Neurogenesis and Gliogenesis in Health and Disease

A special issue of Brain Sciences (ISSN 2076-3425). This special issue belongs to the section "Molecular and Cellular Neuroscience".

Deadline for manuscript submissions: closed (5 February 2021) | Viewed by 32242

Special Issue Editors


E-Mail Website
Guest Editor
Department of Anesthesiology, University of California San Diego, San Diego, CA, USA
Interests: neural stem cells; addiction; depression; neurodegeneration; neuroimmune disorders

E-Mail Website
Guest Editor
National Centre for Biological Sciences (NCBS), Tata Institute of Fundamental Research (TIFR), GKVK Campus, Bellary Road, Bangalore-560065, India
Interests: adult neurogenesis; radial glia-like cells; microglia; transcriptional regulation; dentate gyrus; hippocampus

Special Issue Information

Dear Colleagues,

The adult mammalian brain has the capacity to generate neural stem cells and neural progenitor cells with neurogenic potential. Over the past few decades, new approaches have revealed the regenerative capacity of the adult brain and have begun to discover their potential role in maintaining a healthy brain and assisting with disease states. This Special Issue will discuss the current knowledge on the capacity of the mammalian brain to generate the stem/progenitor cells and their role in several disease states.

Dr. Chitra Mandyam
Dr. Hiyaa Ghosh
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Brain Sciences is an international peer-reviewed open access monthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2200 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • Neural stem cells
  • Addiction
  • Depression
  • Neurodegeneration
  • Neuroimmune disorders

Published Papers (8 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review, Other

18 pages, 23513 KiB  
Article
Functional Activation of Newborn Neurons Following Alcohol-Induced Reactive Neurogenesis
by Natalie N. Nawarawong, Chelsea G. Nickell, Deann M. Hopkins, James R. Pauly and Kimberly Nixon
Brain Sci. 2021, 11(4), 499; https://doi.org/10.3390/brainsci11040499 - 15 Apr 2021
Cited by 7 | Viewed by 3256
Abstract
Abstinence after alcohol dependence leads to structural and functional recovery in many regions of the brain, especially the hippocampus. Significant increases in neural stem cell (NSC) proliferation and subsequent “reactive neurogenesis” coincides with structural recovery in hippocampal dentate gyrus (DG). However, whether these [...] Read more.
Abstinence after alcohol dependence leads to structural and functional recovery in many regions of the brain, especially the hippocampus. Significant increases in neural stem cell (NSC) proliferation and subsequent “reactive neurogenesis” coincides with structural recovery in hippocampal dentate gyrus (DG). However, whether these reactively born neurons are integrated appropriately into neural circuits remains unknown. Therefore, adult male rats were exposed to a binge model of alcohol dependence. On day 7 of abstinence, the peak of reactive NSC proliferation, rats were injected with bromodeoxyuridine (BrdU) to label dividing cells. After six weeks, rats underwent Morris Water Maze (MWM) training then were sacrificed ninety minutes after the final training session. Using fluorescent immunohistochemistry for c-Fos (neuronal activation), BrdU, and Neuronal Nuclei (NeuN), we investigated whether neurons born during reactive neurogenesis were incorporated into a newly learned MWM neuronal ensemble. Prior alcohol exposure increased the number of BrdU+ cells and newborn neurons (BrdU+/NeuN+ cells) in the DG versus controls. However, prior ethanol exposure had no significant impact on MWM-induced c-Fos expression. Despite increased BrdU+ neurons, no difference in the number of activated newborn neurons (BrdU+/c-Fos+/NeuN+) was observed. These data suggest that neurons born during alcohol-induced reactive neurogenesis are functionally integrated into hippocampal circuitry. Full article
(This article belongs to the Special Issue Neurogenesis and Gliogenesis in Health and Disease)
Show Figures

Graphical abstract

14 pages, 3808 KiB  
Article
Generation of the Human Pluripotent Stem-Cell-Derived Astrocyte Model with Forebrain Identity
by Ulla-Kaisa Peteri, Juho Pitkonen, Kagistia Hana Utami, Jere Paavola, Laurent Roybon, Mahmoud A. Pouladi and Maija L. Castrén
Brain Sci. 2021, 11(2), 209; https://doi.org/10.3390/brainsci11020209 - 09 Feb 2021
Cited by 9 | Viewed by 3620
Abstract
Astrocytes form functionally and morphologically distinct populations of cells with brain-region-specific properties. Human pluripotent stem cells (hPSCs) offer possibilities to generate astroglia for studies investigating mechanisms governing the emergence of astrocytic diversity. We established a method to generate human astrocytes from hPSCs with [...] Read more.
Astrocytes form functionally and morphologically distinct populations of cells with brain-region-specific properties. Human pluripotent stem cells (hPSCs) offer possibilities to generate astroglia for studies investigating mechanisms governing the emergence of astrocytic diversity. We established a method to generate human astrocytes from hPSCs with forebrain patterning and final specification with ciliary neurotrophic factor (CNTF). Transcriptome profiling and gene enrichment analysis monitored the sequential expression of genes determining astrocyte differentiation and confirmed activation of forebrain differentiation pathways at Day 30 (D30) and D60 of differentiation in vitro. More than 90% of astrocytes aged D95 in vitro co-expressed the astrocytic markers glial fibrillary acidic protein (GFAP) and S100β. Intracellular calcium responses to ATP indicated differentiation of the functional astrocyte population with constitutive monocyte chemoattractant protein-1 (MCP-1/CCL2) and tissue inhibitor of metalloproteinases-2 (TIMP-2) expression. The method was reproducible across several hPSC lines, and the data demonstrated the usefulness of forebrain astrocyte modeling in research investigating forebrain pathology. Full article
(This article belongs to the Special Issue Neurogenesis and Gliogenesis in Health and Disease)
Show Figures

Graphical abstract

14 pages, 5087 KiB  
Article
Differential Timing and Coordination of Neurogenesis and Astrogenesis in Developing Mouse Hippocampal Subregions
by Allison M. Bond, Daniel A. Berg, Stephanie Lee, Alan S. Garcia-Epelboim, Vijay S. Adusumilli, Guo-li Ming and Hongjun Song
Brain Sci. 2020, 10(12), 909; https://doi.org/10.3390/brainsci10120909 - 26 Nov 2020
Cited by 24 | Viewed by 3792
Abstract
Neocortical development has been extensively studied and therefore is the basis of our understanding of mammalian brain development. One fundamental principle of neocortical development is that neurogenesis and gliogenesis are temporally segregated processes. However, it is unclear how neurogenesis and gliogenesis are coordinated [...] Read more.
Neocortical development has been extensively studied and therefore is the basis of our understanding of mammalian brain development. One fundamental principle of neocortical development is that neurogenesis and gliogenesis are temporally segregated processes. However, it is unclear how neurogenesis and gliogenesis are coordinated in non-neocortical regions of the cerebral cortex, such as the hippocampus, also known as the archicortex. Here, we show that the timing of neurogenesis and astrogenesis in the Cornu Ammonis (CA) 1 and CA3 regions of mouse hippocampus mirrors that of the neocortex; neurogenesis occurs embryonically, followed by astrogenesis during early postnatal development. In contrast, we find that neurogenesis in the dentate gyrus begins embryonically but is a protracted process which peaks neonatally and continues at low levels postnatally. As a result, astrogenesis, which occurs during early postnatal development, overlaps with the process of neurogenesis in the dentate gyrus. During all stages, neurogenesis overwhelms astrogenesis in the dentate gyrus. In addition, we find that the timing of peak astrogenesis varies by hippocampal subregion. Together, our results show differential timing and coordination of neurogenesis and astrogenesis in developing mouse hippocampal subregions and suggest that neurogenesis and gliogenesis occur simultaneously during dentate gyrus development, challenging the conventional principle that neurogenesis and gliogenesis are temporally separated processes. Full article
(This article belongs to the Special Issue Neurogenesis and Gliogenesis in Health and Disease)
Show Figures

Figure 1

20 pages, 3195 KiB  
Article
Histone Lysine Demethylase JMJD2D/KDM4D and Family Members Mediate Effects of Chronic Social Defeat Stress on Mouse Hippocampal Neurogenesis and Mood Disorders
by Swati Maitra, Nitin Khandelwal, Scherazad Kootar, Pooja Sant, Salil S. Pathak, Sujatha Reddy, Annapoorna P. K., Upadhyayula Suryanarayana Murty, Sumana Chakravarty and Arvind Kumar
Brain Sci. 2020, 10(11), 833; https://doi.org/10.3390/brainsci10110833 - 09 Nov 2020
Cited by 8 | Viewed by 3515
Abstract
Depression, anxiety and related mood disorders are major psychiatric illnesses worldwide, and chronic stress appears to be one of the primary underlying causes. Therapeutics to treat these debilitating disorders without a relapse are limited due to the incomplete molecular understanding of their etiopathology. [...] Read more.
Depression, anxiety and related mood disorders are major psychiatric illnesses worldwide, and chronic stress appears to be one of the primary underlying causes. Therapeutics to treat these debilitating disorders without a relapse are limited due to the incomplete molecular understanding of their etiopathology. In addition to the well-studied genetic component, research in the past two decades has implicated diverse epigenetic mechanisms in mediating the negative effects of chronic stressful events on neural circuits. This includes the cognitive circuitry, where the dynamic hippocampal dentate gyrus (DG) neurogenesis gets affected in depression and related affective disorders. Most of these epigenetic studies have focused on the impact of acetylation/deacetylation and methylation of several histone lysine residues on neural gene expression. However, there is a dearth of investigation into the role of demethylation of these lysine residues in chronic stress-induced changes in neurogenesis that results in altered behaviour. Here, using the chronic social defeat stress (CSDS) paradigm to induce depression and anxiety in C57BL/6 mice and ex vivo DG neural stem/progenitor cell (NSCs/NPCs) culture we show the role of the members of the JMJD2/KDM4 family of histone lysine demethylases (KDMs) in mediating stress-induced changes in DG neurogenesis and mood disorders. The study suggests a critical role of JMJD2D in DG neurogenesis. Altered enrichment of JMJD2D on the promoters of Id2 (inhibitor of differentiation 2) and Sox2 (SRY-Box Transcription Factor 2) was observed during proliferation and differentiation of NSCs/NPCs obtained from the DG. This would affect the demethylation of repressive epigenetic mark H3K9, thus activating or repressing these and possibly other genes involved in regulating proliferation and differentiation of DG NSCs/NPCs. Treatment of the NSCs/NPCs culture with Dimethyloxallyl Glycine (DMOG), an inhibitor of JMJDs, led to attenuation in their proliferation capacity. Additionally, systemic administration of DMOG in mice for 10 days induced depression-like and anxiety-like phenotype without any stress exposure. Full article
(This article belongs to the Special Issue Neurogenesis and Gliogenesis in Health and Disease)
Show Figures

Graphical abstract

Review

Jump to: Research, Other

22 pages, 434 KiB  
Review
Glial PAMPering and DAMPening of Adult Hippocampal Neurogenesis
by Luke Parkitny and Mirjana Maletic-Savatic
Brain Sci. 2021, 11(10), 1299; https://doi.org/10.3390/brainsci11101299 - 29 Sep 2021
Cited by 3 | Viewed by 2354
Abstract
Adult neurogenesis represents a mature brain’s capacity to integrate newly generated neurons into functional circuits. Impairment of neurogenesis contributes to the pathophysiology of various mood and cognitive disorders such as depression and Alzheimer’s Disease. The hippocampal neurogenic niche hosts neural progenitors, glia, and [...] Read more.
Adult neurogenesis represents a mature brain’s capacity to integrate newly generated neurons into functional circuits. Impairment of neurogenesis contributes to the pathophysiology of various mood and cognitive disorders such as depression and Alzheimer’s Disease. The hippocampal neurogenic niche hosts neural progenitors, glia, and vasculature, which all respond to intrinsic and environmental cues, helping determine their current state and ultimate fate. In this article we focus on the major immune communication pathways and mechanisms through which glial cells sense, interact with, and modulate the neurogenic niche. We pay particular attention to those related to the sensing of and response to innate immune danger signals. Receptors for danger signals were first discovered as a critical component of the innate immune system response to pathogens but are now also recognized to play a crucial role in modulating non-pathogenic sterile inflammation. In the neurogenic niche, viable, stressed, apoptotic, and dying cells can activate danger responses in neuroimmune cells, resulting in neuroprotection or neurotoxicity. Through these mechanisms glial cells can influence hippocampal stem cell fate, survival, neuronal maturation, and integration. Depending on the context, such responses may be appropriate and on-target, as in the case of learning-associated synaptic pruning, or excessive and off-target, as in neurodegenerative disorders. Full article
(This article belongs to the Special Issue Neurogenesis and Gliogenesis in Health and Disease)
15 pages, 1111 KiB  
Review
Plasticity in the Hippocampus, Neurogenesis and Drugs of Abuse
by Yosef Avchalumov and Chitra D. Mandyam
Brain Sci. 2021, 11(3), 404; https://doi.org/10.3390/brainsci11030404 - 22 Mar 2021
Cited by 21 | Viewed by 5898
Abstract
Synaptic plasticity in the hippocampus assists with consolidation and storage of long-lasting memories. Decades of research has provided substantial information on the cellular and molecular mechanisms underlying synaptic plasticity in the hippocampus, and this review discusses these mechanisms in brief. Addiction is a [...] Read more.
Synaptic plasticity in the hippocampus assists with consolidation and storage of long-lasting memories. Decades of research has provided substantial information on the cellular and molecular mechanisms underlying synaptic plasticity in the hippocampus, and this review discusses these mechanisms in brief. Addiction is a chronic relapsing disorder with loss of control over drug taking and drug seeking that is caused by long-lasting memories of drug experience. Relapse to drug use is caused by exposure to context and cues associated with the drug experience, and is a major clinical problem that contributes to the persistence of addiction. This review also briefly discusses some evidence that drugs of abuse alter plasticity in the hippocampus, and that development of novel treatment strategies that reverse or prevent drug-induced synaptic alterations in the hippocampus may reduce relapse behaviors associated with addiction. Full article
(This article belongs to the Special Issue Neurogenesis and Gliogenesis in Health and Disease)
Show Figures

Figure 1

Other

Jump to: Research, Review

1 pages, 157 KiB  
Erratum
Erratum: Lee, J.G., et al. The Neuroprotective Effects of Melatonin: Possible Role in the Pathophysiology of Neuropsychiatric Disease. Brain Sciences 2019, 9(10), 285
by Jung Goo Lee, Young Sup Woo, Sung Woo Park, Dae-Hyun Seog, Mi Kyoung Seo and Won-Myong Bahk
Brain Sci. 2019, 9(12), 341; https://doi.org/10.3390/brainsci9120341 - 25 Nov 2019
Cited by 4 | Viewed by 1949
Abstract
The authors wish to make an erratum to the published version of their paper [...] Full article
(This article belongs to the Special Issue Neurogenesis and Gliogenesis in Health and Disease)
12 pages, 1260 KiB  
Perspective
The Neuroprotective Effects of Melatonin: Possible Role in the Pathophysiology of Neuropsychiatric Disease
by Jung Goo Lee, Young Sup Woo, Sung Woo Park, Dae-Hyun Seog, Mi Kyoung Seo and Won-Myong Bahk
Brain Sci. 2019, 9(10), 285; https://doi.org/10.3390/brainsci9100285 - 21 Oct 2019
Cited by 37 | Viewed by 6700
Abstract
Melatonin is a hormone that is secreted by the pineal gland. To date, melatonin is known to regulate the sleep cycle by controlling the circadian rhythm. However, recent advances in neuroscience and molecular biology have led to the discovery of new actions and [...] Read more.
Melatonin is a hormone that is secreted by the pineal gland. To date, melatonin is known to regulate the sleep cycle by controlling the circadian rhythm. However, recent advances in neuroscience and molecular biology have led to the discovery of new actions and effects of melatonin. In recent studies, melatonin was shown to have antioxidant activity and, possibly, to affect the development of Alzheimer’s disease (AD). In addition, melatonin has neuroprotective effects and affects neuroplasticity, thus indicating potential antidepressant properties. In the present review, the new functions of melatonin are summarized and a therapeutic target for the development of new drugs based on the mechanism of action of melatonin is proposed. Full article
(This article belongs to the Special Issue Neurogenesis and Gliogenesis in Health and Disease)
Show Figures

Figure 1

Back to TopTop