJ, Joyce JA, Holland EC. Sonic hedgehog pathway activation is induced by acute brain injury and regulated by injury-related inflammation. J Neurosci 2009;29:10299-10308.
    Pubmed
  81. Bonfanti L. From hydra regeneration to human brain structural plasticity: a long trip through narrowing roads. ScientificWorldJournal 2011;11:1270-1299.
    Pubmed
  82. Bonfanti L, Mackay-Sim A. Exploring neurogenesis outside the niche: atypical locations of mammalian neural stem/progenitor cells. Arch Ital Biol 2010;148:43-45.
    Pubmed
  83. Gritti A, Vescovi AL, Galli R. Adult neural stem cells: plasticity and developmental potential. J Physiol Paris 2002;96:81-90.
    Pubmed
  84. Bauer HC, Tempfer H, Bernroider G, Bauer H. Neuronal stem cells in adults. Exp Gerontol 2006;41:111-116.
    Pubmed
  85. Coskun V, Wu H, Blanchi B, Tsao S, Kim K, Zhao J, Biancotti JC, Hutnick L, Krueger RC, Fan G, de Vellis J, Sun YE. CD133+ neural stem cells in the ependyma of mammalian postnatal forebrain. Proc Natl Acad Sci U S A 2008;105:1026-1031.
    Pubmed
  86. Zhang RL, Zhang ZG, Wang Y, LeTourneau Y, Liu XS, Zhang X, Gregg SR, Wang L, Chopp M. Stroke induces ependymal cell transformation into radial glia in the subventricular zone of the adult rodent brain. J Cereb Blood Flow Metab 2007;27:1201-1212.
    Pubmed
  87. Niwa A, Nishibori M, Hamasaki S, Kobori T, Liu K, Wake H, Mori S, Yoshino T, Takahashi H. Voluntary exercise induces neurogenesis in the hypothalamus and ependymal lining of the third ventricle. Brain Struct Funct 2016;221:1653-1666.
    Pubmed
  88. Maggi R, Zasso J, Conti L. Neurodevelopmental origin and adult neurogenesis of the neuroendocrine hypothalamus. Front Cell Neurosci 2015;8:440.
    Pubmed
  89. Qin Y, Zhang W, Yang P. Current states of endogenous stem cells in adult spinal cord. J Neurosci Res 2015;93:391-398.
    Pubmed
  90. Hugnot JP, Franzen R. The spinal cord ependymal region: a stem cell niche in the caudal central nervous system. Front Biosci (Landmark Ed) 2011;16:1044-1059.
    Pubmed
  91. Yoshimi K, Ren YR, Seki T, Yamada M, Ooizumi H, Onodera M, Saito Y, Murayama S, Okano H, Mizuno Y, Mochizuki H. Possibility for neurogenesis in substantia nigra of parkinsonian brain. Ann Neurol 2005;58:31-40.
    Pubmed
  92. Wang S, Okun MS, Suslov O, Zheng T, McFarland NR, Vedam-Mai V, Foote KD, Roper SN, Yachnis AT, Siebzehnrubl FA, Steindler DA. Neurogenic potential of progenitor cells isolated from postmortem human Parkinsonian brains. Brain Res 2012;1464:61-72.
    Pubmed
  93. Wang JM. Allopregnanolone and neurogenesis in the nigrostriatal tract. Front Cell Neurosci 2014;8:224.
    Pubmed
  94. Paul G, Zachrisson O, Varrone A, Almqvist P, Jerling M, Lind G, Rehncrona S, Linderoth B, Bjartmarz H, Shafer LL, Coffey R, Svensson M, Mercer KJ, Forsberg A, Halldin C, Svenningsson P, Widner H, Frisén J, Pålhagen S, Haegerstrand A. Safety and tolerability of intracerebroventricular PDGF-BB in Parkinson's disease patients. J Clin Invest 2015;125:1339-1346.
    Pubmed
  95. Lin R, Iacovitti L. Classic and novel stem cell niches in brain homeostasis and repair. Brain Res 2015;1628:327-342.
    Pubmed
  96. Li L, Candelario KM, Thomas K, Wang R, Wright K, Messier A, Cunningham LA. Hypoxia inducible factor-1alpha (HIF-1alpha) is required for neural stem cell maintenance and vascular stability in the adult mouse SVZ. J Neurosci 2014;34:16713-16719.
    Pubmed
  97. Cunningham LA, Candelario K, Li L. Roles for HIF-1alpha in neural stem cell function and the regenerative response to stroke. Behav Brain Res 2012;227:410-417.
    Pubmed
  98. Hu Q, Liang X, Chen D, Chen Y, Doycheva D, Tang J, Tang J, Zhang JH. Delayed hyperbaric oxygen therapy promotes neurogenesis through reactive oxygen species/hypoxia-inducible factor-1alpha/beta-catenin pathway in middle cerebral artery occlusion rats. Stroke 2014;45:1807-1814.
    Pubmed
  99. Vieira HL, Alves PM, Vercelli A. Modulation of neuronal stem cell differentiation by hypoxia and reactive oxygen species. Prog Neurobiol 2011;93:444-455.
    Pubmed
  100. Colak D, Mori T, Brill MS, Pfeifer A, Falk S, Deng C, Monteiro R, Mummery C, Sommer L, Götz M. Adult neurogenesis requires Smad4-mediated bone morphogenic protein signaling in stem cells. J Neurosci 2008;28:434-446.
    Pubmed
  101. Hack MA, Sugimori M, Lundberg C, Nakafuku M, Götz M. Regionalization and fate specification in neurospheres: the role of Olig2 and Pax6. Mol Cell Neurosci 2004;25:664-678.
    Pubmed
  102. 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.
    Pubmed
  103. Heins N, Malatesta P, Cecconi F, Nakafuku M, Tucker KL, Hack MA, Chapouton P, Barde YA, Götz M. Glial cells generate neurons: the role of the transcription factor Pax6. Nat Neurosci 2002;5:308-315.
    Pubmed
  104. Berninger B, Costa MR, Koch U, Schroeder T, Sutor B, Grothe B, Götz M. Functional properties of neurons derived from in vitro reprogrammed postnatal astroglia. J Neurosci 2007;27:8654-8664.
    Pubmed
  105. Blum R, Heinrich C, Sánchez R, Lepier A, Gundelfinger ED, Berninger B, Götz M. Neuronal network formation from reprogrammed early postnatal rat cortical glial cells. Cereb Cortex 2011;21:413-424.
    Pubmed
  106. Heinrich C, Blum R, Gascón S, Masserdotti G, Tripathi P, Sánchez R, Tiedt S, Schroeder T, Götz M, Berninger B. Directing astroglia from the cerebral cortex into subtype specific functional neurons. PLoS Biol 2010;8:e1000373.
    Pubmed
  107. Jackson EL, Garcia-Verdugo JM, Gil-Perotin S, Roy M, Quinones-Hinojosa A, VandenBerg S, Alvarez-Buylla A. PDGFR alpha-positive B cells are neural stem cells in the adult SVZ that form glioma-like growths in response to increased PDGF signaling. Neuron 2006;51:187-199.
    Pubmed