1
|
Cebrian-Silla A, Nascimento MA, Mancia W, Gonzalez-Granero S, Romero-Rodriguez R, Obernier K, Steffen DM, Lim DA, Garcia-Verdugo JM, Alvarez-Buylla A. Neural stem cell relay from B1 to B2 cells in the adult mouse ventricular-subventricular zone. Cell Rep 2025; 44:115264. [PMID: 40019835 PMCID: PMC11979704 DOI: 10.1016/j.celrep.2025.115264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 11/27/2024] [Accepted: 01/13/2025] [Indexed: 03/05/2025] Open
Abstract
Neurogenesis and gliogenesis continue in the ventricular-subventricular zone (V-SVZ) of the adult rodent brain. V-SVZ astroglial cells with apical contact with the ventricle (B1 cells) function as neural stem cells (NSCs). B1 cells sharply decline during early postnatal life; in contrast, neurogenesis decreases at a slower rate. Here, we show that a second population of astroglia (B2 cells) that do not contact the ventricle also function as NSCs in the adult mouse brain. B2 cell numbers increase postnatally, are sustained in adults, and decrease with aging. We reveal the transcriptomic profile of B1 and B2 cells and show that, like B1 cells, B2 cells can be quiescent or activated. Transplantation and lineage tracing of B2 cells demonstrate their function as primary progenitors for adult neurogenesis. This study reveals that NSC function is progressively relayed from B1 to B2 progenitors helping explain how neurogenesis is maintained into adult life.
Collapse
Affiliation(s)
- Arantxa Cebrian-Silla
- Eli and Edythe Broad Institute for Stem Cell Research and Regeneration Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA.
| | - Marcos Assis Nascimento
- Eli and Edythe Broad Institute for Stem Cell Research and Regeneration Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Walter Mancia
- Eli and Edythe Broad Institute for Stem Cell Research and Regeneration Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Susana Gonzalez-Granero
- BTELab, Research Foundation of the General University Hospital of Valencia, Valencia 46014, Spain
| | - Ricardo Romero-Rodriguez
- Eli and Edythe Broad Institute for Stem Cell Research and Regeneration Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kirsten Obernier
- Quantitative Biosciences Institute, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - David M Steffen
- Eli and Edythe Broad Institute for Stem Cell Research and Regeneration Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Daniel A Lim
- Eli and Edythe Broad Institute for Stem Cell Research and Regeneration Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jose Manuel Garcia-Verdugo
- Laboratory of Comparative Neurobiology, Cavanilles Institute of Biodiversity and Evolutionary Biology, University of Valencia and CIBERNED-ISCIII, Paterna, 46980 Valencia, Spain
| | - Arturo Alvarez-Buylla
- Eli and Edythe Broad Institute for Stem Cell Research and Regeneration Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
2
|
Groh AMR, Song YL, Tea F, Lu B, Huynh S, Afanasiev E, Bigotte M, Del Bigio MR, Stratton JA. Multiciliated ependymal cells: an update on biology and pathology in the adult brain. Acta Neuropathol 2024; 148:39. [PMID: 39254862 DOI: 10.1007/s00401-024-02784-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 09/11/2024]
Abstract
Mature multiciliated ependymal cells line the cerebral ventricles where they form a partial barrier between the cerebrospinal fluid (CSF) and brain parenchyma and regulate local CSF microcirculation through coordinated ciliary beating. Although the ependyma is a highly specialized brain interface with barrier, trophic, and perhaps even regenerative capacity, it remains a misfit in the canon of glial neurobiology. We provide an update to seminal reviews in the field by conducting a scoping review of the post-2010 mature multiciliated ependymal cell literature. We delineate how recent findings have either called into question or substantiated classical views of the ependymal cell. Beyond this synthesis, we document the basic methodologies and study characteristics used to describe multiciliated ependymal cells since 1980. Our review serves as a comprehensive resource for future investigations of mature multiciliated ependymal cells.
Collapse
Affiliation(s)
- Adam M R Groh
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montréal, QC, Canada
| | - Yeji Lori Song
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montréal, QC, Canada
| | - Fiona Tea
- Department of Neuroscience, University of Montreal, Montréal, QC, Canada
| | - Brianna Lu
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montréal, QC, Canada
| | - Stephanie Huynh
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montréal, QC, Canada
| | - Elia Afanasiev
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montréal, QC, Canada
| | - Maxime Bigotte
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montréal, QC, Canada
| | - Marc R Del Bigio
- Department of Pathology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Jo Anne Stratton
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montréal, QC, Canada.
| |
Collapse
|
3
|
Cebrian-Silla A, Assis Nascimento M, Mancia W, Gonzalez-Granero S, Romero-Rodriguez R, Obernier K, Steffen DM, Lim DA, Garcia-Verdugo JM, Alvarez-Buylla A. Neural Stem Cell Relay from B1 to B2 cells in the adult mouse Ventricular-Subventricular Zone. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.28.600695. [PMID: 39005355 PMCID: PMC11244865 DOI: 10.1101/2024.06.28.600695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Neurogenesis and gliogenesis continue in the Ventricular-Subventricular Zone (V-SVZ) of the adult rodent brain. B1 cells are astroglial cells derived from radial glia that function as primary progenitors or neural stem cells (NSCs) in the V-SVZ. B1 cells, which have a small apical contact with the ventricle, decline in numbers during early postnatal life, yet neurogenesis continues into adulthood. Here we found that a second population of V-SVZ astroglial cells (B2 cells), that do not contact the ventricle, function as NSCs in the adult brain. B2 cell numbers increase postnatally, remain constant in 12-month-old mice and decrease by 18 months. Transcriptomic analysis of ventricular-contacting and non-contacting B cells revealed key molecular differences to distinguish B1 from B2 cells. Transplantation and lineage tracing of B2 cells demonstrate their function as primary progenitors for adult neurogenesis. This study reveals how NSC function is relayed from B1 to B2 progenitors to maintain adult neurogenesis.
Collapse
|
4
|
North HF, Weissleder C, Bitar M, Barry G, Fullerton JM, Webster MJ, Weickert CS. RNA-sequencing suggests extracellular matrix and vasculature dysregulation could impair neurogenesis in schizophrenia cases with elevated inflammation. SCHIZOPHRENIA (HEIDELBERG, GERMANY) 2024; 10:50. [PMID: 38704390 PMCID: PMC11069512 DOI: 10.1038/s41537-024-00466-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 03/25/2024] [Indexed: 05/06/2024]
Abstract
A subgroup of schizophrenia cases with elevated inflammation have reduced neurogenesis markers and increased macrophage density in the human subependymal zone (SEZ; also termed subventricular zone or SVZ) neurogenic niche. Inflammation can impair neurogenesis; however, it is unclear which other pathways are associated with reduced neurogenesis. This research aimed to discover transcriptomic differences between inflammatory subgroups of schizophrenia in the SEZ. Total RNA sequencing was performed on SEZ tissue from schizophrenia cases, designated into low inflammation (n = 13) and high inflammation (n = 14) subgroups, based on cluster analysis of inflammation marker gene expression. 718 genes were differentially expressed in high compared to low inflammation schizophrenia (FDR p < 0.05) and were most significantly over-represented in the pathway 'Hepatic Fibrosis/Hepatic Stellate-Cell Activation'. Genes in this pathway relate to extracellular matrix stability (including ten collagens) and vascular remodelling suggesting increased angiogenesis. Collagen-IV, a key element of the basement membrane and fractones, had elevated gene expression. Immunohistochemistry revealed novel collagen-IV+ fractone bulbs within the human SEZ hypocellular gap. Considering the extracellular matrix's regulatory role in SEZ neurogenesis, fibrosis-related alterations in high inflammation schizophrenia may disrupt neurogenesis. Increased angiogenesis could facilitate immune cell transmigration, potentially explaining elevated macrophages in high inflammation schizophrenia. This discovery-driven analysis sheds light on how inflammation may contribute to schizophrenia neuropathology in the neurogenic niche.
Collapse
Affiliation(s)
- Hayley F North
- Neuroscience Research Australia, Sydney, NSW, Australia
- Discipline of Psychiatry and Mental Health, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Christin Weissleder
- Neuroscience Research Australia, Sydney, NSW, Australia
- Mechanism and therapy for genetic brain diseases, Institut Imagine, Paris, France
| | | | - Guy Barry
- OncoLife Therapeutics, Yeronga, QLD, Australia
| | - Janice M Fullerton
- Neuroscience Research Australia, Sydney, NSW, Australia
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Maree J Webster
- Laboratory of Brain Research, Stanley Medical Research Institute, 9800, Medical Center Drive, Rockville, MD, USA
| | - Cynthia Shannon Weickert
- Neuroscience Research Australia, Sydney, NSW, Australia.
- Discipline of Psychiatry and Mental Health, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia.
- Department of Neuroscience and Physiology, Upstate Medical University, Syracuse, NY, USA.
| |
Collapse
|
5
|
Rastoldo G, Tighilet B. The Vestibular Nuclei: A Cerebral Reservoir of Stem Cells Involved in Balance Function in Normal and Pathological Conditions. Int J Mol Sci 2024; 25:1422. [PMID: 38338702 PMCID: PMC10855768 DOI: 10.3390/ijms25031422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 01/18/2024] [Accepted: 01/20/2024] [Indexed: 02/12/2024] Open
Abstract
In this review, we explore the intriguing realm of neurogenesis in the vestibular nuclei-a critical brainstem region governing balance and spatial orientation. We retrace almost 20 years of research into vestibular neurogenesis, from its discovery in the feline model in 2007 to the recent discovery of a vestibular neural stem cell niche. We explore the reasons why neurogenesis is important in the vestibular nuclei and the triggers for activating the vestibular neurogenic niche. We develop the symbiotic relationship between neurogenesis and gliogenesis to promote vestibular compensation. Finally, we examine the potential impact of reactive neurogenesis on vestibular compensation, highlighting its role in restoring balance through various mechanisms.
Collapse
Affiliation(s)
- Guillaume Rastoldo
- Aix Marseille Université-CNRS, Laboratoire de Neurosciences Cognitives, LNC UMR 7291, 13331 Marseille, France;
| | - Brahim Tighilet
- Aix Marseille Université-CNRS, Laboratoire de Neurosciences Cognitives, LNC UMR 7291, 13331 Marseille, France;
- GDR Vertige CNRS Unité GDR2074, 13331 Marseille, France
| |
Collapse
|
6
|
Salmina AB, Alexandrova OP, Averchuk AS, Korsakova SA, Saridis MR, Illarioshkin SN, Yurchenko SO. Current progress and challenges in the development of brain tissue models: How to grow up the changeable brain in vitro? J Tissue Eng 2024; 15:20417314241235527. [PMID: 38516227 PMCID: PMC10956167 DOI: 10.1177/20417314241235527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 02/12/2024] [Indexed: 03/23/2024] Open
Abstract
In vitro modeling of brain tissue is a promising but not yet resolved problem in modern neurobiology and neuropharmacology. Complexity of the brain structure and diversity of cell-to-cell communication in (patho)physiological conditions make this task almost unachievable. However, establishment of novel in vitro brain models would ultimately lead to better understanding of development-associated or experience-driven brain plasticity, designing efficient approaches to restore aberrant brain functioning. The main goal of this review is to summarize the available data on methodological approaches that are currently in use, and to identify the most prospective trends in development of neurovascular unit, blood-brain barrier, blood-cerebrospinal fluid barrier, and neurogenic niche in vitro models. The manuscript focuses on the regulation of adult neurogenesis, cerebral microcirculation and fluids dynamics that should be reproduced in the in vitro 4D models to mimic brain development and its alterations in brain pathology. We discuss approaches that are critical for studying brain plasticity, deciphering the individual person-specific trajectory of brain development and aging, and testing new drug candidates in the in vitro models.
Collapse
Affiliation(s)
- Alla B Salmina
- Brain Science Institute, Research Center of Neurology, Moscow, Russia
- Bauman Moscow State Technical University, Moscow, Russia
| | - Olga P Alexandrova
- Brain Science Institute, Research Center of Neurology, Moscow, Russia
- Bauman Moscow State Technical University, Moscow, Russia
| | - Anton S Averchuk
- Brain Science Institute, Research Center of Neurology, Moscow, Russia
- Bauman Moscow State Technical University, Moscow, Russia
| | | | | | | | | |
Collapse
|
7
|
Valamparamban GF, Spéder P. Homemade: building the structure of the neurogenic niche. Front Cell Dev Biol 2023; 11:1275963. [PMID: 38107074 PMCID: PMC10722289 DOI: 10.3389/fcell.2023.1275963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 11/16/2023] [Indexed: 12/19/2023] Open
Abstract
Neural stem/progenitor cells live in an intricate cellular environment, the neurogenic niche, which supports their function and enables neurogenesis. The niche is made of a diversity of cell types, including neurons, glia and the vasculature, which are able to signal to and are structurally organised around neural stem/progenitor cells. While the focus has been on how individual cell types signal to and influence the behaviour of neural stem/progenitor cells, very little is actually known on how the niche is assembled during development from multiple cellular origins, and on the role of the resulting topology on these cells. This review proposes to draw a state-of-the art picture of this emerging field of research, with the aim to expose our knowledge on niche architecture and formation from different animal models (mouse, zebrafish and fruit fly). We will span its multiple aspects, from the existence and importance of local, adhesive interactions to the potential emergence of larger-scale topological properties through the careful assembly of diverse cellular and acellular components.
Collapse
Affiliation(s)
| | - Pauline Spéder
- Institut Pasteur, Université Paris Cité, CNRS UMR3738, Structure and Signals in the Neurogenic Niche, Paris, France
| |
Collapse
|
8
|
Blasco-Chamarro L, Fariñas I. Fine-tuned rest: unveiling the regulatory landscape of adult quiescent neural stem cells. Neuroscience 2023:S0306-4522(23)00298-1. [PMID: 37437796 DOI: 10.1016/j.neuroscience.2023.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 06/29/2023] [Accepted: 07/03/2023] [Indexed: 07/14/2023]
Abstract
Cell quiescence is an essential mechanism that allows cells to temporarily halt proliferation while preserving the potential to resume it at a later time. The molecular mechanisms underlying cell quiescence are complex and involve the regulation of various signaling pathways, transcription factors and epigenetic modifications. The importance of unveiling the mechanisms regulating the quiescent state is undeniable, as its long-term maintenance is key to sustain tissue homeostasis throughout life. Neural stem cells (NSCs) are maintained in the subependymal zone (SEZ) niche of adult mammalian brains mostly as long-lasting quiescent cells, owing to multiple intrinsic and extrinsic cues that actively regulate this state. Differently from other non-proliferative states, quiescence is a reversible and tightly regulated condition that can re-activate to support the formation of new neurons throughout adult lifespan. Decoding its regulatory mechanisms in homeostasis and unveiling how it is modulated in the context of the aged brain or during tumorigenesis, could bring us closer to the development of new potential strategies to intervene in adult neurogenesis with therapeutic purposes. Starting with a general conceptualization of the quiescent state in different stem cell niches, we here review what we have learned about NSC quiescence in the SEZ, encompassing the experimental strategies used for its study, to end up discussing the modulation of quiescence in the context of a physiology or pathological NSC dysregulation.
Collapse
Affiliation(s)
- Laura Blasco-Chamarro
- Biomedical Research Network on Neurodegenerative Diseases (CIBERNED); Department of Cell Biology; Biotechnology and Biomedicine Institute (BioTecMed), University of Valencia, Spain
| | - Isabel Fariñas
- Biomedical Research Network on Neurodegenerative Diseases (CIBERNED); Department of Cell Biology; Biotechnology and Biomedicine Institute (BioTecMed), University of Valencia, Spain.
| |
Collapse
|
9
|
Yeo RW, Zhou OY, Zhong BL, Sun ED, Navarro Negredo P, Nair S, Sharmin M, Ruetz TJ, Wilson M, Kundaje A, Dunn AR, Brunet A. Chromatin accessibility dynamics of neurogenic niche cells reveal defects in neural stem cell adhesion and migration during aging. NATURE AGING 2023; 3:866-893. [PMID: 37443352 PMCID: PMC10353944 DOI: 10.1038/s43587-023-00449-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 06/02/2023] [Indexed: 07/15/2023]
Abstract
The regenerative potential of brain stem cell niches deteriorates during aging. Yet the mechanisms underlying this decline are largely unknown. Here we characterize genome-wide chromatin accessibility of neurogenic niche cells in vivo during aging. Interestingly, chromatin accessibility at adhesion and migration genes decreases with age in quiescent neural stem cells (NSCs) but increases with age in activated (proliferative) NSCs. Quiescent and activated NSCs exhibit opposing adhesion behaviors during aging: quiescent NSCs become less adhesive, whereas activated NSCs become more adhesive. Old activated NSCs also show decreased migration in vitro and diminished mobilization out of the niche for neurogenesis in vivo. Using tension sensors, we find that aging increases force-producing adhesions in activated NSCs. Inhibiting the cytoskeletal-regulating kinase ROCK reduces these adhesions, restores migration in old activated NSCs in vitro, and boosts neurogenesis in vivo. These results have implications for restoring the migratory potential of NSCs and for improving neurogenesis in the aged brain.
Collapse
Affiliation(s)
- Robin W Yeo
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Olivia Y Zhou
- Department of Genetics, Stanford University, Stanford, CA, USA
- Stanford Biophysics Program, Stanford University, Stanford, CA, USA
- Stanford Medical Scientist Training Program, Stanford University, Stanford, CA, USA
| | - Brian L Zhong
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Eric D Sun
- Department of Genetics, Stanford University, Stanford, CA, USA
- Biomedical Informatics Graduate Program, Stanford University, Stanford, CA, USA
| | | | - Surag Nair
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Mahfuza Sharmin
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Tyson J Ruetz
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Mikaela Wilson
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Anshul Kundaje
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Alexander R Dunn
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Anne Brunet
- Department of Genetics, Stanford University, Stanford, CA, USA.
- Glenn Laboratories for the Biology of Aging, Stanford University, Stanford, CA, USA.
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
10
|
Paez-Gonzalez P, Lopez-de-San-Sebastian J, Ceron-Funez R, Jimenez AJ, Rodríguez-Perez LM. Therapeutic strategies to recover ependymal barrier after inflammatory damage: relevance for recovering neurogenesis during development. Front Neurosci 2023; 17:1204197. [PMID: 37397456 PMCID: PMC10308384 DOI: 10.3389/fnins.2023.1204197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 05/22/2023] [Indexed: 07/04/2023] Open
Abstract
The epithelium covering the surfaces of the cerebral ventricular system is known as the ependyma, and is essential for maintaining the physical and functional integrity of the central nervous system. Additionally, the ependyma plays an essential role in neurogenesis, neuroinflammatory modulation and neurodegenerative diseases. Ependyma barrier is severely affected by perinatal hemorrhages and infections that cross the blood brain barrier. The recovery and regeneration of ependyma after damage are key to stabilizing neuroinflammatory and neurodegenerative processes that are critical during early postnatal ages. Unfortunately, there are no effective therapies to regenerate this tissue in human patients. Here, the roles of the ependymal barrier in the context of neurogenesis and homeostasis are reviewed, and future research lines for development of actual therapeutic strategies are discussed.
Collapse
Affiliation(s)
- Patricia Paez-Gonzalez
- Department of Cell Biology, Genetics and Physiology, University of Malaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Málaga, Spain
| | | | - Raquel Ceron-Funez
- Department of Cell Biology, Genetics and Physiology, University of Malaga, Málaga, Spain
| | - Antonio J. Jimenez
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Málaga, Spain
| | - Luis Manuel Rodríguez-Perez
- Department of Cell Biology, Genetics and Physiology, University of Malaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Málaga, Spain
- Department of Human Physiology, Human Histology, Pathological Anatomy and Sports, University of Malaga, Málaga, Spain
| |
Collapse
|
11
|
Yazdani N, Willits RK. Mimicking the neural stem cell niche: An engineer’s view of cell: material interactions. FRONTIERS IN CHEMICAL ENGINEERING 2023. [DOI: 10.3389/fceng.2022.1086099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Neural stem cells have attracted attention in recent years to treat neurodegeneration. There are two neurogenic regions in the brain where neural stem cells reside, one of which is called the subventricular zone (SVZ). The SVZ niche is a complicated microenvironment providing cues to regulate self-renewal and differentiation while maintaining the neural stem cell’s pool. Many scientists have spent years understanding the cellular and structural characteristics of the SVZ niche, both in homeostasis and pathological conditions. On the other hand, engineers focus primarily on designing platforms using the knowledge they acquire to understand the effect of individual factors on neural stem cell fate decisions. This review provides a general overview of what we know about the components of the SVZ niche, including the residing cells, extracellular matrix (ECM), growth factors, their interactions, and SVZ niche changes during aging and neurodegenerative diseases. Furthermore, an overview will be given on the biomaterials used to mimic neurogenic niche microenvironments and the design considerations applied to add bioactivity while meeting the structural requirements. Finally, it will discuss the potential gaps in mimicking the microenvironment.
Collapse
|
12
|
Mesquita FCP, Leite ES, Morrissey J, Freitas C, Coelho-Sampaio T, Hochman-Mendez C. Polymerized Laminin-521: A Feasible Substrate for Expanding Induced Pluripotent Stem Cells at a Low Protein Concentration. Cells 2022; 11:cells11243955. [PMID: 36552719 PMCID: PMC9777247 DOI: 10.3390/cells11243955] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/29/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
Laminins (LNs) play a central role in the self-assembly and maintenance of basement membranes and are involved in critical interactions between cells and other extracellular matrix (ECM) proteins. Among the defined, xeno-free ECM culture matrices, LNs-namely LN521-have emerged as promising coating systems for the large-scale expansion of induced pluripotent stem cells (iPSCs). The biologic activity of LNs is enhanced by their acidification-induced self-polymerization into a cell-associated network called polylaminin (polyLN), which can recapitulate the native-like polymeric array in a cell-free system. Here, we show for the first time to our knowledge that polyLN521 displays a native-like hexagonal-like structure and that, at basal and low concentrations, it permits the large-scale expansion of human iPSCs. Human iPSCs expanded with polyLN521 maintained the pluripotent state and showed no impairment of karyotype stability or telomere length. These results suggest that low-concentration polyLN521 is a stable and cost-effective coating for large-scale iPSC expansion.
Collapse
Affiliation(s)
- Fernanda C. P. Mesquita
- Department of Regenerative Medicine Research, The Texas Heart Institute, 6770 Bertner Avenue, MC 1-135, Houston, TX 77030, USA
| | - Eliel S. Leite
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho B1-011, 373, Rio de Janeiro 21941-902, Brazil
| | - Jacquelynn Morrissey
- Department of Regenerative Medicine Research, The Texas Heart Institute, 6770 Bertner Avenue, MC 1-135, Houston, TX 77030, USA
| | - Catarina Freitas
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho B1-011, 373, Rio de Janeiro 21941-902, Brazil
| | - Tatiana Coelho-Sampaio
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho B1-011, 373, Rio de Janeiro 21941-902, Brazil
| | - Camila Hochman-Mendez
- Department of Regenerative Medicine Research, The Texas Heart Institute, 6770 Bertner Avenue, MC 1-135, Houston, TX 77030, USA
- Correspondence:
| |
Collapse
|
13
|
Rastoldo G, Watabe I, Lapotre A, Tonetto A, López-Juárez A, Tighilet B. Vestibular Nuclei: A New Neural Stem Cell Niche? Cells 2022; 11:cells11223598. [PMID: 36429025 PMCID: PMC9688605 DOI: 10.3390/cells11223598] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/06/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022] Open
Abstract
We previously reported adult reactive neurogliogenesis in the deafferented vestibular nuclei following unilateral vestibular neurectomy (UVN) in the feline and the rodent model. Recently, we demonstrated that UVN induced a significant increase in a population of cells colocalizing the transcription factor sex determining region Y-box 2 (SOX2) and the glial fibrillary acidic protein (GFAP) three days after the lesion in the deafferented medial vestibular nucleus. These two markers expressed on the same cell population could indicate the presence of lesion-reactive multipotent neural stem cells in the vestibular nuclei. The aim of our study was to provide insight into the potential neurogenic niche status of the vestibular nuclei in physiological conditions by using specific markers of stem cells (Nestin, SOX2, GFAP), cell proliferation (BrdU) and neuronal differentiation (NeuN). The present study confirmed the presence of quiescent and activated adult neural stem cells generating some new neurons in the vestibular nuclei of control rats. These unique features provide evidence that the vestibular nuclei represent a novel NSC site for the generation of neurons and/or glia in the adult rodent under physiological conditions.
Collapse
Affiliation(s)
- Guillaume Rastoldo
- Aix Marseille Université-CNRS, Laboratoire de Neurosciences Cognitives, LNC UMR 7291, 13331 Marseille, France
| | - Isabelle Watabe
- Aix Marseille Université-CNRS, Laboratoire de Neurosciences Cognitives, LNC UMR 7291, 13331 Marseille, France
| | - Agnes Lapotre
- Aix Marseille Université-CNRS, Laboratoire de Neurosciences Cognitives, LNC UMR 7291, 13331 Marseille, France
| | - Alain Tonetto
- Fédération de Recherche Sciences Chimiques Marseille FR 1739, Pôle PRATIM, 13331 Marseille, France
| | - Alejandra López-Juárez
- Department of Chemical, Electronic and Biomedical Engineering, Division of Sciences and Engineering, University of Guanajuato, Guanajuato 38116, Mexico
| | - Brahim Tighilet
- Aix Marseille Université-CNRS, Laboratoire de Neurosciences Cognitives, LNC UMR 7291, 13331 Marseille, France
- GDR Physiopathologie Vestibulaire-Unité GDR2074 CNRS, 13331 Marseille, France
- Correspondence: ; Tel.: +33-413550881; Fax: +33-413550869
| |
Collapse
|
14
|
Dunville K, Tonelli F, Novelli E, Codino A, Massa V, Frontino AM, Galfrè S, Biondi F, Gustincich S, Caleo M, Pandolfini L, Alia C, Cremisi F. Laminin 511 and WNT signalling sustain prolonged expansion of hiPSC-derived hippocampal progenitors. Development 2022; 149:276383. [DOI: 10.1242/dev.200353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 08/08/2022] [Indexed: 11/20/2022]
Abstract
ABSTRACT
Using the timely re-activation of WNT signalling in neuralizing human induced pluripotent stem cells (hiPSCs), we have produced neural progenitor cells with a gene expression profile typical of human embryonic dentate gyrus (DG) cells. Notably, in addition to continuous WNT signalling, a specific laminin isoform is crucial to prolonging the neural stem state and to extending progenitor cell proliferation for over 200 days in vitro. Laminin 511 is indeed specifically required to support proliferation and to inhibit differentiation of hippocampal progenitor cells for extended time periods when compared with a number of different laminin isoforms assayed. Global gene expression profiles of these cells suggest that a niche of laminin 511 and WNT signalling is sufficient to maintain their capability to undergo typical hippocampal neurogenesis. Moreover, laminin 511 signalling sustains the expression of a set of genes responsible for the maintenance of a hippocampal neurogenic niche. Finally, xenograft of human DG progenitors into the DG of adult immunosuppressed host mice produces efficient integration of neurons that innervate CA3 layer cells spanning the same area of endogenous hippocampal neuron synapses.
Collapse
Affiliation(s)
- Keagan Dunville
- Laboratorio di Biologia, Scuola Normale Superiore 1 , Pisa, 56126 , Italy
| | - Fabrizio Tonelli
- Laboratorio di Biologia, Scuola Normale Superiore 1 , Pisa, 56126 , Italy
| | - Elena Novelli
- Istituto di Neuroscienze, Consiglio Nazionale delle Ricerche 2 , Pisa, 56124 , Italy
| | - Azzurra Codino
- Center for Human Technologies, Central RNA Lab, Istituto Italiano di Tecnologia 3 , Genova, 16152 , Italy
| | - Verediana Massa
- Istituto di Neuroscienze, Consiglio Nazionale delle Ricerche 2 , Pisa, 56124 , Italy
| | | | - Silvia Galfrè
- Department of Biology and Biotechnologies ‘Charles Darwin’, Università La Sapienza 4 , Roma, 00185 , Italy
| | - Francesca Biondi
- Istituto di Neuroscienze, Consiglio Nazionale delle Ricerche 2 , Pisa, 56124 , Italy
| | - Stefano Gustincich
- Center for Human Technologies, Central RNA Lab, Istituto Italiano di Tecnologia 3 , Genova, 16152 , Italy
| | - Matteo Caleo
- Istituto di Neuroscienze, Consiglio Nazionale delle Ricerche 2 , Pisa, 56124 , Italy
| | - Luca Pandolfini
- Center for Human Technologies, Central RNA Lab, Istituto Italiano di Tecnologia 3 , Genova, 16152 , Italy
| | - Claudia Alia
- Istituto di Neuroscienze, Consiglio Nazionale delle Ricerche 2 , Pisa, 56124 , Italy
| | - Federico Cremisi
- Laboratorio di Biologia, Scuola Normale Superiore 1 , Pisa, 56126 , Italy
| |
Collapse
|
15
|
Lim R, Banerjee A, Biswas R, Chari AN, Raghavan S. Mechanotransduction through adhesion molecules: Emerging roles in regulating the stem cell niche. Front Cell Dev Biol 2022; 10:966662. [PMID: 36172276 PMCID: PMC9511051 DOI: 10.3389/fcell.2022.966662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 07/18/2022] [Indexed: 11/23/2022] Open
Abstract
Stem cells have been shown to play an important role in regenerative medicine due to their proliferative and differentiation potential. The challenge, however, lies in regulating and controlling their potential for this purpose. Stem cells are regulated by growth factors as well as an array of biochemical and mechanical signals. While the role of biochemical signals and growth factors in regulating stem cell homeostasis is well explored, the role of mechanical signals has only just started to be investigated. Stem cells interact with their niche or to other stem cells via adhesion molecules that eventually transduce mechanical cues to maintain their homeostatic function. Here, we present a comprehensive review on our current understanding of the influence of the forces perceived by cell adhesion molecules on the regulation of stem cells. Additionally, we provide insights on how this deeper understanding of mechanobiology of stem cells has translated toward therapeutics.
Collapse
Affiliation(s)
- Ryan Lim
- A∗STAR Skin Research Lab (ASRL), Agency for Science, Technology and Research (A*STAR) 8A Biomedical Grove, Singapore, Singapore
| | - Avinanda Banerjee
- A∗STAR Skin Research Lab (ASRL), Agency for Science, Technology and Research (A*STAR) 8A Biomedical Grove, Singapore, Singapore
| | - Ritusree Biswas
- Institute for Stem Cell Science and Regenerative Medicine (inStem), GKVK Campus, Bangalore, India
- Sastra University, Thanjavur, TN, India
| | - Anana Nandakumar Chari
- A∗STAR Skin Research Lab (ASRL), Agency for Science, Technology and Research (A*STAR) 8A Biomedical Grove, Singapore, Singapore
| | - Srikala Raghavan
- A∗STAR Skin Research Lab (ASRL), Agency for Science, Technology and Research (A*STAR) 8A Biomedical Grove, Singapore, Singapore
- Institute for Stem Cell Science and Regenerative Medicine (inStem), GKVK Campus, Bangalore, India
| |
Collapse
|
16
|
Norton ES, Whaley LA, Ulloa-Navas MJ, García-Tárraga P, Meneses KM, Lara-Velazquez M, Zarco N, Carrano A, Quiñones-Hinojosa A, García-Verdugo JM, Guerrero-Cázares H. Glioblastoma disrupts the ependymal wall and extracellular matrix structures of the subventricular zone. Fluids Barriers CNS 2022; 19:58. [PMID: 35821139 PMCID: PMC9277938 DOI: 10.1186/s12987-022-00354-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 06/27/2022] [Indexed: 12/04/2022] Open
Abstract
Background Glioblastoma (GBM) is the most aggressive and common type of primary brain tumor in adults. Tumor location plays a role in patient prognosis, with tumors proximal to the lateral ventricles (LVs) presenting with worse overall survival, increased expression of stem cell genes, and increased incidence of distal tumor recurrence. This may be due in part to interaction of GBM with factors of the subventricular zone (SVZ), including those contained within the cerebrospinal fluid (CSF). However, direct interaction of GBM tumors with CSF has not been proved and would be hindered in the presence of an intact ependymal cell layer. Methods Here, we investigate the ependymal cell barrier and its derived extracellular matrix (ECM) fractones in the vicinity of a GBM tumor. Patient-derived GBM cells were orthotopically implanted into immunosuppressed athymic mice in locations distal and proximal to the LV. A PBS vehicle injection in the proximal location was included as a control. At four weeks post-xenograft, brain tissue was examined for alterations in ependymal cell health via immunohistochemistry, scanning electron microscopy, and transmission electron microscopy. Results We identified local invading GBM cells within the LV wall and increased influx of CSF into the LV-proximal GBM tumor bulk compared to controls. In addition to the physical disruption of the ependymal cell barrier, we also identified increased signs of compromised ependymal cell health in LV-proximal tumor-bearing mice. These signs include increased accumulation of lipid droplets, decreased cilia length and number, and decreased expression of cell channel proteins. We additionally identified elevated numbers of small fractones in the SVZ within this group, suggesting increased indirect CSF-contained molecule signaling to tumor cells. Conclusions Our data is the first to show that LV-proximal GBMs physically disrupt the ependymal cell barrier in animal models, resulting in disruptions in ependymal cell biology and increased CSF interaction with the tumor bulk. These findings point to ependymal cell health and CSF-contained molecules as potential axes for therapeutic targeting in the treatment of GBM. Supplementary Information The online version contains supplementary material available at 10.1186/s12987-022-00354-8.
Collapse
Affiliation(s)
- Emily S Norton
- Department of Neurosurgery, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL, 32224, USA.,Neuroscience Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL, USA.,Regenerative Sciences Training Program, Center for Regenerative Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Lauren A Whaley
- Department of Neurosurgery, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL, 32224, USA.,Department of Biology, University of North Florida, Jacksonville, FL, USA
| | - María José Ulloa-Navas
- Laboratory of Comparative Neurobiology, Cavanilles Institute of Biodiversity and Evolutionary Biology, University of Valencia, CIBERNED, Paterna, Spain.,Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Patricia García-Tárraga
- Laboratory of Comparative Neurobiology, Cavanilles Institute of Biodiversity and Evolutionary Biology, University of Valencia, CIBERNED, Paterna, Spain
| | - Kayleah M Meneses
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA.,Biochemistry and Molecular Biology Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL, USA
| | | | - Natanael Zarco
- Department of Neurosurgery, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Anna Carrano
- Department of Neurosurgery, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | | | - José Manuel García-Verdugo
- Laboratory of Comparative Neurobiology, Cavanilles Institute of Biodiversity and Evolutionary Biology, University of Valencia, CIBERNED, Paterna, Spain
| | - Hugo Guerrero-Cázares
- Department of Neurosurgery, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL, 32224, USA.
| |
Collapse
|
17
|
Melrose J. Fractone Stem Cell Niche Components Provide Intuitive Clues in the Design of New Therapeutic Procedures/Biomatrices for Neural Repair. Int J Mol Sci 2022; 23:5148. [PMID: 35563536 PMCID: PMC9103880 DOI: 10.3390/ijms23095148] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/25/2022] [Accepted: 05/02/2022] [Indexed: 02/04/2023] Open
Abstract
The aim of this study was to illustrate recent developments in neural repair utilizing hyaluronan as a carrier of olfactory bulb stem cells and in new bioscaffolds to promote neural repair. Hyaluronan interacts with brain hyalectan proteoglycans in protective structures around neurons in perineuronal nets, which also have roles in the synaptic plasticity and development of neuronal cognitive properties. Specialist stem cell niches termed fractones located in the sub-ventricular and sub-granular regions of the dentate gyrus of the hippocampus migrate to the olfactory bulb, which acts as a reserve of neuroprogenitor cells in the adult brain. The extracellular matrix associated with the fractone stem cell niche contains hyaluronan, perlecan and laminin α5, which regulate the quiescent recycling of stem cells and also provide a means of escaping to undergo the proliferation and differentiation to a pluripotent migratory progenitor cell type that can participate in repair processes in neural tissues. Significant improvement in the repair of spinal cord injury and brain trauma has been reported using this approach. FGF-2 sequestered by perlecan in the neuroprogenitor niche environment aids in these processes. Therapeutic procedures have been developed using olfactory ensheathing stem cells and hyaluronan as a carrier to promote neural repair processes. Now that recombinant perlecan domain I and domain V are available, strategies may also be expected in the near future using these to further promote neural repair strategies.
Collapse
Affiliation(s)
- James Melrose
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, Northern Sydney Local Health District, St. Leonards, NSW 2065, Australia;
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
- Sydney Medical School, Northern, The University of Sydney, Royal North Shore Hospital, St. Leonards, NSW 2065, Australia
- Faculty of Medicine and Health, University of Sydney, Royal North Shore Hospital, St. Leonards, NSW 2065, Australia
| |
Collapse
|
18
|
Schaberg E, Götz M, Faissner A. The extracellular matrix molecule tenascin-C modulates cell cycle progression and motility of adult neural stem/progenitor cells from the subependymal zone. Cell Mol Life Sci 2022; 79:244. [PMID: 35430697 PMCID: PMC9013340 DOI: 10.1007/s00018-022-04259-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 02/16/2022] [Accepted: 03/18/2022] [Indexed: 11/30/2022]
Abstract
Adult neurogenesis has been described in two canonical regions of the adult central nervous system (CNS) of rodents, the subgranular zone (SGZ) of the hippocampus and the subependymal zone (SEZ) of the lateral ventricles. The stem cell niche of the SEZ provides a privileged environment composed of a specialized extracellular matrix (ECM) that comprises the glycoproteins tenascin-C (Tnc) and laminin-1 (LN1). In the present study, we investigated the function of these ECM glycoproteins in the adult stem cell niche. Adult neural stem/progenitor cells (aNSPCs) of the SEZ were prepared from wild type (Tnc+/+) and Tnc knockout (Tnc−/−) mice and analyzed using molecular and cell biological approaches. A delayed maturation of aNSPCs in Tnc−/− tissue was reflected by a reduced capacity to form neurospheres in response to epidermal growth factor (EGF). To examine a potential influence of the ECM on cell proliferation, aNSPCs of both genotypes were studied by cell tracking using digital video microscopy. aNSPCs were cultivated on three different substrates, namely, poly-d-lysine (PDL) and PDL replenished with either LN1 or Tnc for up to 6 days in vitro. On each of the three substrates aNSPCs displayed lineage trees that could be investigated with regard to cell cycle length. The latter appeared reduced in Tnc−/− aNSPCs on PDL and LN1 substrates, less so on Tnc that seemed to compensate the absence of the ECM compound to some extent. Close inspection of the lineage trees revealed a subpopulation of late dividing aNSPCslate that engaged into cycling after a notable delay. aNSPCslate exhibited a clearly different morphology, with a larger cell body and conspicuous processes. aNSPCslate reiterated the reduction in cell cycle length on all substrates tested, which was not rescued on Tnc substrates. When the migratory activity of aNSPC-derived progeny was determined, Tnc−/− neuroblasts displayed significantly longer migration tracks. This was traced to an increased rate of migration episodes compared to the wild-type cells that rested for longer time periods. We conclude that Tnc intervenes in the proliferation of aNSPCs and modulates the motility of neuroblasts in the niche of the SEZ.
Collapse
Affiliation(s)
- Elena Schaberg
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, Universitätsstrasse 150, 44780, Bochum, Germany
| | - Magdalena Götz
- Physiological Genomics, Biomedical Center, LMU, Planegg-Martinsried, Germany
- Institute of Stem Cell Research, Helmholtz Center Munich, Biomedical Center, LMU, Planegg-Martinsried, Germany
- Synergy, Excellence Cluster for Systems Neurology, BMC, LMU, Planegg-Martinsried, Germany
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, Universitätsstrasse 150, 44780, Bochum, Germany.
| |
Collapse
|
19
|
Quaresima S, Istiaq A, Jono H, Cacci E, Ohta K, Lupo G. Assessing the Role of Ependymal and Vascular Cells as Sources of Extracellular Cues Regulating the Mouse Ventricular-Subventricular Zone Neurogenic Niche. Front Cell Dev Biol 2022; 10:845567. [PMID: 35450289 PMCID: PMC9016221 DOI: 10.3389/fcell.2022.845567] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 03/18/2022] [Indexed: 11/13/2022] Open
Abstract
Neurogenesis persists in selected regions of the adult mouse brain; among them, the ventricular-subventricular zone (V-SVZ) of the lateral ventricles represents a major experimental paradigm due to its conspicuous neurogenic output. Postnatal V-SVZ neurogenesis is maintained by a resident population of neural stem cells (NSCs). Although V-SVZ NSCs are largely quiescent, they can be activated to enter the cell cycle, self-renew and generate progeny that gives rise to olfactory bulb interneurons. These adult-born neurons integrate into existing circuits to modify cognitive functions in response to external stimuli, but cells shed by V-SVZ NSCs can also reach injured brain regions, suggesting a latent regenerative potential. The V-SVZ is endowed with a specialized microenvironment, which is essential to maintain the proliferative and neurogenic potential of NSCs, and to preserve the NSC pool from exhaustion by finely tuning their quiescent and active states. Intercellular communication is paramount to the stem cell niche properties of the V-SVZ, and several extracellular signals acting in the niche milieu have been identified. An important part of these signals comes from non-neural cell types, such as local vascular cells, ependymal and glial cells. Understanding the crosstalk between NSCs and other niche components may aid therapeutic approaches for neuropathological conditions, since neurodevelopmental disorders, age-related cognitive decline and neurodegenerative diseases have been associated with dysfunctional neurogenic niches. Here, we review recent advances in the study of the complex interactions between V-SVZ NSCs and their cellular niche. We focus on the extracellular cues produced by ependymal and vascular cells that regulate NSC behavior in the mouse postnatal V-SVZ, and discuss the potential implication of these molecular signals in pathological conditions.
Collapse
Affiliation(s)
- Sabrina Quaresima
- Department of Biology and Biotechnology “C. Darwin”, Sapienza University of Rome, Rome, Italy
| | - Arif Istiaq
- Department of Stem Cell Biology, Faculty of Arts and Science, Kyushu University, Fukuoka, Japan
- Department of Brain Morphogenesis, Institute of Molecular Embryology and Genetics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hirofumi Jono
- Department of Pharmacy, Kumamoto University Hospital, Kumamoto, Japan
- Department of Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Emanuele Cacci
- Department of Biology and Biotechnology “C. Darwin”, Sapienza University of Rome, Rome, Italy
| | - Kunimasa Ohta
- Department of Stem Cell Biology, Faculty of Arts and Science, Kyushu University, Fukuoka, Japan
- *Correspondence: Kunimasa Ohta, ; Giuseppe Lupo,
| | - Giuseppe Lupo
- Department of Biology and Biotechnology “C. Darwin”, Sapienza University of Rome, Rome, Italy
- *Correspondence: Kunimasa Ohta, ; Giuseppe Lupo,
| |
Collapse
|
20
|
Smith MM, Hayes AJ, Melrose J. Pentosan Polysulphate (PPS), a Semi-Synthetic Heparinoid DMOAD With Roles in Intervertebral Disc Repair Biology emulating The Stem Cell Instructive and Tissue Reparative Properties of Heparan Sulphate. Stem Cells Dev 2022; 31:406-430. [PMID: 35102748 DOI: 10.1089/scd.2022.0007] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
This review highlights the attributes of pentosan polysulphate (PPS) in the promotion of intervertebral disc (IVD) repair processes. PPS has been classified as a disease modifying osteoarthritic drug (DMOAD) and many studies have demonstrated its positive attributes in the countering of degenerative changes occurring in cartilaginous tissues during the development of osteoarthritis (OA). Degenerative changes in the IVD also involve inflammatory cytokines, degradative proteases and cell signalling pathways similar to those operative in the development of OA in articular cartilage. PPS acts as a heparan sulphate (HS) mimetic to effect its beneficial effects in cartilage. The IVD contains small cell membrane HS-proteoglycans (HSPGs) such as syndecan, and glypican and a large multifunctional HS/chondroitin sulphate (CS) hybrid proteoglycan (HSPG2/perlecan) that have important matrix stabilising properties and sequester, control and present growth factors from the FGF, VEGF, PDGF and BMP families to cellular receptors to promote cell proliferation, differentiation and matrix synthesis. HSPG2 also has chondrogenic properties and stimulates the synthesis of extracellular matrix (ECM) components, expansion of cartilaginous rudiments and has roles in matrix stabilisation and repair. Perlecan is a perinuclear and nuclear proteoglycan in IVD cells with roles in chromatin organisation and control of transcription factor activity, immunolocalises to stem cell niches in cartilage, promotes escape of stem cells from quiescent recycling, differentiation and attainment of pluripotency and migratory properties. These participate in tissue development and morphogenesis, ECM remodelling and repair. PPS also localises in the nucleus of stromal stem cells, promotes development of chondroprogenitor cell lineages, ECM synthesis and repair and discal repair by resident disc cells. The availability of recombinant perlecan and PPS offer new opportunities in repair biology. These multifunctional agents offer welcome new developments in repair strategies for the IVD.
Collapse
Affiliation(s)
- Margaret M Smith
- The University of Sydney Raymond Purves Bone and Joint Research Laboratories, 247198, St Leonards, New South Wales, Australia;
| | - Anthony J Hayes
- Cardiff School of Biosciences, University of Cardiff, UK, Bioimaging Unit, Cardiff, Wales, United Kingdom of Great Britain and Northern Ireland;
| | - James Melrose
- Kolling Institute, University of Sydney, Royal North Shore Hospital, Raymond Purves Lab, Sydney Medical School Northern, Level 10, Kolling Institute B6, Royal North Shore Hospital, St. Leonards, New South Wales, Australia, 2065.,University of New South Wales, 7800, Graduate School of Biomedical Engineering, University of NSW, Sydney, New South Wales, Australia, 2052;
| |
Collapse
|
21
|
Deng S, Gan L, Liu C, Xu T, Zhou S, Guo Y, Zhang Z, Yang GY, Tian H, Tang Y. Roles of Ependymal Cells in the Physiology and Pathology of the Central Nervous System. Aging Dis 2022; 14:468-483. [PMID: 37008045 PMCID: PMC10017161 DOI: 10.14336/ad.2022.0826-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 08/26/2022] [Indexed: 11/18/2022] Open
Abstract
Ependymal cells are indispensable components of the central nervous system (CNS). They originate from neuroepithelial cells of the neural plate and show heterogeneity, with at least three types that are localized in different locations of the CNS. As glial cells in the CNS, accumulating evidence demonstrates that ependymal cells play key roles in mammalian CNS development and normal physiological processes by controlling the production and flow of cerebrospinal fluid (CSF), brain metabolism, and waste clearance. Ependymal cells have been attached to great importance by neuroscientists because of their potential to participate in CNS disease progression. Recent studies have demonstrated that ependymal cells participate in the development and progression of various neurological diseases, such as spinal cord injury and hydrocephalus, raising the possibility that they may serve as a potential therapeutic target for the disease. This review focuses on the function of ependymal cells in the developmental CNS as well as in the CNS after injury and discusses the underlying mechanisms of controlling the functions of ependymal cells.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Yaohui Tang
- Correspondence should be addressed to: Dr. Yaohui Tang, Med-X Research Institute and School of Biomedical Engineering Shanghai Jiao Tong University, Shanghai, China. .
| |
Collapse
|
22
|
Kerever A, Arikawa-Hirasawa E. Optimal Extracellular Matrix Niches for Neurogenesis: Identifying Glycosaminoglycan Chain Composition in the Subventricular Neurogenic Zone. Front Neuroanat 2021; 15:764458. [PMID: 34671246 PMCID: PMC8520954 DOI: 10.3389/fnana.2021.764458] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 09/14/2021] [Indexed: 11/17/2022] Open
Abstract
In the adult mammalian brain, new neurons are generated in a restricted region called the neurogenic niche, which refers to the specific regulatory microenvironment of neural stem cells (NSCs). Among the constituents of neurogenic niches, the extracellular matrix (ECM) has emerged as a key player in NSC maintenance, proliferation, and differentiation. In particular, heparan sulfate (HS) proteoglycans are capable of regulating various growth factor signaling pathways that influence neurogenesis. In this review, we summarize our current understanding of the ECM niche in the adult subventricular zone (SVZ), with a special focus on basement membrane (BM)-like structures called fractones, and discuss how fractones, particularly their composition of glycosaminoglycans (GAGs), may influence neurogenesis.
Collapse
Affiliation(s)
- Aurelien Kerever
- Research Institute for Diseases of Old Age, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Eri Arikawa-Hirasawa
- Research Institute for Diseases of Old Age, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| |
Collapse
|
23
|
McClenahan F, Dimitriou C, Koutsakis C, Dimitrakopoulos D, Arampatzis A, Kakouri P, Kourla M, Oikonomou S, Andreopoulou E, Patsonis M, Meri DK, Rasool RT, Franklin RJ, Kazanis I. Isolation of neural stem and oligodendrocyte progenitor cells from the brain of live rats. Stem Cell Reports 2021; 16:2534-2547. [PMID: 34560001 PMCID: PMC8514974 DOI: 10.1016/j.stemcr.2021.08.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 08/23/2021] [Accepted: 08/24/2021] [Indexed: 11/09/2022] Open
Abstract
Postnatal brain neural stem and progenitor cells (NSPCs) cluster in anatomically inaccessible stem cell niches, such as the subependymal zone (SEZ). Here, we describe a method for the isolation of NSPCs from live animals, which we term “milking.” The intracerebroventricular injection of a release cocktail, containing neuraminidase, integrin-β1-blocking antibody, and fibroblast growth factor 2, induces the controlled flow of NSPCs in the cerebrospinal fluid, where they are collected via liquid biopsies. Isolated cells retain key in vivo self-renewal properties and their cell-type profile reflects the cell composition of their source area, while the function of the niche is sustained even 8 months post-milking. By changing the target area more caudally, we also isolate oligodendrocyte progenitor cells (OPCs) from the corpus callosum. This novel approach for sampling NSPCs and OPCs paves the way for performing longitudinal studies in experimental animals, for more in vivo relevant cell culture assays, and for future clinical neuro-regenerative applications. Isolation of brain neural stem and oligodendrocyte progenitor cells from live rats Cells are induced to flow from their niche into the cerebrospinal fluid Neurogenesis persists despite long-term ependymal damage/loss Collected cells retain the properties of endogenous neural stem/progenitor cells
Collapse
Affiliation(s)
- Freyja McClenahan
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, CB2 0AW Cambridge, UK
| | - Christina Dimitriou
- Lab of Developmental Biology, Department of Biology, University of Patras, 26504 Patras, Greece
| | - Christos Koutsakis
- Lab of Developmental Biology, Department of Biology, University of Patras, 26504 Patras, Greece
| | | | - Asterios Arampatzis
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, CB2 0AW Cambridge, UK
| | - Paraskevi Kakouri
- Lab of Developmental Biology, Department of Biology, University of Patras, 26504 Patras, Greece
| | - Michaela Kourla
- Lab of Developmental Biology, Department of Biology, University of Patras, 26504 Patras, Greece
| | - Sofia Oikonomou
- Lab of Developmental Biology, Department of Biology, University of Patras, 26504 Patras, Greece
| | - Evangelia Andreopoulou
- Lab of Developmental Biology, Department of Biology, University of Patras, 26504 Patras, Greece
| | - Melina Patsonis
- Lab of Developmental Biology, Department of Biology, University of Patras, 26504 Patras, Greece
| | - Danai-Kassandra Meri
- Lab of Developmental Biology, Department of Biology, University of Patras, 26504 Patras, Greece
| | - Rana-Tahir Rasool
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, CB2 0AW Cambridge, UK
| | - Robin Jm Franklin
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, CB2 0AW Cambridge, UK
| | - Ilias Kazanis
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, CB2 0AW Cambridge, UK; Lab of Developmental Biology, Department of Biology, University of Patras, 26504 Patras, Greece.
| |
Collapse
|
24
|
Kim HJ, Lee E, Nam M, Chung JK, Joo S, Nam Y, Sun W. Contribution of Extracellular Matrix Component Landscapes in the Adult Subventricular Zone to the Positioning of Neural Stem/Progenitor Cells. Exp Neurobiol 2021; 30:275-284. [PMID: 34483142 PMCID: PMC8424380 DOI: 10.5607/en21012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 08/11/2021] [Accepted: 08/28/2021] [Indexed: 01/01/2023] Open
Abstract
Neurogenesis persists in restricted regions of the adult brain, including the subventricular zone (SVZ). Adult neural stem cells (NSCs) in the SVZ proliferate and give rise to new neurons and glial cells depending on intrinsic and environmental cues. Among the multiple factors that contribute to the chemical, physical, and mechanical components of the neurogenic niche, we focused on the composition of the extracellular matrix (ECM) of vasculature and fractones in the SVZ. The SVZ consists of ECM-rich blood vessels and fractones during development and adulthood, and adult neural stem/progenitor cells (NS/PCs) preferentially attach to the laminin-rich basal lamina. To examine the ECM preference of adult NS/PCs, we designed a competition assay using cell micropatterning. Although both laminin and collagen type IV, which are the main components of basal lamina, act as physical scaffolds, adult NS/PCs preferred to adhere to laminin over collagen type IV. Interestingly, the ECM preference of adult NS/PCs could be manipulated by chemokines such as stromal-derived factor 1 (SDF1) and α6 integrin. As SDF1 re-routes NSCs and their progenitors toward the injury site after brain damage, these results suggest that the alteration in ECM preferences may provide a molecular basis for context-dependent NS/PC positioning.
Collapse
Affiliation(s)
- Hyun Jung Kim
- Department of Anatomy, College of Medicine, Korea University, Seoul 02841, Korea.,Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejon 34141, Korea
| | - Eunsoo Lee
- Department of Anatomy, College of Medicine, Korea University, Seoul 02841, Korea.,Fluorescence Core Imaging Center, Ewha Womans University, Seoul 03760, Korea
| | - Myungwoo Nam
- Department of Anatomy, College of Medicine, Korea University, Seoul 02841, Korea
| | - Jae Kwon Chung
- Department of Anatomy, College of Medicine, Korea University, Seoul 02841, Korea
| | - Sunghoon Joo
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Yoonkey Nam
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Woong Sun
- Department of Anatomy, College of Medicine, Korea University, Seoul 02841, Korea
| |
Collapse
|
25
|
Diversity of Adult Neural Stem and Progenitor Cells in Physiology and Disease. Cells 2021; 10:cells10082045. [PMID: 34440814 PMCID: PMC8392301 DOI: 10.3390/cells10082045] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/23/2021] [Accepted: 08/03/2021] [Indexed: 02/07/2023] Open
Abstract
Adult neural stem and progenitor cells (NSPCs) contribute to learning, memory, maintenance of homeostasis, energy metabolism and many other essential processes. They are highly heterogeneous populations that require input from a regionally distinct microenvironment including a mix of neurons, oligodendrocytes, astrocytes, ependymal cells, NG2+ glia, vasculature, cerebrospinal fluid (CSF), and others. The diversity of NSPCs is present in all three major parts of the CNS, i.e., the brain, spinal cord, and retina. Intrinsic and extrinsic signals, e.g., neurotrophic and growth factors, master transcription factors, and mechanical properties of the extracellular matrix (ECM), collectively regulate activities and characteristics of NSPCs: quiescence/survival, proliferation, migration, differentiation, and integration. This review discusses the heterogeneous NSPC populations in the normal physiology and highlights their potentials and roles in injured/diseased states for regenerative medicine.
Collapse
|
26
|
Kerever A, Nagahara F, Keino-Masu K, Masu M, van Kuppevelt TH, Vivès RR, Arikawa-Hirasawa E. Regulation of fractone heparan sulfate composition in young and aged subventricular zone neurogenic niches. Glycobiology 2021; 31:1531-1542. [PMID: 34324645 DOI: 10.1093/glycob/cwab081] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/30/2021] [Accepted: 07/23/2021] [Indexed: 11/14/2022] Open
Abstract
Fractones, specialized extracellular matrix structures found in the subventricular zone (SVZ) neurogenic niche, can capture growth factors, such as basic fibroblast growth factor, from the extracellular milieu through a heparin-binding mechanism for neural stem cell presentation, which promotes neurogenesis. During aging, a decline in neurogenesis correlates with a change in the composition of heparan sulfate (HS) within fractones. In this study, we used antibodies that recognize specific short oligosaccharides with varying sulfation to evaluate the HS composition in fractones in young and aged brains. To further understand the conditions that regulate 6-O sulfation levels and its impact on neurogenesis, we used endosulfatase Sulf1 and Sulf2 double knock out (DKO) mice. Fractones in the SVZ of Sulf1/2 DKO mice showed immunoreactivity for the HS epitope, suggesting higher 6-O sulfation. While neurogenesis declined in the aged SVZ of both WT and Sulf1/2 DKO mice, we observed a larger number of neuroblasts in the young and aged SVZ of Sulf1/2 DKO mice. Together, these results show that the removal of 6-O-sulfation in fractones HS by endosulfatases inhibits neurogenesis in the SVZ. Our findings advance the current understanding regarding the extracellular environment that is best suited for neural stem cells to thrive, which is critical for the design of future stem cell therapies.
Collapse
Affiliation(s)
- Aurelien Kerever
- Research Institute for Diseases of Old Age, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Fumina Nagahara
- Research Institute for Diseases of Old Age, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kazuko Keino-Masu
- Department of Molecular Neurobiology, Faculty of Medicine, University of Tsukuba
| | - Masayuki Masu
- Department of Molecular Neurobiology, Faculty of Medicine, University of Tsukuba
| | - Toin H van Kuppevelt
- Department of Biochemistry, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Romain R Vivès
- University Grenoble Alpes, CNRS, CEA, IBS, Grenoble, France
| | - Eri Arikawa-Hirasawa
- Research Institute for Diseases of Old Age, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| |
Collapse
|
27
|
Omiya H, Yamaguchi S, Watanabe T, Kuniya T, Harada Y, Kawaguchi D, Gotoh Y. BMP signaling suppresses Gemc1 expression and ependymal differentiation of mouse telencephalic progenitors. Sci Rep 2021; 11:613. [PMID: 33436697 PMCID: PMC7804439 DOI: 10.1038/s41598-020-79610-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 11/18/2020] [Indexed: 01/29/2023] Open
Abstract
The lateral ventricles of the adult mammalian brain are lined by a single layer of multiciliated ependymal cells, which generate a flow of cerebrospinal fluid through directional beating of their cilia as well as regulate neurogenesis through interaction with adult neural stem cells. Ependymal cells are derived from a subset of embryonic neural stem-progenitor cells (NPCs, also known as radial glial cells) that becomes postmitotic during the late embryonic stage of development. Members of the Geminin family of transcriptional regulators including GemC1 and Mcidas play key roles in the differentiation of ependymal cells, but it remains largely unclear what extracellular signals regulate these factors and ependymal differentiation during embryonic and early-postnatal development. We now show that the levels of Smad1/5/8 phosphorylation and Id1/4 protein expression-both of which are downstream events of bone morphogenetic protein (BMP) signaling-decline in cells of the ventricular-subventricular zone in the mouse lateral ganglionic eminence in association with ependymal differentiation. Exposure of postnatal NPC cultures to BMP ligands or to a BMP receptor inhibitor suppressed and promoted the emergence of multiciliated ependymal cells, respectively. Moreover, treatment of embryonic NPC cultures with BMP ligands reduced the expression level of the ependymal marker Foxj1 and suppressed the emergence of ependymal-like cells. Finally, BMP ligands reduced the expression levels of Gemc1 and Mcidas in postnatal NPC cultures, whereas the BMP receptor inhibitor increased them. Our results thus implicate BMP signaling in suppression of ependymal differentiation from NPCs through regulation of Gemc1 and Mcidas expression during embryonic and early-postnatal stages of mouse telencephalic development.
Collapse
Affiliation(s)
- Hanae Omiya
- grid.26999.3d0000 0001 2151 536XGraduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033 Japan
| | - Shima Yamaguchi
- grid.26999.3d0000 0001 2151 536XGraduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033 Japan
| | - Tomoyuki Watanabe
- grid.26999.3d0000 0001 2151 536XGraduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033 Japan
| | - Takaaki Kuniya
- grid.26999.3d0000 0001 2151 536XGraduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033 Japan
| | - Yujin Harada
- grid.26999.3d0000 0001 2151 536XGraduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033 Japan
| | - Daichi Kawaguchi
- grid.26999.3d0000 0001 2151 536XGraduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033 Japan
| | - Yukiko Gotoh
- grid.26999.3d0000 0001 2151 536XGraduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033 Japan ,grid.26999.3d0000 0001 2151 536XInternational Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033 Japan
| |
Collapse
|
28
|
Dray N, Than-Trong E, Bally-Cuif L. Neural stem cell pools in the vertebrate adult brain: Homeostasis from cell-autonomous decisions or community rules? Bioessays 2020; 43:e2000228. [PMID: 33295062 DOI: 10.1002/bies.202000228] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 10/31/2020] [Accepted: 11/04/2020] [Indexed: 12/15/2022]
Abstract
Adult stem cell populations must coordinate their own maintenance with the generation of differentiated cell types to sustain organ physiology, in a spatially controlled manner and over long periods. Quantitative analyses of clonal dynamics have revealed that, in epithelia, homeostasis is achieved at the population rather than at the single stem cell level, suggesting that feedback mechanisms coordinate stem cell maintenance and progeny generation. In the central nervous system, however, little is known of the possible community processes underlying neural stem cell maintenance. Recent work, in part based on intravital imaging made possible in the adult zebrafish, conclusively highlights that homeostasis in neural stem cell pools may rely on population asymmetry and long-term spatiotemporal coordination of neural stem cell states and fates. These results suggest that neural stem cell assemblies in the vertebrate brain behave as self-organized systems, such that the stem cells themselves generate their own intrinsic niche.
Collapse
Affiliation(s)
- Nicolas Dray
- Zebrafish Neurogenetics Unit, CNRS, Team supported by the Ligue Nationale Contre le Cancer, Institut Pasteur, UMR3738, Paris, France
| | - Emmanuel Than-Trong
- Zebrafish Neurogenetics Unit, CNRS, Team supported by the Ligue Nationale Contre le Cancer, Institut Pasteur, UMR3738, Paris, France.,Ecole doctorale Biosigne, Le Kremlin Bicêtre, Université Paris-Saclay, France
| | - Laure Bally-Cuif
- Zebrafish Neurogenetics Unit, CNRS, Team supported by the Ligue Nationale Contre le Cancer, Institut Pasteur, UMR3738, Paris, France
| |
Collapse
|
29
|
Harkins D, Cooper HM, Piper M. The role of lipids in ependymal development and the modulation of adult neural stem cell function during aging and disease. Semin Cell Dev Biol 2020; 112:61-68. [PMID: 32771376 DOI: 10.1016/j.semcdb.2020.07.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/24/2020] [Accepted: 07/29/2020] [Indexed: 01/10/2023]
Abstract
Within the adult mammalian central nervous system, the ventricular-subventricular zone (V-SVZ) lining the lateral ventricles houses neural stem cells (NSCs) that continue to produce neurons throughout life. Developmentally, the V-SVZ neurogenic niche arises during corticogenesis following the terminal differentiation of telencephalic radial glial cells (RGCs) into either adult neural stem cells (aNSCs) or ependymal cells. In mice, these two cellular populations form rosettes during the late embryonic and early postnatal period, with ependymal cells surrounding aNSCs. These aNSCs and ependymal cells serve a number of key purposes, including the generation of neurons throughout life (aNSCs), and acting as a barrier between the CSF and the parenchyma and promoting CSF bulk flow (ependymal cells). Interestingly, the development of this neurogenic niche, as well as its ongoing function, has been shown to be reliant on different aspects of lipid biology. In this review we discuss the developmental origins of the rodent V-SVZ neurogenic niche, and highlight research which has implicated a role for lipids in the physiology of this part of the brain. We also discuss the role of lipids in the maintenance of the V-SVZ niche, and discuss new research which has suggested that alterations to lipid biology could contribute to ependymal cell dysfunction in aging and disease.
Collapse
Affiliation(s)
- Danyon Harkins
- School of Biomedical Sciences, The University of Queensland, Brisbane, 4072, Australia
| | - Helen M Cooper
- Queensland Brain Institute, The University of Queensland, Brisbane, 4072, Australia
| | - Michael Piper
- School of Biomedical Sciences, The University of Queensland, Brisbane, 4072, Australia; Queensland Brain Institute, The University of Queensland, Brisbane, 4072, Australia.
| |
Collapse
|
30
|
Iriyama S, Yasuda M, Nishikawa S, Takai E, Hosoi J, Amano S. Decrease of laminin-511 in the basement membrane due to photoaging reduces epidermal stem/progenitor cells. Sci Rep 2020; 10:12592. [PMID: 32724130 PMCID: PMC7387558 DOI: 10.1038/s41598-020-69558-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 07/14/2020] [Indexed: 02/08/2023] Open
Abstract
Daily sunlight exposure damages the epidermal basement membrane (BM) and disrupts epidermal homeostasis. Inter-follicular epidermal stem cells (IFE-SCs) regulate epidermal proliferation and differentiation, which supports epidermal homeostasis. Here, we examine how photoaging affects the function of IFE-SCs and we identify key components in their cellular environment (niche). We found that sun-exposed skin showed a decrease of MCSP-positive and β1-integrin-positive cells concomitantly with a decrease of laminin-511 at the dermal-epidermal junction (DEJ), as compared with sun-protected skin. Higher levels of laminin-511 were associated with not only increased efficiency of colony formation, but also higher expression levels of MCSP as well as other stem cell markers such as Lrig1, ITGB1, CD44, CD46, DLL1, and K15 in keratinocytes from skin of 12- to 62-year-old subjects. UVB exposure to cultured human skin impaired laminin-511 integrity at the dermal-epidermal junction and reduced MCSP-positive basal epidermal cells as well as K15-positive cells. Combined treatment with matrix metalloproteinase and heparanase inhibitors protected the integrity of laminin-511 and inhibited the reduction of MCSP-positive cells and K15-positive cells. These results suggest that photoaging may reduce the levels of MCSP-positive and K15-positive epidermal stem/progenitor cells in the epidermis via loss of laminin-511 at the dermal-epidermal junction.
Collapse
Affiliation(s)
- Shunsuke Iriyama
- Shiseido Global Innovation Center, 1-2-11 Takashima, Nishi-ku, Yokohama, 220-0011, Japan.
| | - Masahito Yasuda
- Department of Dermatology, Gunma University Graduate School of Medicine, 3-39-22 Showa-Machi, Maebashi, Gunma, 371-8511, Japan
| | - Saori Nishikawa
- Shiseido Global Innovation Center, 1-2-11 Takashima, Nishi-ku, Yokohama, 220-0011, Japan
| | - Eisuke Takai
- Shiseido Global Innovation Center, 1-2-11 Takashima, Nishi-ku, Yokohama, 220-0011, Japan
| | - Junichi Hosoi
- Shiseido Global Innovation Center, 1-2-11 Takashima, Nishi-ku, Yokohama, 220-0011, Japan
| | - Satoshi Amano
- Shiseido Global Innovation Center, 1-2-11 Takashima, Nishi-ku, Yokohama, 220-0011, Japan
| |
Collapse
|
31
|
Arreguin AJ, Colognato H. Brain Dysfunction in LAMA2-Related Congenital Muscular Dystrophy: Lessons From Human Case Reports and Mouse Models. Front Mol Neurosci 2020; 13:118. [PMID: 32792907 PMCID: PMC7390928 DOI: 10.3389/fnmol.2020.00118] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/09/2020] [Indexed: 12/26/2022] Open
Abstract
Laminin α2 gene (LAMA2)-related Congenital Muscular Dystrophy (CMD) was distinguished by a defining central nervous system (CNS) abnormality—aberrant white matter signals by MRI—when first described in the 1990s. In the past 25 years, researchers and clinicians have expanded our knowledge of brain involvement in LAMA2-related CMD, also known as Congenital Muscular Dystrophy Type 1A (MDC1A). Neurological changes in MDC1A can be structural, including lissencephaly and agyria, as well as functional, including epilepsy and intellectual disability. Mouse models of MDC1A include both spontaneous and targeted LAMA2 mutations and range from a partial loss of LAMA2 function (e.g., dy2J/dy2J), to a complete loss of LAMA2 expression (dy3K/dy3K). Diverse cellular and molecular changes have been reported in the brains of MDC1A mouse models, including blood-brain barrier dysfunction, altered neuro- and gliogenesis, changes in synaptic plasticity, and decreased myelination, providing mechanistic insight into potential neurological dysfunction in MDC1A. In this review article, we discuss selected studies that illustrate the potential scope and complexity of disturbances in brain development in MDC1A, and as well as highlight mechanistic insights that are emerging from mouse models.
Collapse
Affiliation(s)
- Andrea J Arreguin
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, United States.,Medical Scientist Training Program (MSTP), Stony Brook University, Stony Brook, NY, United States
| | - Holly Colognato
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, United States
| |
Collapse
|
32
|
Coelho-Sampaio T, Tenchov B, Nascimento MA, Hochman-Mendez C, Morandi V, Caarls MB, Altankov G. Type IV collagen conforms to the organization of polylaminin adsorbed on planar substrata. Acta Biomater 2020; 111:242-253. [PMID: 32450232 DOI: 10.1016/j.actbio.2020.05.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 05/14/2020] [Accepted: 05/14/2020] [Indexed: 12/15/2022]
Abstract
Tissue engineering demands the development of scaffolds that mimic natural extracellular matrices (ECM). Despite the success in obtaining synthetic interstitial ECM, the production of an artificial basement membrane (BM), the specialized thin sheet of ECM that is pivotal for the functional organization of most tissues and internal organs, is still not achieved. With the long-term aim of developing a flat BM-like structure here we investigated the behavior of acid-soluble Col IV during simultaneous assembly with laminin (LM) in acidic conditions. The underlying rationale was the previously observed phenomenon of acid-triggered LM polymerization, giving rise to biomimetic polylaminin (polyLM) that can be adsorbed on the substrate. Unexpectedly, we found that Col IV (that does not polymerize in acidic conditions) readily incorporated into the polyLM layer, forming a network that mimics to a great extent the characteristic polygonal morphology of single polyLM observable at micrometric scale. Scanning calorimetry and light scattering measurements supported the notion that polyLM and Col IV could directly interact. The biological properties of the proposed artificial BM-like structure were characterized using human keratinocytes (HACAT) and umbilical vein endothelial cells (HUVEC). HACAT formed stratified cell layers on the hybrid polyLM/Col IV layer, but not on Matrigel, nor on LM or Col IV alone, while HUVEC improved cortical F-actin and tight juctions organization on polyLM/Col IV. Thus, the proposed artificial BM reproduces not only morphological but also some functional properties of the natural BM. STATEMENT OF SIGNIFICANCE: Basement membranes (BMs) are flat biological matrices separating tissue compartments in the body. Their peculiar sheet-like structure is thought to result from the association of two independent protein networks of laminin and collagen IV. While pursuing the development of an artificial BM, we found that, when mixed with acid-induced polymerized laminin, collagen IV immediately conformed to the laminin shape. This implies that the protein networks may not be independently assembled as believed so far, but instead that laminin may command the assembly of collagen IV. Our hybrid matrix was structurally more stable than the commercial BM extract Matrigel and, unlike the latter, supported in vitro formation of a stratified layer of keratinocytes that approximated the organization of the natural epidermis.
Collapse
|
33
|
Kiyozumi D, Nakano I, Sato-Nishiuchi R, Tanaka S, Sekiguchi K. Laminin is the ECM niche for trophoblast stem cells. Life Sci Alliance 2020; 3:3/2/e201900515. [PMID: 31937556 PMCID: PMC6977391 DOI: 10.26508/lsa.201900515] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 01/06/2020] [Accepted: 01/07/2020] [Indexed: 01/07/2023] Open
Abstract
Laminin functions as an ECM niche factor for trophoblast stem cells and secures trophoblast stem cell expansion through its interactions with integrin. The niche is a specialized microenvironment for tissue stem cells in vivo. It has long been emphasized that niche ECM molecules act on tissue stem cells to regulate their behavior, but the molecular entities of these interactions remain to be fully elucidated. Here, we report that laminin forms the in vivo ECM niche for trophoblast stem cells (TSCs), the tissue stem cells of the placenta. TSCs expressed fibronectin-binding, vitronectin-binding, and laminin-binding integrins, whereas the integrin ligands present in the TSC niche were collagen and laminin. Therefore, the only niche integrin ligand available for TSCs in vivo was laminin. Laminin promoted TSC adhesion and proliferation in vitro in an integrin binding–dependent manner. Importantly, when the integrin-binding ability of laminin was genetically ablated in mice, the size of the TSC population was significantly reduced compared with that in control mice. The present findings underscore an ECM niche function of laminin to support tissue stem cell maintenance in vivo.
Collapse
Affiliation(s)
- Daiji Kiyozumi
- Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Osaka, Japan
| | - Itsuko Nakano
- Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Osaka, Japan
| | - Ryoko Sato-Nishiuchi
- Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Osaka, Japan
| | - Satoshi Tanaka
- Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Kiyotoshi Sekiguchi
- Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Osaka, Japan
| |
Collapse
|
34
|
Suzuki N, Hyodo M, Hayashi C, Mabuchi Y, Sekimoto K, Onchi C, Sekiguchi K, Akazawa C. Laminin α2, α4, and α5 Chains Positively Regulate Migration and Survival of Oligodendrocyte Precursor Cells. Sci Rep 2019; 9:19882. [PMID: 31882770 PMCID: PMC6934537 DOI: 10.1038/s41598-019-56488-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 12/11/2019] [Indexed: 12/31/2022] Open
Abstract
In the developing central nervous system (CNS), oligodendrocyte precursor cells (OPCs) migrate along blood vessels and are widely distributed in the CNS. Meanwhile, OPCs require survival factors from the extracellular microenvironment. In other tissues, laminins, heterotrimetric (αβγ) extracellular matrix proteins, promote cell migration and survival. However, the expression pattern and functions of laminins in OPC development remain poorly understood. In the present study, we first investigated the expression of laminin α chains, which bind to cell surface receptors such as integrins, in the postnatal murine brain. We found that laminin α1, α2, α4, and α5 chains were expressed around blood vessels and OPCs attached the laminin α chain-positive vessels. We then evaluated the effect of these laminins on OPCs activity using recombinant laminin E8s (LME8s) that are minimally active fragments of the laminin isoforms. OPCs attached on LM211E8, LM411E8, and LM511E8, containing laminin α2, α4, and α5 chains, respectively, through integrin β1. Further, these three LME8s promoted migration of OPCs, and OPC survival was prolonged on either LM411E8 or LM511E8 via the activation of focal adhesion kinase. Together, our findings suggest that laminins expressed surrounding blood vessels positively regulate migration and survival of OPCs through the integrin β1-FAK pathway.
Collapse
Affiliation(s)
- Nobuharu Suzuki
- Department of Molecular and Cellular Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan. .,Department of Biochemistry and Biophysics, Graduate School of Health Care Sciences, TMDU, Tokyo, Japan.
| | - Mai Hyodo
- Department of Biochemistry and Biophysics, Graduate School of Health Care Sciences, TMDU, Tokyo, Japan
| | - Chikako Hayashi
- Department of Molecular and Cellular Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan.,Department of Biochemistry and Biophysics, Graduate School of Health Care Sciences, TMDU, Tokyo, Japan
| | - Yo Mabuchi
- Department of Biochemistry and Biophysics, Graduate School of Health Care Sciences, TMDU, Tokyo, Japan.,Department of Biochemistry and Biophysics, Graduate School of Medical and Dental Sciences, TMDU, Tokyo, Japan
| | - Kaori Sekimoto
- Department of Biochemistry and Biophysics, Graduate School of Health Care Sciences, TMDU, Tokyo, Japan
| | - Chinami Onchi
- Department of Molecular and Cellular Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Kiyotoshi Sekiguchi
- Division of Matrixome Research and Application, Institute for Protein Research, Osaka University, Suita, Osaka, Japan
| | - Chihiro Akazawa
- Department of Biochemistry and Biophysics, Graduate School of Health Care Sciences, TMDU, Tokyo, Japan. .,Department of Biochemistry and Biophysics, Graduate School of Medical and Dental Sciences, TMDU, Tokyo, Japan.
| |
Collapse
|
35
|
Ahmed M, Marziali LN, Arenas E, Feltri ML, Ffrench-Constant C. Laminin α2 controls mouse and human stem cell behaviour during midbrain dopaminergic neuron development. Development 2019; 146:dev.172668. [PMID: 31371375 DOI: 10.1242/dev.172668] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 07/24/2019] [Indexed: 01/16/2023]
Abstract
Development of the central nervous system requires coordination of the proliferation and differentiation of neural stem cells. Here, we show that laminin alpha 2 (lm-α2) is a component of the midbrain dopaminergic neuron (mDA) progenitor niche in the ventral midbrain (VM) and identify a concentration-dependent role for laminin α2β1γ1 (lm211) in regulating mDA progenitor proliferation and survival via a distinct set of receptors. At high concentrations, lm211-rich environments maintain mDA progenitors in a proliferative state via integrins α6β1 and α7β1, whereas low concentrations of lm211 support mDA lineage survival via dystroglycan receptors. We confirmed our findings in vivo, demonstrating that the VM was smaller in the absence of lm-α2, with increased apoptosis; furthermore, the progenitor pool was depleted through premature differentiation, resulting in fewer mDA neurons. Examination of mDA neuron subtype composition showed a reduction in later-born mDA neurons of the ventral tegmental area, which control a range of cognitive behaviours. Our results identify a novel role for laminin in neural development and provide a possible mechanism for autism-like behaviours and the brainstem hypoplasia seen in some individuals with mutations of LAMA2.
Collapse
Affiliation(s)
- Maqsood Ahmed
- MRC Centre of Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Leandro N Marziali
- Departments of Biochemistry and Neurology, School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Ernest Arenas
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm 17177, Sweden
| | - M Laura Feltri
- Departments of Biochemistry and Neurology, School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | | |
Collapse
|
36
|
Morante-Redolat JM, Porlan E. Neural Stem Cell Regulation by Adhesion Molecules Within the Subependymal Niche. Front Cell Dev Biol 2019; 7:102. [PMID: 31245371 PMCID: PMC6581678 DOI: 10.3389/fcell.2019.00102] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 05/27/2019] [Indexed: 12/14/2022] Open
Abstract
In the mammalian adult brain, neural stem cells persist in neurogenic niches. The subependymal zone is the most prolific neurogenic niche in adult rodents, where residing stem cells generate large numbers of immature neurons that migrate into the olfactory bulb, where they differentiate into different types of interneurons. Subependymal neural stem cells derive from embryonic radial glia and retain some of their features like apico-basal polarity, with apical processes piercing the ependymal layer, and a basal process contacting blood vessels, constituting an epithelial niche. Conservation of the cytoarchitecture of the niche is of crucial importance for the maintenance of stem cells and for their neurogenic potential. In this minireview we will focus on extracellular matrix and adhesion molecules in the adult subependymal zone, showing their involvement not only as structural elements sustaining the niche architecture and topology, but also in the maintenance of stemness and regulation of the quiescence-proliferation balance.
Collapse
Affiliation(s)
- Jose Manuel Morante-Redolat
- Departamento de Biología Celular, Biología Funcional y Antropología Física, Universitat de València, Burjassot, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina, Universitat de València, Burjassot, Spain
| | - Eva Porlan
- Departamento de Neuropatología Molecular, Centro de Biología Molecular Severo Ochoa Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain.,Departamento de Biología Molecular, Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain.,Hospital La Paz Institute for Health Research (IdiPAZ), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
37
|
Obernier K, Alvarez-Buylla A. Neural stem cells: origin, heterogeneity and regulation in the adult mammalian brain. Development 2019; 146:146/4/dev156059. [PMID: 30777863 DOI: 10.1242/dev.156059] [Citation(s) in RCA: 351] [Impact Index Per Article: 58.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In the adult rodent brain, neural stem cells (NSCs) persist in the ventricular-subventricular zone (V-SVZ) and the subgranular zone (SGZ), which are specialized niches in which young neurons for the olfactory bulb (OB) and hippocampus, respectively, are generated. Recent studies have significantly modified earlier views on the mechanisms of NSC self-renewal and neurogenesis in the adult brain. Here, we discuss the molecular control, heterogeneity, regional specification and cell division modes of V-SVZ NSCs, and draw comparisons with NSCs in the SGZ. We highlight how V-SVZ NSCs are regulated by local signals from their immediate neighbors, as well as by neurotransmitters and factors that are secreted by distant neurons, the choroid plexus and vasculature. We also review recent advances in single cell RNA analyses that reveal the complexity of adult neurogenesis. These findings set the stage for a better understanding of adult neurogenesis, a process that one day may inspire new approaches to brain repair.
Collapse
Affiliation(s)
- Kirsten Obernier
- Eli and Edythe Broad Institute for Stem Cell Research and Regeneration Medicine, University of California, San Francisco, CA 94143, USA.,Department of Neurological Surgery, University of California, San Francisco, CA 94143, USA
| | - Arturo Alvarez-Buylla
- Eli and Edythe Broad Institute for Stem Cell Research and Regeneration Medicine, University of California, San Francisco, CA 94143, USA .,Department of Neurological Surgery, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
38
|
Sato Y, Kiyozumi D, Futaki S, Nakano I, Shimono C, Kaneko N, Ikawa M, Okabe M, Sawamoto K, Sekiguchi K. Ventricular-subventricular zone fractones are speckled basement membranes that function as a neural stem cell niche. Mol Biol Cell 2018; 30:56-68. [PMID: 30379609 PMCID: PMC6337917 DOI: 10.1091/mbc.e18-05-0286] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Neural stem cells (NSCs) are retained in the adult ventricular–subventricular zone (V-SVZ), a specialized neurogenic niche with a unique cellular architecture. It currently remains unclear whether or how NSCs utilize basement membranes (BMs) in this niche. Here, we examine the molecular compositions and functions of BMs in the adult mouse V-SVZ. Whole-mount V-SVZ immunostaining revealed that fractones, which are fingerlike processes of extravascular BMs, are speckled BMs unconnected to the vasculature, and differ in their molecular composition from vascular BMs. Glial fibrillary acidic protein (GFAP)-positive astrocytes and NSCs produce and adhere to speckled BMs. Furthermore, Gfap-Cre-mediated Lamc1flox(E1605Q) knockin mice, in which integrin-binding activities of laminins are specifically nullified in GFAP-positive cells, exhibit a decreased number and size of speckled BMs and reduced in vitro neurosphere-forming activity. Our results reveal niche activities of fractones/speckled BMs for NSCs and provide molecular insights into how laminin–integrin interactions regulate NSCs in vivo.
Collapse
Affiliation(s)
- Yuya Sato
- Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Suita, Osaka 565-0871, Japan
| | - Daiji Kiyozumi
- Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Suita, Osaka 565-0871, Japan
| | - Sugiko Futaki
- Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Suita, Osaka 565-0871, Japan
| | - Itsuko Nakano
- Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Suita, Osaka 565-0871, Japan
| | - Chisei Shimono
- Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Suita, Osaka 565-0871, Japan
| | - Naoko Kaneko
- Department of Developmental and Regenerative Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8610, Japan
| | - Masahito Ikawa
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Masaru Okabe
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Kazunobu Sawamoto
- Department of Developmental and Regenerative Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8610, Japan.,Division of Neural Development and Regeneration, National Institute for Physiological Sciences, Okazaki, Aichi 444-8585, Japan
| | - Kiyotoshi Sekiguchi
- Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Suita, Osaka 565-0871, Japan.,Laboratory of Matrixome Research and Application, Institute for Protein Research, Osaka University, Suita, Osaka 565-0871, Japan
| |
Collapse
|