1
|
Monticelli S, Giangrande A. Evolutionary Conservation of the Gcm/Glide Cascade: Of Glia and Beyond. BRAIN, BEHAVIOR AND EVOLUTION 2024; 100:58-66. [PMID: 39586239 DOI: 10.1159/000542753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 10/25/2024] [Indexed: 11/27/2024]
Abstract
BACKGROUND Glia represent a major cell population of the nervous system, and they take part in virtually any process sustaining the development, the functioning, and the pathology of the nervous system. Glial cells diversified significantly during evolution and distinct signals have been adopted to initiate glial development in mammals as compared to flies. In the invertebrate model Drosophila melanogaster, the transcription factor Gcm is necessary and sufficient to generate glial cells. Although Gcm orthologs have been found in protostomes and deuterostomes, they do not act in glial fate commitment as in flies, calling for further investigations of the evolutionarily conserved role of Gcm. SUMMARY Here, we review the impact of the fly Gcm transcription factor in the differentiation of phagocytic competent cells inside and outside the nervous system, glia, and macrophages, respectively. Then, we discuss the evolutionary conservation of Gcm and the neural/nonneural functions of Gcm orthologs. Finally, we present a recent work from Pavlidaki et al. [Cell Rep. 2022;41(3):111506] showing that the Gcm cascade is conserved from fly macrophages to mammalian microglia to counteract acute and chronic inflammation. KEY MESSAGES Gcm has an ancestral role in immunity, and its anti-inflammatory effect is evolutionarily conserved. This opens new avenues to assess Gcm function in other species/animal models, its potential involvement in inflammation-related processes, such as regeneration, and to expand the investigation on glia evolution.
Collapse
Affiliation(s)
- Sara Monticelli
- IGBMC, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, UMR 7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, UMR-S 1258, Illkirch, France
- Université de Strasbourg, IGBMC UMR 7104- UMR-S 1258, Illkirch, France
| | - Angela Giangrande
- IGBMC, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, UMR 7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, UMR-S 1258, Illkirch, France
- Université de Strasbourg, IGBMC UMR 7104- UMR-S 1258, Illkirch, France
| |
Collapse
|
2
|
Iram T, Garcia MA, Amand J, Kaur A, Atkins M, Iyer M, Lam M, Ambiel N, Jorgens DM, Keller A, Wyss-Coray T, Kern F, Zuchero JB. SRF transcriptionally regulates the oligodendrocyte cytoskeleton during CNS myelination. Proc Natl Acad Sci U S A 2024; 121:e2307250121. [PMID: 38483990 PMCID: PMC10962977 DOI: 10.1073/pnas.2307250121] [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: 05/10/2023] [Accepted: 02/10/2024] [Indexed: 03/19/2024] Open
Abstract
Myelination of neuronal axons is essential for nervous system development. Myelination requires dramatic cytoskeletal dynamics in oligodendrocytes, but how actin is regulated during myelination is poorly understood. We recently identified serum response factor (SRF)-a transcription factor known to regulate expression of actin and actin regulators in other cell types-as a critical driver of myelination in the aged brain. Yet, a major gap remains in understanding the mechanistic role of SRF in oligodendrocyte lineage cells. Here, we show that SRF is required cell autonomously in oligodendrocytes for myelination during development. Combining ChIP-seq with RNA-seq identifies SRF-target genes in oligodendrocyte precursor cells and oligodendrocytes that include actin and other key cytoskeletal genes. Accordingly, SRF knockout oligodendrocytes exhibit dramatically reduced actin filament levels early in differentiation, consistent with its role in actin-dependent myelin sheath initiation. Surprisingly, oligodendrocyte-restricted loss of SRF results in upregulation of gene signatures associated with aging and neurodegenerative diseases. Together, our findings identify SRF as a transcriptional regulator that controls the expression of cytoskeletal genes required in oligodendrocytes for myelination. This study identifies an essential pathway regulating oligodendrocyte biology with high relevance to brain development, aging, and disease.
Collapse
Affiliation(s)
- Tal Iram
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA94305
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA94305
| | - Miguel A. Garcia
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA94305
| | - Jérémy Amand
- Department of Clinical Bioinformatics, Helmholtz Institute for Pharmaceutical Research Saarland–Helmholtz Centre for Infection Research, Saarland University Campus, Saarbrücken66123, Germany
- Clinical Bioinformatics, Saarland University, Saarbrücken66123, Germany
| | - Achint Kaur
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA94305
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA94305
| | - Micaiah Atkins
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA94305
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA94305
| | - Manasi Iyer
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA94305
| | - Mable Lam
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA94305
| | - Nicholas Ambiel
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA94305
| | | | - Andreas Keller
- Department of Clinical Bioinformatics, Helmholtz Institute for Pharmaceutical Research Saarland–Helmholtz Centre for Infection Research, Saarland University Campus, Saarbrücken66123, Germany
- Clinical Bioinformatics, Saarland University, Saarbrücken66123, Germany
| | - Tony Wyss-Coray
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA94305
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA94305
| | - Fabian Kern
- Department of Clinical Bioinformatics, Helmholtz Institute for Pharmaceutical Research Saarland–Helmholtz Centre for Infection Research, Saarland University Campus, Saarbrücken66123, Germany
- Clinical Bioinformatics, Saarland University, Saarbrücken66123, Germany
| | - J. Bradley Zuchero
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA94305
| |
Collapse
|
3
|
Thumu SCR, Jain M, Soman S, Das S, Verma V, Nandi A, Gutmann DH, Jayaprakash B, Nair D, Clement JP, Marathe S, Ramanan N. SRF-deficient astrocytes provide neuroprotection in mouse models of excitotoxicity and neurodegeneration. eLife 2024; 13:e95577. [PMID: 38289036 PMCID: PMC10857791 DOI: 10.7554/elife.95577] [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: 12/27/2023] [Accepted: 01/15/2024] [Indexed: 02/10/2024] Open
Abstract
Reactive astrogliosis is a common pathological hallmark of CNS injury, infection, and neurodegeneration, where reactive astrocytes can be protective or detrimental to normal brain functions. Currently, the mechanisms regulating neuroprotective astrocytes and the extent of neuroprotection are poorly understood. Here, we report that conditional deletion of serum response factor (SRF) in adult astrocytes causes reactive-like hypertrophic astrocytes throughout the mouse brain. These SrfGFAP-ERCKO astrocytes do not affect neuron survival, synapse numbers, synaptic plasticity or learning and memory. However, the brains of Srf knockout mice exhibited neuroprotection against kainic-acid induced excitotoxic cell death. Relevant to human neurodegenerative diseases, SrfGFAP-ERCKO astrocytes abrogate nigral dopaminergic neuron death and reduce β-amyloid plaques in mouse models of Parkinson's and Alzheimer's disease, respectively. Taken together, these findings establish SRF as a key molecular switch for the generation of reactive astrocytes with neuroprotective functions that attenuate neuronal injury in the setting of neurodegenerative diseases.
Collapse
Affiliation(s)
| | - Monika Jain
- Centre for Neuroscience, Indian Institute of ScienceBangaloreIndia
| | - Sumitha Soman
- Centre for Neuroscience, Indian Institute of ScienceBangaloreIndia
| | - Soumen Das
- Centre for Neuroscience, Indian Institute of ScienceBangaloreIndia
| | - Vijaya Verma
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific ResearchBangaloreIndia
| | - Arnab Nandi
- Centre for Neuroscience, Indian Institute of ScienceBangaloreIndia
| | - David H Gutmann
- Department of Neurology, Washington University School of MedicineSt. LouisUnited States
| | | | - Deepak Nair
- Centre for Neuroscience, Indian Institute of ScienceBangaloreIndia
| | - James P Clement
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific ResearchBangaloreIndia
| | - Swananda Marathe
- Centre for Neuroscience, Indian Institute of ScienceBangaloreIndia
| | | |
Collapse
|
4
|
Song J, Dikwella N, Sinske D, Roselli F, Knöll B. SRF deletion results in earlier disease onset in a mouse model of amyotrophic lateral sclerosis. JCI Insight 2023; 8:e167694. [PMID: 37339001 PMCID: PMC10445689 DOI: 10.1172/jci.insight.167694] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 06/16/2023] [Indexed: 06/22/2023] Open
Abstract
Changes in neuronal activity modulate the vulnerability of motoneurons (MNs) in neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS). So far, the molecular basis of neuronal activity's impact in ALS is poorly understood. Herein, we investigated the impact of deleting the neuronal activity-stimulated transcription factor (TF) serum response factor (SRF) in MNs of SOD1G93A mice. SRF was present in vulnerable MMP9+ MNs. Ablation of SRF in MNs induced an earlier disease onset starting around 7-8 weeks after birth, as revealed by enhanced weight loss and decreased motor ability. This earlier disease onset in SRF-depleted MNs was accompanied by a mild elevation of neuroinflammation and neuromuscular synapse degeneration, whereas overall MN numbers and mortality were unaffected. In SRF-deficient mice, MNs showed impaired induction of autophagy-encoding genes, suggesting a potentially new SRF function in transcriptional regulation of autophagy. Complementary, constitutively active SRF-VP16 enhanced autophagy-encoding gene transcription and autophagy progression in cells. Furthermore, SRF-VP16 decreased ALS-associated aggregate induction. Chemogenetic modulation of neuronal activity uncovered SRF as having important TF-mediating activity-dependent effects, which might be beneficial to reduce ALS disease burden. Thus, our data identify SRF as a gene regulator connecting neuronal activity with the cellular autophagy program initiated in degenerating MNs.
Collapse
Affiliation(s)
- Jialei Song
- Institute of Neurobiochemistry and
- Department of Neurology, Ulm University, Ulm, Germany
| | - Natalie Dikwella
- Institute of Neurobiochemistry and
- Department of Neurology, Ulm University, Ulm, Germany
| | | | - Francesco Roselli
- Department of Neurology, Ulm University, Ulm, Germany
- German Center for Neurodegenerative Diseases-Ulm (DZNE-Ulm), Ulm, Germany
| | | |
Collapse
|
5
|
Tabuchi A, Ihara D. SRF in Neurochemistry: Overview of Recent Advances in Research on the Nervous System. Neurochem Res 2022; 47:2545-2557. [PMID: 35668335 DOI: 10.1007/s11064-022-03632-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/20/2022] [Accepted: 05/07/2022] [Indexed: 10/18/2022]
Abstract
Serum response factor (SRF) is a representative transcription factor that plays crucial roles in various biological phenomena by regulating immediate early genes (IEGs) and genes related to cell morphology and motility, among others. Over the years, the signal transduction pathways activating SRF have been clarified and SRF-target genes have been identified. In this overview, we initially briefly summarize the basic biology of SRF and its cofactors, ternary complex factor (TCF) and megakaryoblastic leukemia (MKL)/myocardin-related transcription factor (MRTF). Progress in the generation of nervous system-specific knockout (KO) or genetically modified mice as well as genetic analyses over the last few decades has not only identified novel SRF-target genes but also highlighted the neurochemical importance of SRF and its cofactors. Therefore, here we next present the phenotypes of mice with nervous system-specific KO of SRF or its cofactors by depicting recent findings associated with brain development, plasticity, epilepsy, stress response, and drug addiction, all of which result from function or dysfunction of the SRF axis. Last, we develop a hypothesis regarding the possible involvement of SRF and its cofactors in human neurological disorders including neurodegenerative, psychiatric, and neurodevelopmental diseases. This overview should deepen our understanding, highlight promising future directions for developing novel therapeutic strategies, and lead to illumination of the mechanisms underlying higher brain functions based on neuronal structure and function.
Collapse
Affiliation(s)
- Akiko Tabuchi
- Laboratory of Molecular Neurobiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan.
| | - Daisuke Ihara
- Laboratory of Molecular Neurobiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| |
Collapse
|
6
|
Ngo C, Kothary R. MicroRNAs in oligodendrocyte development and remyelination. J Neurochem 2022; 162:310-321. [PMID: 35536759 DOI: 10.1111/jnc.15618] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 03/14/2022] [Accepted: 04/20/2022] [Indexed: 11/28/2022]
Abstract
Oligodendrocytes are the glial cells responsible for the formation of myelin around axons of the central nervous system (CNS). Myelin is an insulating layer that allows electrical impulses to transmit quickly and efficiently along neurons. If myelin is damaged, as in chronic demyelinating disorders such as multiple sclerosis (MS), these impulses slow down. Remyelination by oligodendrocytes is often ineffective in MS, in part because of the failure of oligodendrocyte precursor cells (OPCs) to differentiate into mature, myelinating oligodendrocytes. The process of oligodendrocyte differentiation is tightly controlled by several regulatory networks involving transcription factors, intracellular signaling pathways, and extrinsic cues. Understanding the factors that regulate oligodendrocyte development is essential for the discovery of new therapeutic strategies capable of enhancing remyelination. Over the past decade, microRNAs (miRNAs) have emerged as key regulators of oligodendrocyte development, exerting effects on cell specification, proliferation, differentiation, and myelination. This article will review the role of miRNAs on oligodendrocyte biology and discuss their potential as promising therapeutic tools for remyelination.
Collapse
Affiliation(s)
- Clarissa Ngo
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Program in Biomedical Sciences, Faculty of Science, University of Ottawa, Ottawa, Ontario, Canada
| | - Rashmi Kothary
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada.,Centre for Neuromuscular Disease, University of Ottawa, Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, and Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
7
|
Long JY, Wang XJ, Li XY, Kong XH, Yang G, Zhang D, Yang YT, Shi Z, Ma XP. Spinal Microglia and Astrocytes: Two Key Players in Chronic Visceral Pain Pathogenesis. Neurochem Res 2022; 47:545-551. [PMID: 34797501 DOI: 10.1007/s11064-021-03486-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/05/2021] [Accepted: 11/08/2021] [Indexed: 10/19/2022]
Abstract
Chronic visceral pain (CVP) is one of the common symptoms of many diseases triggered by underlying diseases of the internal organs of the human body. Its causes include vascular mechanisms, mechanical factors, persistent inflammation, and unexplained functional mechanisms. Although the pathogenesis is unclear, more and more research has begun to shift from the neuronal aspect to the glial cells in recent years. Some data highlight that the spinal glial cells, particularly the microglia and astrocytes, play an essential role in CVP. Based on this, we highlight the mechanisms of microglia and astrocytes in CVP concerning the release of cytokines, chemokines, and neuroactive substances and alterations in intracellular signaling pathways during the process. Finally, because CVP is widespread in various diseases, we present future perspectives targeting microglia and astrocytes for treatment.
Collapse
Affiliation(s)
- Jun-Yi Long
- Shanghai Research Institute of Acupuncture and Meridian, No.650 South Wanping Road, Xuhui District, Shanghai, 200030, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, 200120, China
| | - Xue-Jun Wang
- Shanghai Research Institute of Acupuncture and Meridian, No.650 South Wanping Road, Xuhui District, Shanghai, 200030, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, 200120, China
| | - Xiao-Ying Li
- Shanghai Research Institute of Acupuncture and Meridian, No.650 South Wanping Road, Xuhui District, Shanghai, 200030, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, 200120, China
| | - Xie-He Kong
- Shanghai Research Institute of Acupuncture and Meridian, No.650 South Wanping Road, Xuhui District, Shanghai, 200030, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, 200120, China
| | - Guang Yang
- Shanghai Research Institute of Acupuncture and Meridian, No.650 South Wanping Road, Xuhui District, Shanghai, 200030, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, 200120, China
| | - Dan Zhang
- Shanghai Research Institute of Acupuncture and Meridian, No.650 South Wanping Road, Xuhui District, Shanghai, 200030, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, 200120, China
| | - Yan-Ting Yang
- Shanghai Research Institute of Acupuncture and Meridian, No.650 South Wanping Road, Xuhui District, Shanghai, 200030, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, 200120, China
| | - Zheng Shi
- Shanghai Research Institute of Acupuncture and Meridian, No.650 South Wanping Road, Xuhui District, Shanghai, 200030, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, 200120, China
| | - Xiao-Peng Ma
- Shanghai Research Institute of Acupuncture and Meridian, No.650 South Wanping Road, Xuhui District, Shanghai, 200030, China.
- Shanghai University of Traditional Chinese Medicine, Shanghai, 200120, China.
| |
Collapse
|
8
|
SRF Is Required for Maintenance of Astrocytes in Non-Reactive State in the Mammalian Brain. eNeuro 2021; 8:ENEURO.0447-19.2020. [PMID: 33441399 PMCID: PMC7877468 DOI: 10.1523/eneuro.0447-19.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 12/24/2020] [Accepted: 12/24/2020] [Indexed: 11/21/2022] Open
Abstract
Astrocytes play several critical roles in the normal functioning of the mammalian brain, including ion homeostasis, synapse formation, and synaptic plasticity. Following injury and infection or in the setting of neurodegeneration, astrocytes become hypertrophic and reactive, a process termed astrogliosis. Although acute reactive gliosis is beneficial in limiting further tissue damage, chronic gliosis becomes detrimental for neuronal recovery and regeneration. Several extracellular factors have been identified that generate reactive astrocytes; however, very little is known about the cell-autonomous transcriptional mechanisms that regulate the maintenance of astrocytes in the normal non-reactive state. Here, we show that conditional deletion of the stimulus-dependent transcription factor, serum response factor (SRF) in astrocytes (SrfGFAPCKO) results in astrogliosis marked by hypertrophic morphology and increased expression of GFAP, vimentin, and nestin. These reactive astrocytes were not restricted to any specific brain region and were seen in both white and gray matter in the entire brain. This astrogliosis persisted throughout adulthood concomitant with microglial activation. Importantly, the Srf mutant mouse brain did not exhibit any cell death or blood brain barrier (BBB) deficits suggesting that apoptosis and leaky BBB are not the causes for the reactive phenotype. The mutant astrocytes expressed more A2 reactive astrocyte marker genes and the SrfGFAPCKO mice exhibited normal neuronal numbers indicating that SRF-deficient gliosis astrocytes are not neurotoxic. Together, our findings suggest that SRF plays a critical role in astrocytes to maintain them in a non-reactive state.
Collapse
|
9
|
Sun S, Zhu XJ, Huang H, Guo W, Tang T, Xie B, Xu X, Zhang Z, Shen Y, Dai ZM, Qiu M. WNT signaling represses astrogliogenesis via Ngn2-dependent direct suppression of astrocyte gene expression. Glia 2019; 67:1333-1343. [PMID: 30889310 DOI: 10.1002/glia.23608] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 01/21/2019] [Accepted: 02/19/2019] [Indexed: 12/19/2022]
Abstract
Neural progenitor cells (NPCs) are sequentially specified into neurons and glia during the development of central nervous system. WNT/β-catenin signaling is known to regulate the balance between the proliferation and differentiation of NPCs during neurogenesis. However, the function of WNT/β-catenin signaling during gliogenesis remains poorly defined. Here, we report that activation of WNT/β-catenin signaling disrupts astrogliogenesis in the developing spinal cord. Conversely, inhibition of WNT/β-catenin signaling leads to precocious astrogliogenesis. Further analysis reveals that activation of WNT/β-catenin pathway results in a dramatic increase of neurogenin 2 (Ngn2) expression in transgenic mice, and knockdown of Ngn2 expression in neural precursor cells can reverse the inhibitory effect of WNT/β-catenin on astrocytic differentiation. Moreover, Ngn2 can directly bind to the promoters of several astrocyte specific genes and suppress their expression independent of STATs activity. Together, our studies provide the first in vivo evidence that WNT/β-catenin signaling inhibits early astrogliogenesis via an Ngn2-dependent transcriptional repression mechanism.
Collapse
Affiliation(s)
- Shuhui Sun
- Institute of Life Sciences, Key Laboratory of Organ Development and Regeneration of Zhejiang Province, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, PR China.,College of Life Sciences, Zhejiang University, Hangzhou, PR China
| | - Xiao-Jing Zhu
- Institute of Life Sciences, Key Laboratory of Organ Development and Regeneration of Zhejiang Province, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, PR China
| | - Hao Huang
- Institute of Life Sciences, Key Laboratory of Organ Development and Regeneration of Zhejiang Province, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, PR China
| | - Wei Guo
- Institute of Life Sciences, Key Laboratory of Organ Development and Regeneration of Zhejiang Province, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, PR China.,College of Life Sciences, Zhejiang University, Hangzhou, PR China
| | - Tao Tang
- Department of Anatomical Sciences and Neurobiology, University of Louisville, Louisville, Kentucky
| | - Binghua Xie
- Institute of Life Sciences, Key Laboratory of Organ Development and Regeneration of Zhejiang Province, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, PR China
| | - Xiaofeng Xu
- Institute of Life Sciences, Key Laboratory of Organ Development and Regeneration of Zhejiang Province, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, PR China
| | - Zunyi Zhang
- Institute of Life Sciences, Key Laboratory of Organ Development and Regeneration of Zhejiang Province, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, PR China
| | - Ying Shen
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of Ministry of Health, Zhejiang Province Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Zhong-Min Dai
- Institute of Life Sciences, Key Laboratory of Organ Development and Regeneration of Zhejiang Province, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, PR China
| | - Mengsheng Qiu
- Institute of Life Sciences, Key Laboratory of Organ Development and Regeneration of Zhejiang Province, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, PR China.,College of Life Sciences, Zhejiang University, Hangzhou, PR China
| |
Collapse
|
10
|
Abstract
Glia account for more than half of the cells in the mammalian nervous system, and the past few decades have witnessed a flood of studies that detail novel functions for glia in nervous system development, plasticity and disease. Here, and in the accompanying poster, we review the origins of glia and discuss their diverse roles during development, in the adult nervous system and in the context of disease.
Collapse
Affiliation(s)
- J Bradley Zuchero
- Department of Neurobiology, Stanford University, School of Medicine, Stanford, CA 94305, USA
| | - Ben A Barres
- Department of Neurobiology, Stanford University, School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
11
|
Tataranno ML, Perrone S, Buonocore G. Plasma Biomarkers of Oxidative Stress in Neonatal Brain Injury. Clin Perinatol 2015; 42:529-39. [PMID: 26250915 DOI: 10.1016/j.clp.2015.04.011] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Perinatal encephalopathy is a leading cause of lifelong disability. Increasing evidence indicates that the pathogenesis of perinatal brain damage is much more complex than originally thought, with multiple pathways involved. An important role of oxidative stress (OS) in the pathogenesis of brain injury is recognized for preterm and term infants. This article examines potential reliable and specific OS biomarkers that can be used in premature and term infants for the early detection and follow-up of the most common neonatal brain injuries, such as hypoxic-ischemic encephalopathy, intraventricular hemorrhage, and periventricular leukomalacia. The next step will be to explore the correlation between brain-specific OS biomarkers and functional brain outcomes.
Collapse
Affiliation(s)
- Maria Luisa Tataranno
- Department of Molecular and Developmental Medicine, University of Siena, Via Banchi di Sotto, 55, 53100 Siena, Italy
| | - Serafina Perrone
- Department of Molecular and Developmental Medicine, University of Siena, Via Banchi di Sotto, 55, 53100 Siena, Italy.
| | - Giuseppe Buonocore
- Department of Molecular and Developmental Medicine, University of Siena, Via Banchi di Sotto, 55, 53100 Siena, Italy
| |
Collapse
|
12
|
Nagao M, Lanjakornsiripan D, Itoh Y, Kishi Y, Ogata T, Gotoh Y. High mobility group nucleosome-binding family proteins promote astrocyte differentiation of neural precursor cells. Stem Cells 2015; 32:2983-97. [PMID: 25069414 DOI: 10.1002/stem.1787] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Revised: 06/17/2014] [Accepted: 06/23/2014] [Indexed: 11/07/2022]
Abstract
Astrocytes are the most abundant cell type in the mammalian brain and are important for the functions of the central nervous system. Although previous studies have shown that the STAT signaling pathway or its regulators promote the generation of astrocytes from multipotent neural precursor cells (NPCs) in the developing mammalian brain, the molecular mechanisms that regulate the astrocytic fate decision have still remained largely unclear. Here, we show that the high mobility group nucleosome-binding (HMGN) family proteins, HMGN1, 2, and 3, promote astrocyte differentiation of NPCs during brain development. HMGN proteins were expressed in NPCs, Sox9(+) glial progenitors, and GFAP(+) astrocytes in perinatal and adult brains. Forced expression of either HMGN1, 2, or 3 in NPCs in cultures or in the late embryonic neocortex increased the generation of astrocytes at the expense of neurons. Conversely, knockdown of either HMGN1, 2, or 3 in NPCs suppressed astrocyte differentiation and promoted neuronal differentiation. Importantly, overexpression of HMGN proteins did not induce the phosphorylation of STAT3 or activate STAT reporter genes. In addition, HMGN family proteins did not enhance DNA demethylation and acetylation of histone H3 around the STAT-binding site of the gfap promoter. Moreover, knockdown of HMGN family proteins significantly reduced astrocyte differentiation induced by gliogenic signal ciliary neurotrophic factor, which activates the JAK-STAT pathway. Therefore, we propose that HMGN family proteins are novel chromatin regulatory factors that control astrocyte fate decision/differentiation in parallel with or downstream of the JAK-STAT pathway through modulation of the responsiveness to gliogenic signals.
Collapse
Affiliation(s)
- Motoshi Nagao
- Department of Rehabilitation for the Movement Functions, Research Institute, National Rehabilitation Center for Persons with Disabilities, Saitama, Japan
| | | | | | | | | | | |
Collapse
|
13
|
Two functional promoter polymorphisms of neuregulin 1 gene are associated with progressive forms of multiple sclerosis. J Neurol Sci 2015; 351:154-159. [DOI: 10.1016/j.jns.2015.03.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 02/06/2015] [Accepted: 03/04/2015] [Indexed: 01/19/2023]
|
14
|
Anastasiadou S, Liebenehm S, Sinske D, Meyer zu Reckendorf C, Moepps B, Nordheim A, Knöll B. Neuronal expression of the transcription factor serum response factor modulates myelination in a mouse multiple sclerosis model. Glia 2015; 63:958-76. [PMID: 25639799 DOI: 10.1002/glia.22794] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 01/08/2015] [Accepted: 01/08/2015] [Indexed: 12/14/2022]
Abstract
In multiple sclerosis (MS), neurons in addition to inflammatory cells are now considered to mediate disease origin and progression. So far, molecular and cellular mechanisms of neuronal MS contributions are poorly understood. Herein we analyzed whether neuron-restricted signaling by the neuroprotective transcription factor serum response factor (SRF) modulates de- and remyelination in a rodent MS model. In the mouse cuprizone model, neuron- (Srf (flox/flox;CaMKCreERT2)) but not glia-specific (Srf (flox/flox;PlpCreERT2)) SRF depletion impaired demyelination suggesting impaired debris clearance by astrocytes and microglia. This supports an important role of SRF expression in neurons but not oligodendrocytes in de- and remyelination. During remyelination, NG2- and OLIG2-positive cells of the oligodendrocyte lineage as well as de novo mRNA synthesis of myelin genes were also reduced in neuron-specific Srf mutants. Using the stripe assay, we demonstrate that cortices of cuprizone-fed wild-type mice elicited astrocyte and microglia activation whereas this was abrogated in cuprizone-fed neuron-specific Srf mutants. We identified CCL chemokines (e.g. CCL2) as neuron-derived SRF-regulated paracrine signals rescuing immune cell activation upon neuronal SRF deletion. In summary, we uncovered important roles of neurons and neuronally expressed SRF in MS associated de- and remyelination.
Collapse
Affiliation(s)
- Sofia Anastasiadou
- Institute of Physiological Chemistry, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany
| | | | | | | | | | | | | |
Collapse
|
15
|
Nonaka M, Kim R, Sharry S, Matsushima A, Takemoto-Kimura S, Bito H. Towards a better understanding of cognitive behaviors regulated by gene expression downstream of activity-dependent transcription factors. Neurobiol Learn Mem 2014; 115:21-9. [PMID: 25173698 DOI: 10.1016/j.nlm.2014.08.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 08/18/2014] [Accepted: 08/20/2014] [Indexed: 12/12/2022]
Abstract
In the field of molecular and cellular neuroscience, it is not a trivial task to see the forest for the trees, where numerous, and seemingly independent, molecules often work in concert to control critical steps of synaptic plasticity and signalling. Here, we will first summarize our current knowledge on essential activity-dependent transcription factors (TFs) such as CREB, MEF2, Npas4 and SRF, then examine how various transcription cofactors (TcoFs) also contribute to defining the transcriptional outputs during learning and memory. This review finally attempts a provisory synthesis that sheds new light on some of the emerging principles of neuronal circuit dynamics driven by activity-regulated gene transcription to help better understand the intricate relationship between activity-dependent gene expression and cognitive behavior.
Collapse
Affiliation(s)
- Mio Nonaka
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan; Centre for Cognitive and Neural Systems, University of Edinburgh, 1 George Square, Edinburgh EH8 9JZ, United Kingdom
| | - Ryang Kim
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan; CREST-Japan Science and Technology Agency, Tokyo 102-0076, Japan
| | - Stuart Sharry
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Ayano Matsushima
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan; CREST-Japan Science and Technology Agency, Tokyo 102-0076, Japan
| | - Sayaka Takemoto-Kimura
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Haruhiko Bito
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan; CREST-Japan Science and Technology Agency, Tokyo 102-0076, Japan.
| |
Collapse
|
16
|
Malik N, Wang X, Shah S, Efthymiou AG, Yan B, Heman-Ackah S, Zhan M, Rao M. Comparison of the gene expression profiles of human fetal cortical astrocytes with pluripotent stem cell derived neural stem cells identifies human astrocyte markers and signaling pathways and transcription factors active in human astrocytes. PLoS One 2014; 9:e96139. [PMID: 24848099 PMCID: PMC4029581 DOI: 10.1371/journal.pone.0096139] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Accepted: 04/04/2014] [Indexed: 01/07/2023] Open
Abstract
Astrocytes are the most abundant cell type in the central nervous system (CNS) and have a multitude of functions that include maintenance of CNS homeostasis, trophic support of neurons, detoxification, and immune surveillance. It has only recently been appreciated that astrocyte dysfunction is a primary cause of many neurological disorders. Despite their importance in disease very little is known about global gene expression for human astrocytes. We have performed a microarray expression analysis of human fetal astrocytes to identify genes and signaling pathways that are important for astrocyte development and maintenance. Our analysis confirmed that the fetal astrocytes express high levels of the core astrocyte marker GFAP and the transcription factors from the NFI family which have been shown to play important roles in astrocyte development. A group of novel markers were identified that distinguish fetal astrocytes from pluripotent stem cell-derived neural stem cells (NSCs) and NSC-derived neurons. As in murine astrocytes, the Notch signaling pathway appears to be particularly important for cell fate decisions between the astrocyte and neuronal lineages in human astrocytes. These findings unveil the repertoire of genes expressed in human astrocytes and serve as a basis for further studies to better understand astrocyte biology, especially as it relates to disease.
Collapse
Affiliation(s)
- Nasir Malik
- National Institutes of Health, NIAMS, Bethesda, Maryland, United States of America
- * E-mail:
| | - Xiantao Wang
- National Institutes of Health, NIAMS, Bethesda, Maryland, United States of America
| | - Sonia Shah
- National Institutes of Health, NIAMS, Bethesda, Maryland, United States of America
| | | | - Bin Yan
- Hong Kong Baptist University, Department of Biology, Hong Kong
| | - Sabrina Heman-Ackah
- National Institutes of Health, NIAMS, Bethesda, Maryland, United States of America
| | - Ming Zhan
- The Methodist Hospital Research Institute, Weill Cornell Medical College, Houston, Texas, United States of America
| | - Mahendra Rao
- National Institutes of Health, NIAMS, Bethesda, Maryland, United States of America
- National Institutes of Health, NIH Center for Regenerative Medicine, Bethesda, Maryland, United States of America
| |
Collapse
|
17
|
Stein LR, Imai SI. Specific ablation of Nampt in adult neural stem cells recapitulates their functional defects during aging. EMBO J 2014; 33:1321-40. [PMID: 24811750 DOI: 10.1002/embj.201386917] [Citation(s) in RCA: 151] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Neural stem/progenitor cell (NSPC) proliferation and self-renewal, as well as insult-induced differentiation, decrease markedly with age. The molecular mechanisms responsible for these declines remain unclear. Here, we show that levels of NAD(+) and nicotinamide phosphoribosyltransferase (Nampt), the rate-limiting enzyme in mammalian NAD(+) biosynthesis, decrease with age in the hippocampus. Ablation of Nampt in adult NSPCs reduced their pool and proliferation in vivo. The decrease in the NSPC pool during aging can be rescued by enhancing hippocampal NAD(+) levels. Nampt is the main source of NSPC NAD(+) levels and required for G1/S progression of the NSPC cell cycle. Nampt is also critical in oligodendrocytic lineage fate decisions through a mechanism mediated redundantly by Sirt1 and Sirt2. Ablation of Nampt in the adult NSPCs in vivo reduced NSPC-mediated oligodendrogenesis upon insult. These phenotypes recapitulate defects in NSPCs during aging, giving rise to the possibility that Nampt-mediated NAD(+) biosynthesis is a mediator of age-associated functional declines in NSPCs.
Collapse
Affiliation(s)
- Liana R Stein
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Shin-ichiro Imai
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
18
|
Kawakami-Schulz SV, Verdoni AM, Sattler SG, Jessen E, Kao WWY, Ikeda A, Ikeda S. Serum response factor: positive and negative regulation of an epithelial gene expression network in the destrin mutant cornea. Physiol Genomics 2014; 46:277-89. [PMID: 24550211 DOI: 10.1152/physiolgenomics.00126.2013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Increased angiogenesis, inflammation, and proliferation are hallmarks of diseased tissues, and in vivo models of these disease phenotypes can provide insight into disease pathology. Dstn(corn1) mice, deficient for the actin depolymerizing factor destrin (DSTN), display an increase of serum response factor (SRF) that results in epithelial hyperproliferation, inflammation, and neovascularization in the cornea. Previous work demonstrated that conditional ablation of Srf from the corneal epithelium of Dstn(corn1) mice returns the cornea to a wild-type (WT) like state. This result implicated SRF as a major regulator of genes that contributes to abnormal phenotypes in Dstn(corn1) cornea. The purpose of this study is to identify gene networks that are affected by increased expression of Srf in the Dstn(corn1) cornea. Microarray analysis led to characterization of gene expression changes that occur when conditional knockout of Srf rescues mutant phenotypes in the cornea of Dstn(corn1) mice. Comparison of gene expression values from WT, Dstn(corn1) mutant, and Dstn(corn1) rescued cornea identified >400 differentially expressed genes that are downstream from SRF. Srf ablation had a significant effect on genes associated with epithelial cell-cell junctions and regulation of actin dynamics. The majority of genes affected by SRF are downregulated in the Dstn(corn1) mutant cornea, suggesting that increased SRF negatively affects transcription of SRF gene targets. ChIP-seq analysis on Dstn(corn1) mutant and WT tissue revealed that, despite being present in higher abundance, SRF binding is significantly decreased in the Dstn(corn1) mutant cornea. This study uses a unique model combining genetic and genomic approaches to identify genes that are regulated by SRF. These findings expand current understanding of the role of SRF in both normal and abnormal tissue homeostasis.
Collapse
|
19
|
de Leeuw CN, Dyka FM, Boye SL, Laprise S, Zhou M, Chou AY, Borretta L, McInerny SC, Banks KG, Portales-Casamar E, Swanson MI, D’Souza CA, Boye SE, Jones SJM, Holt RA, Goldowitz D, Hauswirth WW, Wasserman WW, Simpson EM. Targeted CNS Delivery Using Human MiniPromoters and Demonstrated Compatibility with Adeno-Associated Viral Vectors. Mol Ther Methods Clin Dev 2014; 1:5. [PMID: 24761428 PMCID: PMC3992516 DOI: 10.1038/mtm.2013.5] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 11/05/2013] [Indexed: 01/21/2023]
Abstract
Critical for human gene therapy is the availability of small promoter tools to drive gene expression in a highly specific and reproducible manner. We tackled this challenge by developing human DNA MiniPromoters using computational biology and phylogenetic conservation. MiniPromoters were tested in mouse as single-copy knock-ins at the Hprt locus on the X Chromosome, and evaluated for lacZ reporter expression in CNS and non-CNS tissue. Eighteen novel MiniPromoters driving expression in mouse brain were identified, two MiniPromoters for driving pan-neuronal expression, and 17 MiniPromoters for the mouse eye. Key areas of therapeutic interest were represented in this set: the cerebral cortex, embryonic hypothalamus, spinal cord, bipolar and ganglion cells of the retina, and skeletal muscle. We also demonstrated that three retinal ganglion cell MiniPromoters exhibit similar cell-type specificity when delivered via adeno-associated virus (AAV) vectors intravitreally. We conclude that our methodology and characterization has resulted in desirable expression characteristics that are intrinsic to the MiniPromoter, not dictated by copy number effects or genomic location, and results in constructs predisposed to success in AAV. These MiniPromoters are immediately applicable for pre-clinical studies towards gene therapy in humans, and are publicly available to facilitate basic and clinical research, and human gene therapy.
Collapse
Affiliation(s)
- Charles N de Leeuw
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Frank M Dyka
- Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Sanford L Boye
- Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Stéphanie Laprise
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Michelle Zhou
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Alice Y Chou
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Lisa Borretta
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Simone C McInerny
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kathleen G Banks
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Elodie Portales-Casamar
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Magdalena I Swanson
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Cletus A D’Souza
- Canada’s Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Shannon E Boye
- Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Steven JM Jones
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
- Canada’s Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Robert A Holt
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
- Canada’s Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Daniel Goldowitz
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - William W Hauswirth
- Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Wyeth W Wasserman
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Elizabeth M Simpson
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
20
|
Namihira M, Nakashima K. Mechanisms of astrocytogenesis in the mammalian brain. Curr Opin Neurobiol 2013; 23:921-7. [DOI: 10.1016/j.conb.2013.06.002] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Accepted: 06/10/2013] [Indexed: 10/26/2022]
|
21
|
Transcriptional Regulation and Specification of Neural Stem Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 786:129-55. [DOI: 10.1007/978-94-007-6621-1_8] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
22
|
Buller B, Chopp M, Ueno Y, Zhang L, Zhang RL, Morris D, Zhang Y, Zhang ZG. Regulation of serum response factor by miRNA-200 and miRNA-9 modulates oligodendrocyte progenitor cell differentiation. Glia 2012; 60:1906-14. [PMID: 22907787 DOI: 10.1002/glia.22406] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2012] [Accepted: 07/24/2012] [Indexed: 01/06/2023]
Abstract
Serum response factor (SRF) is a transcription factor that transactivates actin-associated genes and has been implicated in oligodendrocyte (OL) differentiation. To date, it has not been investigated in cerebral ischemia. We investigated the dynamics of SRF expression after stroke in vivo and the role of SRF in OL differentiation in vitro. Using immunohistochemistry, we found that SRF was upregulated in OLs and OL precursor cells (OPCs) after stroke. Moreover, upregulation of SRF was concurrent with downregulation of the micro-RNAs (miRNAs) miR-9 and the miR-200 family in the ischemic white matter region, the corpus callosum. Inhibition of SRF activation by CCG-1423, a specific inhibitor of SRF function, blocked OPCs from differentiating into OLs. Overexpression of miR-9 and miR-200 in cultured OPCs suppressed SRF expression and inhibited OPC differentiation. Moreover, co-expression of miR-9 and miR-200 attenuated activity of a luciferase reporter assay containing the Srf 3' untranslated region. Collectively, this study is the first to show that stroke upregulates SRF expression in OPCs and OLs, and that SRF levels are mediated by miRNAs and regulate OPC differentiation.
Collapse
Affiliation(s)
- Benjamin Buller
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan 48202, USA
| | | | | | | | | | | | | | | |
Collapse
|