1
|
Huynh NPT, Osipovitch M, Foti R, Bates J, Mansky B, Cano JC, Benraiss A, Zhao C, Lu QR, Goldman SA. Shared patterns of glial transcriptional dysregulation link Huntington's disease and schizophrenia. Brain 2024; 147:3099-3112. [PMID: 39028640 PMCID: PMC11370805 DOI: 10.1093/brain/awae166] [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: 09/23/2023] [Revised: 04/22/2024] [Accepted: 05/01/2024] [Indexed: 07/21/2024] Open
Abstract
Huntington's disease and juvenile-onset schizophrenia have long been regarded as distinct disorders. However, both manifest cell-intrinsic abnormalities in glial differentiation, with resultant astrocytic dysfunction and hypomyelination. To assess whether a common mechanism might underlie the similar glial pathology of these otherwise disparate conditions, we used comparative correlation network approaches to analyse RNA-sequencing data from human glial progenitor cells (hGPCs) produced from disease-derived pluripotent stem cells. We identified gene sets preserved between Huntington's disease and schizophrenia hGPCs yet distinct from normal controls that included 174 highly connected genes in the shared disease-associated network, focusing on genes involved in synaptic signalling. These synaptic genes were largely suppressed in both schizophrenia and Huntington's disease hGPCs, and gene regulatory network analysis identified a core set of upstream regulators of this network, of which OLIG2 and TCF7L2 were prominent. Among their downstream targets, ADGRL3, a modulator of glutamatergic synapses, was notably suppressed in both schizophrenia and Huntington's disease hGPCs. Chromatin immunoprecipitation sequencing confirmed that OLIG2 and TCF7L2 each bound to the regulatory region of ADGRL3, whose expression was then rescued by lentiviral overexpression of these transcription factors. These data suggest that the disease-associated suppression of OLIG2 and TCF7L2-dependent transcription of glutamate signalling regulators may impair glial receptivity to neuronal glutamate. The consequent loss of activity-dependent mobilization of hGPCs may yield deficient oligodendrocyte production, and hence the hypomyelination noted in these disorders, as well as the disrupted astrocytic differentiation and attendant synaptic dysfunction associated with each. Together, these data highlight the importance of convergent glial molecular pathology in both the pathogenesis and phenotypic similarities of two otherwise unrelated disorders, Huntington's disease and schizophrenia.
Collapse
Affiliation(s)
- Nguyen P T Huynh
- Center for Translational Neuromedicine, University of Copenhagen, Faculty of Health and Medical Sciences, 2200 Copenhagen, Denmark
- Center for Translational Neuromedicine and Department of Neurology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Mikhail Osipovitch
- Center for Translational Neuromedicine, University of Copenhagen, Faculty of Health and Medical Sciences, 2200 Copenhagen, Denmark
| | - Rossana Foti
- Center for Translational Neuromedicine, University of Copenhagen, Faculty of Health and Medical Sciences, 2200 Copenhagen, Denmark
| | - Janna Bates
- Center for Translational Neuromedicine and Department of Neurology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Benjamin Mansky
- Center for Translational Neuromedicine and Department of Neurology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Jose C Cano
- Center for Translational Neuromedicine and Department of Neurology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Abdellatif Benraiss
- Center for Translational Neuromedicine and Department of Neurology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Chuntao Zhao
- Division of Experimental Hematology and Cancer Biology, Department of Pediatrics, Brain Tumor Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Q Richard Lu
- Division of Experimental Hematology and Cancer Biology, Department of Pediatrics, Brain Tumor Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Steven A Goldman
- Center for Translational Neuromedicine, University of Copenhagen, Faculty of Health and Medical Sciences, 2200 Copenhagen, Denmark
- Center for Translational Neuromedicine and Department of Neurology, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
2
|
Marangon D, Castro e Silva JH, Cerrato V, Boda E, Lecca D. Oligodendrocyte Progenitors in Glial Scar: A Bet on Remyelination. Cells 2024; 13:1024. [PMID: 38920654 PMCID: PMC11202012 DOI: 10.3390/cells13121024] [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: 05/07/2024] [Revised: 06/07/2024] [Accepted: 06/10/2024] [Indexed: 06/27/2024] Open
Abstract
Oligodendrocyte progenitor cells (OPCs) represent a subtype of glia, giving rise to oligodendrocytes, the myelin-forming cells in the central nervous system (CNS). While OPCs are highly proliferative during development, they become relatively quiescent during adulthood, when their fate is strictly influenced by the extracellular context. In traumatic injuries and chronic neurodegenerative conditions, including those of autoimmune origin, oligodendrocytes undergo apoptosis, and demyelination starts. Adult OPCs become immediately activated; they migrate at the lesion site and proliferate to replenish the damaged area, but their efficiency is hampered by the presence of a glial scar-a barrier mainly formed by reactive astrocytes, microglia and the deposition of inhibitory extracellular matrix components. If, on the one hand, a glial scar limits the lesion spreading, it also blocks tissue regeneration. Therapeutic strategies aimed at reducing astrocyte or microglia activation and shifting them toward a neuroprotective phenotype have been proposed, whereas the role of OPCs has been largely overlooked. In this review, we have considered the glial scar from the perspective of OPCs, analysing their behaviour when lesions originate and exploring the potential therapies aimed at sustaining OPCs to efficiently differentiate and promote remyelination.
Collapse
Affiliation(s)
- Davide Marangon
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy; (D.M.); (J.H.C.e.S.)
| | - Juliana Helena Castro e Silva
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy; (D.M.); (J.H.C.e.S.)
| | - Valentina Cerrato
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, 10126 Turin, Italy; (V.C.); (E.B.)
- Neuroscience Institute Cavalieri Ottolenghi, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
| | - Enrica Boda
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, 10126 Turin, Italy; (V.C.); (E.B.)
- Neuroscience Institute Cavalieri Ottolenghi, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
| | - Davide Lecca
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy; (D.M.); (J.H.C.e.S.)
| |
Collapse
|
3
|
Xu J, Wang R, Luo W, Mao X, Gao H, Feng X, Chen G, Yang Z, Deng W, Nie Y. Oligodendrocyte progenitor cell-specific delivery of lipid nanoparticles loaded with Olig2 synthetically modified messenger RNA for ischemic stroke therapy. Acta Biomater 2024; 174:297-313. [PMID: 38096960 DOI: 10.1016/j.actbio.2023.12.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 12/02/2023] [Accepted: 12/05/2023] [Indexed: 12/23/2023]
Abstract
The transcription factor Olig2 is highly expressed throughout oligodendroglial development and is needed for the differentiation of oligodendrocyte progenitor cells (OPCs) into oligodendrocytes and remyelination. Although Olig2 overexpression in OPCs is a possible therapeutic target for enhancing myelin repair in ischemic stroke, achieving Olig2 overexpression in vivo remains a formidable technological challenge. To address this challenge, we employed lipid nanoparticle (LNP)-mediated delivery of Olig2 synthetically modified messenger RNA (mRNA) as a viable method for in vivo Olih2 protein overexpression. Specifically, we developed CD140a-targeted LNPs loaded with Olig2 mRNA (C-Olig2) to achieve targeted Olig2 protein expression within PDGFRα+ OPCs, with the goal of promoting remyelination for ischemic stroke therapy. We show that C-Olig2 promotes the differentiation of PDGFRα+ OPCs derived from mouse neural stem cells into mature oligodendrocytes in vitro, suggesting that mRNA-mediated Olig2 overexpression is a rational approach to promote oligodendrocyte differentiation and remyelination. Furthermore, when C-Olig2 was administered to a murine model of ischemic stroke, it led to improvements in blood‒brain barrier (BBB) integrity, enhanced remyelination, and rescued learning and cognitive deficits. Our comprehensive analysis, which included bulk RNA sequencing (RNA-seq) and single-nucleus RNA-seq (snRNA-seq), revealed upregulated biological processes related to learning and memory in the brains of mice treated with C-Olig2 compared to those receiving empty LNPs (Mock). Collectively, our findings highlight the therapeutic potential of multifunctional nanomedicine targeting mRNA expression for ischemic stroke and suggest that this approach holds promise for addressing various brain diseases. STATEMENT OF SIGNIFICANCE: While Olig2 overexpression in OPCs represents a promising therapeutic avenue for enhancing remyelination in ischemic stroke, in vivo strategies for achieving Olig2 expression pose considerable technological challenges. The delivery of mRNA via lipid nanoparticles is considered aa viable approach for in vivo protein expression. In this study, we engineered CD140a-targeted LNPs loaded with Olig2 mRNA (C-Olig2) with the aim of achieving specific Olig2 overexpression in mouse OPCs. Our findings demonstrate that C-Olig2 promotes the differentiation of OPCs into oligodendrocytes in vitro, providing evidence that mRNA-mediated Olig2 overexpression is a rational strategy to foster remyelination. Furthermore, the intravenous administration of C-Olig2 into a murine model of ischemic stroke not only improved blood-brain barrier integrity but also enhanced remyelination and mitigated learning and cognitive deficits. These results underscore the promising therapeutic potential of multifunctional nanomedicine targeting mRNA expression in the context of ischemic stroke.
Collapse
Affiliation(s)
- Jian Xu
- Stroke center, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 510799, China; Department of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China.
| | - Rui Wang
- Stroke center, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 510799, China; Clinical Research Institute, the First People's Hospital of Foshan, Foshan 528000, China
| | - Wei Luo
- Clinical Research Institute, the First People's Hospital of Foshan, Foshan 528000, China
| | - Xiaofan Mao
- Clinical Research Institute, the First People's Hospital of Foshan, Foshan 528000, China
| | - Hong Gao
- Department of Geriatrics, Institute of Translational Medicine, the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - Xinwei Feng
- Stroke center, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 510799, China
| | - Guoqiang Chen
- Department of General Medicine, the First People's Hospital of Foshan, Foshan 528000, China
| | - Zhihua Yang
- Stroke center, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 510799, China.
| | - Wenbin Deng
- Department of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China.
| | - Yichu Nie
- Department of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China; Clinical Research Institute, the First People's Hospital of Foshan, Foshan 528000, China.
| |
Collapse
|
4
|
Li Y, Lim C, Dismuke T, Malawsky DS, Oasa S, Bruce ZC, Offenhäuser C, Baumgartner U, D’Souza RCJ, Edwards SL, French JD, Ock LS, Nair S, Sivakumaran H, Harris L, Tikunov AP, Hwang D, Del Mar Alicea Pauneto C, Maybury M, Hassall T, Wainwright B, Kesari S, Stein G, Piper M, Johns TG, Sokolsky-Papkov M, Terenius L, Vukojević V, Gershon TR, Day BW. Preventing recurrence in Sonic Hedgehog Subgroup Medulloblastoma using the OLIG2 inhibitor CT-179. RESEARCH SQUARE 2023:rs.3.rs-2949436. [PMID: 37333134 PMCID: PMC10275055 DOI: 10.21203/rs.3.rs-2949436/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Recurrence is the primary life-threatening complication for medulloblastoma (MB). In Sonic Hedgehog (SHH)-subgroup MB, OLIG2-expressing tumor stem cells drive recurrence. We investigated the anti-tumor potential of the small-molecule OLIG2 inhibitor CT-179, using SHH-MB patient-derived organoids, patient-derived xenograft (PDX) tumors and mice genetically-engineered to develop SHH-MB. CT-179 disrupted OLIG2 dimerization, DNA binding and phosphorylation and altered tumor cell cycle kinetics in vitro and in vivo, increasing differentiation and apoptosis. CT-179 increased survival time in GEMM and PDX models of SHH-MB, and potentiated radiotherapy in both organoid and mouse models, delaying post-radiation recurrence. Single cell transcriptomic studies (scRNA-seq) confirmed that CT-179 increased differentiation and showed that tumors up-regulated Cdk4 post-treatment. Consistent with increased CDK4 mediating CT-179 resistance, CT-179 combined with CDK4/6 inhibitor palbociclib delayed recurrence compared to either single-agent. These data show that targeting treatment-resistant MB stem cell populations by adding the OLIG2 inhibitor CT-179 to initial MB treatment can reduce recurrence.
Collapse
Affiliation(s)
- Yuchen Li
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
- These authors contributed equally
- The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Chaemin Lim
- These authors contributed equally
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA
- College of Pharmacy, Chung-Ang University, 221 Heukseok-dong, Dongiak-gu, Seoul 06974, Republic of Korea
| | - Taylor Dismuke
- These authors contributed equally
- Department of Neurology, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| | - Daniel S. Malawsky
- Department of Neurology, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
- Wellcome Sanger Institute, Hinxton, Cambridgeshire, UK
| | - Sho Oasa
- Department of Clinical Neuroscience, Center for Molecular Medicine (CMM), Karolinska Institutet, 17176 Stockholm, Sweden
| | - Zara C. Bruce
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
| | | | - Ulrich Baumgartner
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
- The University of Queensland, Brisbane, QLD, 4072, Australia
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, 4072, Australia
| | - Rochelle C. J. D’Souza
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
- The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Stacey L. Edwards
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
| | - Juliet D. French
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
| | - Lucy S.H. Ock
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
| | - Sneha Nair
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
| | - Haran Sivakumaran
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
| | - Lachlan Harris
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
- The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Andrey P. Tikunov
- Department of Neurology, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
- Department of Pediatrics, Emory University, Atlanta, GA 30323, USA
| | - Duhyeong Hwang
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA
- Department of Pharmaceutical Engineering, Dankook University, 119 Dandae-ro, Dongnam-gu, Cheonan 31116, Republic of Korea
| | - Coral Del Mar Alicea Pauneto
- Department of Neurology, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| | - Mellissa Maybury
- Child Health Research Centre, The University of Queensland, Brisbane, QLD, 4101, Australia
| | - Timothy Hassall
- The University of Queensland, Brisbane, QLD, 4072, Australia
- Oncology Service, Queensland Children’s Hospital, Children’s Health Queensland Hospital & Health Service, Brisbane, QLD, 4101, Australia
| | | | - Santosh Kesari
- Curtana Pharmaceuticals, Inc. Austin, TX 78756, United States
| | | | - Michael Piper
- The University of Queensland, Brisbane, QLD, 4072, Australia
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, 4072, Australia
| | | | - Marina Sokolsky-Papkov
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Lars Terenius
- Department of Clinical Neuroscience, Center for Molecular Medicine (CMM), Karolinska Institutet, 17176 Stockholm, Sweden
| | - Vladana Vukojević
- Department of Clinical Neuroscience, Center for Molecular Medicine (CMM), Karolinska Institutet, 17176 Stockholm, Sweden
| | - Timothy R. Gershon
- Department of Neurology, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
- Department of Pediatrics, Emory University, Atlanta, GA 30323, USA
| | - Bryan W. Day
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
- The University of Queensland, Brisbane, QLD, 4072, Australia
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, 4072, Australia
- Lead contact
| |
Collapse
|
5
|
Szu JI, Tsigelny IF, Wojcinski A, Kesari S. Biological functions of the Olig gene family in brain cancer and therapeutic targeting. Front Neurosci 2023; 17:1129434. [PMID: 37274223 PMCID: PMC10232966 DOI: 10.3389/fnins.2023.1129434] [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: 12/21/2022] [Accepted: 04/26/2023] [Indexed: 06/06/2023] Open
Abstract
The Olig genes encode members of the basic helix-loop-helix (bHLH) family of transcription factors. Olig1, Olig2, and Olig3 are expressed in both the developing and mature central nervous system (CNS) and regulate cellular specification and differentiation. Over the past decade extensive studies have established functional roles of Olig1 and Olig2 in development as well as in cancer. Olig2 overexpression drives glioma proliferation and resistance to radiation and chemotherapy. In this review, we summarize the biological functions of the Olig family in brain cancer and how targeting Olig family genes may have therapeutic benefit.
Collapse
Affiliation(s)
- Jenny I. Szu
- Department of Translational Neurosciences, Providence Saint John’s Health Center, Saint John’s Cancer Institute, Santa Monica, CA, United States
| | - Igor F. Tsigelny
- San Diego Supercomputer Center, University of California, San Diego, San Diego, CA, United States
- CureScience, San Diego, CA, United States
| | - Alexander Wojcinski
- Department of Translational Neurosciences, Providence Saint John’s Health Center, Saint John’s Cancer Institute, Santa Monica, CA, United States
- Pacific Neuroscience Institute, Santa Monica, CA, United States
| | - Santosh Kesari
- Department of Translational Neurosciences, Providence Saint John’s Health Center, Saint John’s Cancer Institute, Santa Monica, CA, United States
- Pacific Neuroscience Institute, Santa Monica, CA, United States
| |
Collapse
|
6
|
Fang LP, Liu Q, Meyer E, Welle A, Huang W, Scheller A, Kirchhoff F, Bai X. A subset of OPCs do not express Olig2 during development which can be increased in the adult by brain injuries and complex motor learning. Glia 2023; 71:415-430. [PMID: 36308278 DOI: 10.1002/glia.24284] [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: 06/18/2022] [Revised: 09/29/2022] [Accepted: 10/07/2022] [Indexed: 11/08/2022]
Abstract
Oligodendrocyte precursor cells (OPCs) are uniformly distributed in the mammalian brain; however, their function is rather heterogeneous in respect to their origin, location, receptor/channel expression and age. The basic helix-loop-helix transcription factor Olig2 is expressed in all OPCs as a pivotal determinant of their differentiation. Here, we identified a subset (2%-26%) of OPCs lacking Olig2 in various brain regions including cortex, corpus callosum, CA1 and dentate gyrus. These Olig2 negative (Olig2neg ) OPCs were enriched in the juvenile brain and decreased subsequently with age, being rarely detectable in the adult brain. However, the loss of this population was not due to apoptosis or microglia-dependent phagocytosis. Unlike Olig2pos OPCs, these subset cells were rarely labeled for the mitotic marker Ki67. And, accordingly, BrdU was incorporated only by a three-day long-term labeling but not by a 2-hour short pulse, suggesting these cells do not proliferate any more but were derived from proliferating OPCs. The Olig2neg OPCs exhibited a less complex morphology than Olig2pos ones. Olig2neg OPCs preferentially remain in a precursor stage rather than differentiating into highly branched oligodendrocytes. Changing the adjacent brain environment, for example, by acute injuries or by complex motor learning tasks, stimulated the transition of Olig2pos OPCs to Olig2neg cells in the adult. Taken together, our results demonstrate that OPCs transiently suppress Olig2 upon changes of the brain activity.
Collapse
Affiliation(s)
- Li-Pao Fang
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, Homburg, Germany
| | - Qing Liu
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, Homburg, Germany
| | - Erika Meyer
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, Homburg, Germany.,Laboratory of Brain Ischemia and Neuroprotection, Department of Pharmacology and Therapeutics, State University of Maringá, Maringá, Brazil
| | - Anna Welle
- Department of Genetics and EpiGenetics, University of Saarland, Saarbrücken, Germany
| | - Wenhui Huang
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, Homburg, Germany
| | - Anja Scheller
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, Homburg, Germany
| | - Frank Kirchhoff
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, Homburg, Germany.,Experimental Research Center for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, Craiova
| | - Xianshu Bai
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, Homburg, Germany
| |
Collapse
|
7
|
Jana A, Wang X, Leasure JW, Magana L, Wang L, Kim YM, Dodiya H, Toth PT, Sisodia SS, Rehman J. Increased Type I interferon signaling and brain endothelial barrier dysfunction in an experimental model of Alzheimer's disease. Sci Rep 2022; 12:16488. [PMID: 36182964 PMCID: PMC9526723 DOI: 10.1038/s41598-022-20889-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 09/19/2022] [Indexed: 11/09/2022] Open
Abstract
Blood-brain barrier (BBB) dysfunction is emerging as a key pathogenic factor in the progression of Alzheimer's disease (AD), where increased microvascular endothelial permeability has been proposed to play an important role. However, the molecular mechanisms leading to increased brain microvascular permeability in AD are not fully understood. We studied brain endothelial permeability in female APPswe/PS1∆E9 (APP/PS1) mice which constitute a transgenic mouse model of amyloid-beta (Aβ) amyloidosis and found that permeability increases with aging in the areas showing the greatest amyloid plaque deposition. We performed an unbiased bulk RNA-sequencing analysis of brain endothelial cells (BECs) in female APP/PS1 transgenic mice. We observed that upregulation of interferon signaling gene expression pathways in BECs was among the most prominent transcriptomic signatures in the brain endothelium. Immunofluorescence analysis of isolated BECs from female APP/PS1 mice demonstrated higher levels of the Type I interferon-stimulated gene IFIT2. Immunoblotting of APP/PS1 BECs showed downregulation of the adherens junction protein VE-cadherin. Stimulation of human brain endothelial cells with interferon-β decreased the levels of the adherens junction protein VE-cadherin as well as tight junction proteins Occludin and Claudin-5 and increased barrier leakiness. Depletion of the Type I interferon receptor in human brain endothelial cells prevented interferon-β-induced VE-cadherin downregulation and restored endothelial barrier integrity. Our study suggests that Type I interferon signaling contributes to brain endothelial dysfunction in AD.
Collapse
Affiliation(s)
- Arundhati Jana
- Division of Cardiology, Department of Medicine, College of Medicine, University of Illinois, Chicago, IL, 60612, USA
| | - Xinge Wang
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, 60612, USA.,Department of Biochemistry and Molecular Genetics, University of Illinois, College of Medicine, Chicago, IL, 60607, USA
| | - Joseph W Leasure
- Department of Biochemistry and Molecular Genetics, University of Illinois, College of Medicine, Chicago, IL, 60607, USA
| | - Lissette Magana
- Department of Biochemistry and Molecular Genetics, University of Illinois, College of Medicine, Chicago, IL, 60607, USA
| | - Li Wang
- Division of Cardiology, Department of Medicine, College of Medicine, University of Illinois, Chicago, IL, 60612, USA.,Department of Biochemistry and Molecular Genetics, University of Illinois, College of Medicine, Chicago, IL, 60607, USA
| | - Young-Mee Kim
- Division of Cardiology, Department of Medicine, College of Medicine, University of Illinois, Chicago, IL, 60612, USA.,Department of Biochemistry and Molecular Genetics, University of Illinois, College of Medicine, Chicago, IL, 60607, USA
| | - Hemraj Dodiya
- Department of Neurobiology, University of Chicago, Chicago, IL, 60637, USA.,The Microbiome Center, University of Chicago, Chicago, IL, 60637, USA
| | - Peter T Toth
- Research Resources Center, University of Chicago, Chicago, IL, 60612, USA.,Department of Pharmacology and Regenerative Medicine, University of Chicago, Chicago, IL, 60612, USA
| | - Sangram S Sisodia
- Department of Neurobiology, University of Chicago, Chicago, IL, 60637, USA.,The Microbiome Center, University of Chicago, Chicago, IL, 60637, USA
| | - Jalees Rehman
- Division of Cardiology, Department of Medicine, College of Medicine, University of Illinois, Chicago, IL, 60612, USA. .,Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, 60612, USA. .,Department of Biochemistry and Molecular Genetics, University of Illinois, College of Medicine, Chicago, IL, 60607, USA. .,Department of Pharmacology and Regenerative Medicine, University of Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
8
|
Endogenous Neural Stem Cell Mediated Oligodendrogenesis in the Adult Mammalian Brain. Cells 2022; 11:cells11132101. [PMID: 35805185 PMCID: PMC9265817 DOI: 10.3390/cells11132101] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 06/28/2022] [Accepted: 06/30/2022] [Indexed: 02/08/2023] Open
Abstract
Oligodendrogenesis is essential for replacing worn-out oligodendrocytes, promoting myelin plasticity, and for myelin repair following a demyelinating injury in the adult mammalian brain. Neural stem cells are an important source of oligodendrocytes in the adult brain; however, there are considerable differences in oligodendrogenesis from neural stem cells residing in different areas of the adult brain. Amongst the distinct niches containing neural stem cells, the subventricular zone lining the lateral ventricles and the subgranular zone in the dentate gyrus of the hippocampus are considered the principle areas of adult neurogenesis. In addition to these areas, radial glia-like cells, which are the precursors of neural stem cells, are found in the lining of the third ventricle, where they are called tanycytes, and in the cerebellum, where they are called Bergmann glia. In this review, we will describe the contribution and regulation of each of these niches in adult oligodendrogenesis.
Collapse
|
9
|
Koppenhafer SL, Goss KL, Voigt E, Croushore E, Terry WW, Ostergaard J, Gordon PM, Gordon DJ. Inhibitor of DNA binding 2 (ID2) regulates the expression of developmental genes and tumorigenesis in ewing sarcoma. Oncogene 2022; 41:2873-2884. [PMID: 35422476 PMCID: PMC9107507 DOI: 10.1038/s41388-022-02310-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 03/29/2022] [Accepted: 03/31/2022] [Indexed: 11/24/2022]
Abstract
Sarcomas are difficult to treat and the therapy, even when effective, is associated with long-term and life-threatening side effects. In addition, the treatment regimens for many sarcomas, including Ewing sarcoma, rhabdomyosarcoma, and osteosarcoma, are relatively unchanged over the past two decades, indicating a critical lack of progress. Although differentiation-based therapies are used for the treatment of some cancers, the application of this approach to sarcomas has proven challenging. Here, using a CRISPR-mediated gene knockout approach, we show that Inhibitor of DNA Binding 2 (ID2) is a critical regulator of developmental-related genes and tumor growth in vitro and in vivo in Ewing sarcoma tumors. We also identified that homoharringtonine, which is an inhibitor of protein translation and FDA-approved for the treatment of leukemia, decreases the level of the ID2 protein and significantly reduces tumor growth and prolongs mouse survival in an Ewing sarcoma xenograft model. Furthermore, in addition to targeting ID2, homoharringtonine also reduces the protein levels of ID1 and ID3, which are additional members of the ID family of proteins with well-described roles in tumorigenesis, in multiple types of cancer. Overall, these results provide insight into developmental regulation in Ewing sarcoma tumors and identify a novel, therapeutic approach to target the ID family of proteins using an FDA-approved drug.
Collapse
Affiliation(s)
- Stacia L Koppenhafer
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, University of Iowa, Iowa City, IA, 52242, USA
| | - Kelli L Goss
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, University of Iowa, Iowa City, IA, 52242, USA
| | - Ellen Voigt
- Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Emma Croushore
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, University of Iowa, Iowa City, IA, 52242, USA
| | - William W Terry
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, University of Iowa, Iowa City, IA, 52242, USA
| | - Jason Ostergaard
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Peter M Gordon
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - David J Gordon
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, University of Iowa, Iowa City, IA, 52242, USA.
| |
Collapse
|
10
|
Enriched Environment Effects on Myelination of the Central Nervous System: Role of Glial Cells. Neural Plast 2022; 2022:5766993. [PMID: 35465398 PMCID: PMC9023233 DOI: 10.1155/2022/5766993] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 01/20/2022] [Accepted: 03/09/2022] [Indexed: 12/24/2022] Open
Abstract
Myelination is regulated by various glial cells in the central nervous system (CNS), including oligodendrocytes (OLs), microglia, and astrocytes. Myelination of the CNS requires the generation of functionally mature OLs from OPCs. OLs are the myelin-forming cells in the CNS. Microglia play both beneficial and detrimental roles during myelin damage and repair. Astrocyte is responsible for myelin formation and regeneration by direct interaction with oligodendrocyte lineage cells. These glial cells are influenced by experience-dependent activities such as environmental enrichment (EE). To date, there are few studies that have investigated the association between EE and glial cells. EE with a complex combination of sensorimotor, cognitive, and social stimulation has a significant effect on cognitive impairment and brain plasticity. Hence, one mechanism through EE improving cognitive function may rely on the mutual effect of EE and glial cells. The purpose of this paper is to review recent research into the efficacy of EE for myelination and glial cells at cellular and molecular levels and offers critical insights for future research directions of EE and the treatment of EE in cognitive impairment disease.
Collapse
|
11
|
Sanchez-Petidier M, Guerri C, Moreno-Manzano V. Toll-like receptors 2 and 4 differentially regulate the self-renewal and differentiation of spinal cord neural precursor cells. Stem Cell Res Ther 2022; 13:117. [PMID: 35314006 PMCID: PMC8935849 DOI: 10.1186/s13287-022-02798-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 03/07/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Toll-like receptors (TLRs) represent critical effectors in the host defense response against various pathogens; however, their known function during development has also highlighted a potential role in cell fate determination and neural differentiation. While glial cells and neural precursor cells (NPCs) of the spinal cord express both TLR2 and TLR4, their influence on self-renewal and cell differentiation remains incompletely described. METHODS TLR2, TLR4 knock-out and the wild type mice were employed for spinal cord tissue analysis and NPCs isolation at early post-natal stage. Sox2, FoxJ1 and Ki67 expression among others served to identify the undifferentiated and proliferative NPCs; GFAP, Olig2 and β-III-tubulin markers served to identify astrocytes, oligodendrocytes and neurons respectively after NPC spontaneous differentiation. Multiple comparisons were analyzed using one-way ANOVA, with appropriate corrections such as Tukey's post hoc tests used for comparisons. RESULTS We discovered that the deletion of TLR2 or TLR4 significantly reduced the number of Sox2-expressing NPCs in the neonatal mouse spinal cord. While TLR2-knockout NPCs displayed enhanced self-renewal, increased proliferation and apoptosis, and delayed neural differentiation, the absence of TLR4 promoted the neural differentiation of NPCs without affecting proliferation, producing long projecting neurons. TLR4 knock-out NPCs showed significantly higher expression of Neurogenin1, that would be involved in the activation of this neurogenic program by a ligand and microenvironment-independent mechanism. Interestingly, the absence of both TLR2 and TLR4, which induces also a significant reduction in the expression of TLR1, in NPCs impeded oligodendrocyte precursor cell maturation to a similar degree. CONCLUSIONS Our data suggest that Toll-like receptors are needed to maintain Sox2 positive neural progenitors in the spinal cord, however possess distinct regulatory roles in mouse neonatal spinal cord NPCs-while TLR2 and TLR4 play a similar role in oligodendrocytic differentiation, they differentially influence neural differentiation.
Collapse
Affiliation(s)
- Marina Sanchez-Petidier
- Neuronal and Tissue Regeneration Laboratory, Prince Felipe Research Institute, Valencia, Spain.,Neuropathology Laboratory, Prince Felipe Research Institute, Valencia, Spain
| | - Consuelo Guerri
- Neuropathology Laboratory, Prince Felipe Research Institute, Valencia, Spain.
| | - Victoria Moreno-Manzano
- Neuronal and Tissue Regeneration Laboratory, Prince Felipe Research Institute, Valencia, Spain.
| |
Collapse
|
12
|
The Effects of the Olig Family on the Regulation of Spinal Cord Development and Regeneration. Neurochem Res 2021; 46:2776-2782. [PMID: 34228233 DOI: 10.1007/s11064-021-03383-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 06/15/2021] [Accepted: 06/17/2021] [Indexed: 12/22/2022]
Abstract
Neurons and glial cells in the central nervous system (CNS) are generated from neuroepithelial cells in the ventricular zone that surrounds the embryonic neural tube. The proliferation and distinct differentiation of neural precursors occurs at certain stages and are regulated by a series of transcription factors leading to the generation of neuronal and glial cell subtypes. In this manuscript, we review the effects of the Olig family, namely, members Olig1, Olig2 and Olig3, on the distinct differentiation of glial and neuronal cells in the developing spinal cord and injured neural tissue.
Collapse
|
13
|
Yang LJ, Cui H. Olig2 knockdown alleviates hypoxic-ischemic brain damage in newborn rats. Histol Histopathol 2021; 36:675-684. [PMID: 34013967 DOI: 10.14670/hh-18-344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVES Neuronal damage is an important pathological mechanism in neonatal hypoxic-ischemic brain damage (HIBD). We found in our previous studies that oligodendrocyte transcription factor 2 (Olig2) downregulation was able to increase cell survival in the brain. However, the specific mechanism has yet to be clarified. METHODS Sprague-Dawley rats aged 3 d were randomly divided into three groups: the normal control group, the Olig2-RNAi group, and the RNAi-negative control group. The normal control group received no treatment, the Olig2-RNAi group received the Olig2 RNAi adenovirus, and the RNAi-negative control group was given the control adenovirus after the completion of the HIBD model. Infarct lesions and their volumes were observed by triphenyltetrazolium chloride (TTC) staining 3 d after the completion of the adenovirus local injection. The condition of the tissue was characterized by hematoxylin-eosin staining 7 d after the model was established, and cell viability was determined by azure methylene blue staining. Subcellular damage was analyzed by transmission electron microscopy. Rotarod analysis was performed to detect moving behavior ability and an MWM assay was conducted to evaluate the memory. RESULTS TTC staining showed a smaller brain injury area in the Olig2-RNAi group than in the RNAi-negative control group. Hematoxylin-eosin staining indicated the presence of severe cell injury in the hippocampal region after HIBD, which improved after Olig2 knockdown. Azure methylene blue staining and electron microscopy results suggested that the cells improved after Olig2 knockdown. The rats stayed longer on the rotating rod, and their latency in the water maze test was gradually shortened relative to that of the rats in the Olig2-RNAi negative control group. CONCLUSION Olig2 knockdown can promote the repair of hypoxic-ischemic brain damage in newborn rats.
Collapse
Affiliation(s)
- L J Yang
- Department of Pediatrics, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| | - H Cui
- Department of Pediatrics, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
14
|
Oligodendrocyte Development and Regenerative Therapeutics in Multiple Sclerosis. Life (Basel) 2021; 11:life11040327. [PMID: 33918664 PMCID: PMC8069894 DOI: 10.3390/life11040327] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/02/2021] [Accepted: 04/07/2021] [Indexed: 12/23/2022] Open
Abstract
Myelination by oligodendrocytes (OLs) is an important biological process essential for central nervous system (CNS) development and functions. Oligodendroglial lineage cells undergo several morphological and molecular changes at different stages of their lineage progression into myelinating OLs. The transition steps of the oligodendrocyte progenitor cells (OPCs) to myelinating oligodendrocytes are defined by a specific pattern of regulated gene expression, which is under the control of coordinated signaling pathways. Any abnormal development, loss or failure of oligodendrocytes to myelinate axons can lead to several neurodegenerative diseases like multiple sclerosis (MS). MS is characterized by inflammation and demyelination, and current treatments target only the immune component of the disease, but have little impact on remyelination. Recently, several pharmacological compounds enhancing remyelination have been identified and some of them are in clinical trials. Here, we will review the current knowledge on oligodendrocyte differentiation, myelination and remyelination. We will focus on MS as a pathological condition, the most common chronic inflammatory demyelinating disease of the CNS in young adults.
Collapse
|
15
|
Vancamp P, Butruille L, Demeneix BA, Remaud S. Thyroid Hormone and Neural Stem Cells: Repair Potential Following Brain and Spinal Cord Injury. Front Neurosci 2020; 14:875. [PMID: 32982671 PMCID: PMC7479247 DOI: 10.3389/fnins.2020.00875] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 07/28/2020] [Indexed: 12/22/2022] Open
Abstract
Neurodegenerative diseases are characterized by chronic neuronal and/or glial cell loss, while traumatic injury is often accompanied by the acute loss of both. Multipotent neural stem cells (NSCs) in the adult mammalian brain spontaneously proliferate, forming neuronal and glial progenitors that migrate toward lesion sites upon injury. However, they fail to replace neurons and glial cells due to molecular inhibition and the lack of pro-regenerative cues. A major challenge in regenerative biology therefore is to unveil signaling pathways that could override molecular brakes and boost endogenous repair. In physiological conditions, thyroid hormone (TH) acts on NSC commitment in the subventricular zone, and the subgranular zone, the two largest NSC niches in mammals, including humans. Here, we discuss whether TH could have beneficial actions in various pathological contexts too, by evaluating recent data obtained in mammalian models of multiple sclerosis (MS; loss of oligodendroglial cells), Alzheimer’s disease (loss of neuronal cells), stroke and spinal cord injury (neuroglial cell loss). So far, TH has shown promising effects as a stimulator of remyelination in MS models, while its role in NSC-mediated repair in other diseases remains elusive. Disentangling the spatiotemporal aspects of the injury-driven repair response as well as the molecular and cellular mechanisms by which TH acts, could unveil new ways to further exploit its pro-regenerative potential, while TH (ant)agonists with cell type-specific action could provide safer and more target-directed approaches that translate easier to clinical settings.
Collapse
Affiliation(s)
- Pieter Vancamp
- Laboratory Molecular Physiology and Adaptation, CNRS UMR 7221, Muséum National d'Histoire Naturelle, Department Adaptations of Life, Paris, France
| | - Lucile Butruille
- Laboratory Molecular Physiology and Adaptation, CNRS UMR 7221, Muséum National d'Histoire Naturelle, Department Adaptations of Life, Paris, France
| | - Barbara A Demeneix
- Laboratory Molecular Physiology and Adaptation, CNRS UMR 7221, Muséum National d'Histoire Naturelle, Department Adaptations of Life, Paris, France
| | - Sylvie Remaud
- Laboratory Molecular Physiology and Adaptation, CNRS UMR 7221, Muséum National d'Histoire Naturelle, Department Adaptations of Life, Paris, France
| |
Collapse
|
16
|
Flitsch LJ, Laupman KE, Brüstle O. Transcription Factor-Based Fate Specification and Forward Programming for Neural Regeneration. Front Cell Neurosci 2020; 14:121. [PMID: 32508594 PMCID: PMC7251072 DOI: 10.3389/fncel.2020.00121] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 04/14/2020] [Indexed: 12/11/2022] Open
Abstract
Traditionally, in vitro generation of donor cells for brain repair has been dominated by the application of extrinsic growth factors and morphogens. Recent advances in cell engineering strategies such as reprogramming of somatic cells into induced pluripotent stem cells and direct cell fate conversion have impressively demonstrated the feasibility to manipulate cell identities by the overexpression of cell fate-determining transcription factors. These strategies are now increasingly implemented for transcription factor-guided differentiation of neural precursors and forward programming of pluripotent stem cells toward specific neural subtypes. This review covers major achievements, pros and cons, as well as future prospects of transcription factor-based cell fate specification and the applicability of these approaches for the generation of donor cells for brain repair.
Collapse
Affiliation(s)
- Lea J Flitsch
- Institute of Reconstructive Neurobiology, Life & Brain Center, University of Bonn Medical Faculty and University Hospital Bonn, Bonn, Germany
| | - Karen E Laupman
- Institute of Reconstructive Neurobiology, Life & Brain Center, University of Bonn Medical Faculty and University Hospital Bonn, Bonn, Germany
| | - Oliver Brüstle
- Institute of Reconstructive Neurobiology, Life & Brain Center, University of Bonn Medical Faculty and University Hospital Bonn, Bonn, Germany
| |
Collapse
|
17
|
Gomez-Pinedo U, Cuevas JA, Benito-Martín MS, Moreno-Jiménez L, Esteban-Garcia N, Torre-Fuentes L, Matías-Guiu JA, Pytel V, Montero P, Matías-Guiu J. Vitamin D increases remyelination by promoting oligodendrocyte lineage differentiation. Brain Behav 2020; 10:e01498. [PMID: 31837111 PMCID: PMC6955836 DOI: 10.1002/brb3.1498] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 09/20/2019] [Accepted: 11/02/2019] [Indexed: 12/18/2022] Open
Abstract
INTRODUCTION Several experimental studies have suggested the potential remyelinating effects of vitamin D (VitD) supplements regardless of the presence of VitD deficiency. This study aims to analyze neurogenesis in a model of toxic demyelination in order to evaluate the effects of VitD on demyelination and remyelination. MATERIAL AND METHODS We used 24 male Wistar rats that had received surgical lesions to the corpus callosum and were injected with lysolecithin. Rats were divided into three groups: Group 1 included eight rats with lesions to the corpus callosum but not lysolecithin injections (sham group), group 2 included eight rats with lesions to the corpus callosum that were injected with lysolecithin (lysolecithin group), and group 3 included eight rats with lesions that were injected with lysolecithin and received VitD (VitD group). We analyzed neurogenesis both in the subventricular zone and at the lesion site. RESULTS Administration of VitD promotes the proliferation and differentiation of neural stem cells in the subventricular zone and the migration of these cells to the lesion site in the corpus callosum; these cells subsequently differentiate into oligodendrocyte lineage cells and produce myelin basic protein. This phenomenon was not caused by microglial activation, which was less marked in rats receiving VitD. Megalin expression did not increase at the lesion site, which suggests that VitD is internalized by other mechanisms. CONCLUSION Our results support the hypothesis that regardless of the presence of VitD deficiency, treatment with VitD may contribute to remyelination by promoting the proliferation of oligodendrocyte precursor cells.
Collapse
Affiliation(s)
- Ulises Gomez-Pinedo
- Neurobiology Laboratory, Department of Neurology, Institute of Neurosciences, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, Madrid, Spain
| | - Jesús Adriel Cuevas
- Neurobiology Laboratory, Department of Neurology, Institute of Neurosciences, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, Madrid, Spain
| | - María Soledad Benito-Martín
- Neurobiology Laboratory, Department of Neurology, Institute of Neurosciences, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, Madrid, Spain
| | - Lidia Moreno-Jiménez
- Neurobiology Laboratory, Department of Neurology, Institute of Neurosciences, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, Madrid, Spain
| | - Noelia Esteban-Garcia
- Neurobiology Laboratory, Department of Neurology, Institute of Neurosciences, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, Madrid, Spain
| | - Laura Torre-Fuentes
- Neurobiology Laboratory, Department of Neurology, Institute of Neurosciences, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, Madrid, Spain
| | - Jordi A Matías-Guiu
- Neurobiology Laboratory, Department of Neurology, Institute of Neurosciences, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, Madrid, Spain
| | - Vanesa Pytel
- Neurobiology Laboratory, Department of Neurology, Institute of Neurosciences, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, Madrid, Spain
| | - Paloma Montero
- Neurobiology Laboratory, Department of Neurology, Institute of Neurosciences, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, Madrid, Spain
| | - Jorge Matías-Guiu
- Neurobiology Laboratory, Department of Neurology, Institute of Neurosciences, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
18
|
Aberrant Oligodendrogenesis in Down Syndrome: Shift in Gliogenesis? Cells 2019; 8:cells8121591. [PMID: 31817891 PMCID: PMC6953000 DOI: 10.3390/cells8121591] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 11/28/2019] [Accepted: 12/04/2019] [Indexed: 12/25/2022] Open
Abstract
Down syndrome (DS), or trisomy 21, is the most prevalent chromosomal anomaly accounting for cognitive impairment and intellectual disability (ID). Neuropathological changes of DS brains are characterized by a reduction in the number of neurons and oligodendrocytes, accompanied by hypomyelination and astrogliosis. Recent studies mainly focused on neuronal development in DS, but underestimated the role of glial cells as pathogenic players. Aberrant or impaired differentiation within the oligodendroglial lineage and altered white matter functionality are thought to contribute to central nervous system (CNS) malformations. Given that white matter, comprised of oligodendrocytes and their myelin sheaths, is vital for higher brain function, gathering knowledge about pathways and modulators challenging oligodendrogenesis and cell lineages within DS is essential. This review article discusses to what degree DS-related effects on oligodendroglial cells have been described and presents collected evidence regarding induced cell-fate switches, thereby resulting in an enhanced generation of astrocytes. Moreover, alterations in white matter formation observed in mouse and human post-mortem brains are described. Finally, the rationale for a better understanding of pathways and modulators responsible for the glial cell imbalance as a possible source for future therapeutic interventions is given based on current experience on pro-oligodendroglial treatment approaches developed for demyelinating diseases, such as multiple sclerosis.
Collapse
|
19
|
Tiane A, Schepers M, Rombaut B, Hupperts R, Prickaerts J, Hellings N, van den Hove D, Vanmierlo T. From OPC to Oligodendrocyte: An Epigenetic Journey. Cells 2019; 8:E1236. [PMID: 31614602 PMCID: PMC6830107 DOI: 10.3390/cells8101236] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 10/08/2019] [Accepted: 10/10/2019] [Indexed: 12/15/2022] Open
Abstract
Oligodendrocytes provide metabolic and functional support to neuronal cells, rendering them key players in the functioning of the central nervous system. Oligodendrocytes need to be newly formed from a pool of oligodendrocyte precursor cells (OPCs). The differentiation of OPCs into mature and myelinating cells is a multistep process, tightly controlled by spatiotemporal activation and repression of specific growth and transcription factors. While oligodendrocyte turnover is rather slow under physiological conditions, a disruption in this balanced differentiation process, for example in case of a differentiation block, could have devastating consequences during ageing and in pathological conditions, such as multiple sclerosis. Over the recent years, increasing evidence has shown that epigenetic mechanisms, such as DNA methylation, histone modifications, and microRNAs, are major contributors to OPC differentiation. In this review, we discuss how these epigenetic mechanisms orchestrate and influence oligodendrocyte maturation. These insights are a crucial starting point for studies that aim to identify the contribution of epigenetics in demyelinating diseases and may thus provide new therapeutic targets to induce myelin repair in the long run.
Collapse
Affiliation(s)
- Assia Tiane
- Department of Immunology, Biomedical Research Institute, Hasselt University, Hasselt 3500, Belgium.
- Department Psychiatry and Neuropsychology, European Graduate School of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht 6200 MD, The Netherlands.
| | - Melissa Schepers
- Department of Immunology, Biomedical Research Institute, Hasselt University, Hasselt 3500, Belgium.
- Department Psychiatry and Neuropsychology, European Graduate School of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht 6200 MD, The Netherlands.
| | - Ben Rombaut
- Department of Immunology, Biomedical Research Institute, Hasselt University, Hasselt 3500, Belgium.
- Department Psychiatry and Neuropsychology, European Graduate School of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht 6200 MD, The Netherlands.
| | - Raymond Hupperts
- Department of Neurology, Zuyderland Medical Center, Sittard-Geleen 6130 MB, The Netherlands.
- Department Psychiatry and Neuropsychology, European Graduate School of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht 6200 MD, The Netherlands.
| | - Jos Prickaerts
- Department Psychiatry and Neuropsychology, European Graduate School of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht 6200 MD, The Netherlands.
| | - Niels Hellings
- Department of Immunology, Biomedical Research Institute, Hasselt University, Hasselt 3500, Belgium.
| | - Daniel van den Hove
- Department Psychiatry and Neuropsychology, European Graduate School of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht 6200 MD, The Netherlands.
- Department of Psychiatry, Psychosomatics and Psychotherapy, University of Wuerzburg, Wuerzburg 97080, Germany.
| | - Tim Vanmierlo
- Department of Immunology, Biomedical Research Institute, Hasselt University, Hasselt 3500, Belgium.
- Department Psychiatry and Neuropsychology, European Graduate School of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht 6200 MD, The Netherlands.
| |
Collapse
|
20
|
Tomas-Roig J, Havemann-Reinecke U. Gene expression signature in brain regions exposed to long-term psychosocial stress following acute challenge with cannabinoid drugs. Psychoneuroendocrinology 2019; 102:1-8. [PMID: 30476795 DOI: 10.1016/j.psyneuen.2018.11.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 11/15/2018] [Accepted: 11/15/2018] [Indexed: 12/13/2022]
Abstract
Repeated exposure to life stressors can overwhelm the body's capacity to restore homeostasis and result in severe negative consequences. Cannabinoid CB1 receptors are highly expressed in the Central Nervous System (CNS) and regulate both glucocorticoid signalling and neurotransmitter release. In rodents, WIN55212.2 is a full agonist at the cannabinoid receptor type-1, while Rimonabant is a potent and selective cannabinoid inverse agonist at this receptor. This study aims to investigate the effect of long-term psychosocial stress following acute challenge with cannabinoid drugs on gene expression in distinct brain regions; this is done by employing digital multiplexed gene expression analysis. We found that repeated stress increased cortical mRNA levels of dopamine receptor D2, while the expression of neuregulin-1 decreased in both the prefrontal cortex and cerebellum. Further, we found that the acute injection of the agonist WIN55212.2 reduced striatal levels of dopamine receptor D2, while the use of inverse agonist Rimonabant acted in the opposite direction. The analysis of the interaction between the drugs and repeated stress revealed that defeat mice treated with WIN55212.2 showed lower expression of a set of myelin-related genes, as did the expression of SRY-box 10 and dopamine receptors-D1 and -D2 in the prefrontal cortex when compared to vehicle. In addition, in the hippocampus of stressed mice treated with WIN55212.2, we found an elevated expression of oligodendrocyte transcription factor-1, -2 and zinc finger protein 488 when compared to vehicle. In comparison to vehicle, an increase in 2',3'-Cyclic nucleotide 3'-phosphodiesterase and oligodendrocyte transcription factor-1 occurred in the cerebellum of stressed animals treated with the agonist. Moreover, treatment with Rimonabant under the influence of stress induced an overexpression of a set of myelin-related genes in the prefrontal cortex when compared to WIN-treated animals. In conclusion, repeated stress interfered with the dopaminergic system in the prefrontal cortex. We demonstrated that the expression of dopamine receptor D2 in the striatum was mediated by the CB1 receptor. Stressed mice exposed to either WIN55212.2 or Rimonabant displayed pronounced deficits in CNS myelination. In addition, the pharmacological blockage of CB1 receptor in stressed mice deregulated the expression of dopamine receptors and might lead to dysfunctions in dopamine metabolism.
Collapse
Affiliation(s)
- J Tomas-Roig
- Dept. of Psychiatry and Psychotherapy, University of Göttingen, Germany; Center Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany; Girona Neuroimmunology and Multiple Sclerosis Unit (UNIEMTG), Dr. Josep Trueta University Hospital and Neurodegeneration and Neuroinflammation Research Group, Girona Biomedical Research Institute (IDIBGI), Spain.
| | - U Havemann-Reinecke
- Dept. of Psychiatry and Psychotherapy, University of Göttingen, Germany; Center Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
| |
Collapse
|
21
|
Remyelination promoting therapies in multiple sclerosis animal models: a systematic review and meta-analysis. Sci Rep 2019; 9:822. [PMID: 30696832 PMCID: PMC6351564 DOI: 10.1038/s41598-018-35734-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 10/30/2018] [Indexed: 12/16/2022] Open
Abstract
An unmet but urgent medical need is the development of myelin repair promoting therapies for Multiple Sclerosis (MS). Many such therapies have been pre-clinically tested using different models of toxic demyelination such as cuprizone, ethidium bromide, or lysolecithin and some of the therapies already entered clinical trials. However, keeping track on all these possible new therapies and their efficacy has become difficult with the increasing number of studies. In this study, we aimed at summarizing the current evidence on such therapies through a systematic review and at providing an estimate of the effects of tested interventions by a meta-analysis. We show that 88 different therapies have been pre-clinically tested for remyelination. 25 of them (28%) entered clinical trials. Our meta-analysis also identifies 16 promising therapies which did not enter a clinical trial for MS so far, among them Pigment epithelium-derived factor, Plateled derived growth factor, and Tocopherol derivate TFA-12.We also show that failure in bench to bedside translation from certain therapies may in part be attributable to poor study quality. By addressing these problems, clinical translation might be smoother and possibly animal numbers could be reduced.
Collapse
|
22
|
Chen S, Kumar N, Mao Z, Sitruk-Ware R, Brinton RD. Therapeutic progestin segesterone acetate promotes neurogenesis: implications for sustaining regeneration in female brain. Menopause 2018; 25:1138-1151. [PMID: 29846284 PMCID: PMC7731586 DOI: 10.1097/gme.0000000000001135] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Neurogenesis is the principal regenerative mechanism to sustain the plasticity potential in adult brains. Decreased neurogenesis parallels the cognition decline with aging, and has been suggested as a common hallmark in the progression of many neurodegeneration diseases. We previously reported that acute exposure to segesterone acetate (ST-1435; Nestorone), alone or in combination with 17β-estradiol (E2), increased human neural stem cells proliferation and survival both in vitro and in vivo. The present study expanded our previous findings to investigate the more clinical related chronic exposure in combination with E2 on the regenerative capacity of adult brain. METHODS To mimic the chronic contraception exposure in women, 3-month old female mice (n = 110) were treated with ST-1435, with or without co-administration of E2, for 4 weeks. Neural cell proliferation and survival, and oligodendrocyte generation were assessed. The involvement of insulin-like growth factor 1 signaling was studied. RESULTS Our results demonstrated that chronic ST-1435 and E2 alone or in combination increased neurogenesis by a comparable magnitude, with minimum to no antagonistic or additive effects between ST-1435 and E2. In addition, chronic exposure of ST-1435 or ST-1435 + E2 stimulated oligodendrocyte generation, indicating potential elevated myelination. Insulin-like growth factor-1 (IGF-1) and IGF-1 receptor (IGF-1R) were also up-regulated after chronic ST-1435 and E2 exposure, suggesting the involvement of IGF-1 signaling as the potential underlined regulatory pathway transducing ST-1435 effect. CONCLUSION These findings provide preclinical evidence and mechanistic insights for the development of ST-1435 as a neuroregenerative therapy to promote intrinsic regenerative capacity in female brains against aging and neurodegenerative disorders.
Collapse
Affiliation(s)
- Shuhua Chen
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ, USA
| | - Narendar Kumar
- Center for Biomedical Research, Population Council,, New York, NY, USA
| | - Zisu Mao
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ, USA
| | | | - Roberta Diaz Brinton
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ, USA
- Department of Pharmacology, University of Arizona, Tucson, AZ, USA
- Department of Neurology, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
23
|
Fauveau M, Wilmet B, Deboux C, Benardais K, Bachelin C, Temporão AC, Kerninon C, Nait Oumesmar B. SOX17 transcription factor negatively regulates oligodendrocyte precursor cell differentiation. Glia 2018; 66:2221-2232. [PMID: 30152028 DOI: 10.1002/glia.23483] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 05/16/2018] [Accepted: 06/08/2018] [Indexed: 11/08/2022]
Abstract
Oligodendrocyte development is a critical process timely and spatially regulated to ensure proper myelination of the central nervous system. HMG-box transcription factors are key regulators of oligodendrocyte lineage progression. Among these factors, Sox17 was previously identified as a positive regulator of oligodendrocyte development. However, the role of Sox17 in oligodendroglial cell lineage progression and differentiation is still poorly understood. To define the functional role of Sox17, we generated new transgenic mouse models with inducible overexpression of Sox17, specifically in oligodendroglial cells. Here, we report that gain of Sox17 function has no effect on oligodendrocyte progenitor cells (OPCs) specification. During early postnatal development, Sox17 overexpression increases the pool of OPCs at the expense of differentiated oligodendrocytes. However, the oligodendroglial cell population, OPC proliferation and apoptosis remained unchanged in Sox17 transgenic mice. RNA sequencing, quantitative RT-PCR and immunohistochemical analysis showed that Sox17 represses the expression of the major myelin genes, resulting in a severe CNS hypomyelination. Overall, our data highlight an unexpected role for Sox17 as a negative regulator of OPC differentiation and myelination, suggesting stage specific functions for this factor during oligodendroglial cell lineage progression.
Collapse
Affiliation(s)
- Melissa Fauveau
- Inserm U1127, Institut du Cerveau et de la Moelle Epinière, ICM, Paris, France.,Sorbonne Université UMR-S1127, Paris, France.,CNRS, UMR 7225, Paris, France
| | - Baptiste Wilmet
- Inserm U1127, Institut du Cerveau et de la Moelle Epinière, ICM, Paris, France.,Sorbonne Université UMR-S1127, Paris, France.,CNRS, UMR 7225, Paris, France
| | - Cyrille Deboux
- Inserm U1127, Institut du Cerveau et de la Moelle Epinière, ICM, Paris, France.,Sorbonne Université UMR-S1127, Paris, France.,CNRS, UMR 7225, Paris, France
| | - Karelle Benardais
- Inserm U1127, Institut du Cerveau et de la Moelle Epinière, ICM, Paris, France.,Sorbonne Université UMR-S1127, Paris, France.,CNRS, UMR 7225, Paris, France
| | - Corinne Bachelin
- Inserm U1127, Institut du Cerveau et de la Moelle Epinière, ICM, Paris, France.,Sorbonne Université UMR-S1127, Paris, France.,CNRS, UMR 7225, Paris, France
| | - Ana C Temporão
- Inserm U1127, Institut du Cerveau et de la Moelle Epinière, ICM, Paris, France.,Sorbonne Université UMR-S1127, Paris, France.,CNRS, UMR 7225, Paris, France
| | - Christophe Kerninon
- Inserm U1127, Institut du Cerveau et de la Moelle Epinière, ICM, Paris, France.,Sorbonne Université UMR-S1127, Paris, France.,CNRS, UMR 7225, Paris, France
| | - Brahim Nait Oumesmar
- Inserm U1127, Institut du Cerveau et de la Moelle Epinière, ICM, Paris, France.,Sorbonne Université UMR-S1127, Paris, France.,CNRS, UMR 7225, Paris, France
| |
Collapse
|
24
|
Therapeutic Potential of Pien Tze Huang on Experimental Autoimmune Encephalomyelitis Rat. J Immunol Res 2018; 2018:2952471. [PMID: 29682587 PMCID: PMC5848133 DOI: 10.1155/2018/2952471] [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: 09/08/2017] [Accepted: 12/31/2017] [Indexed: 01/31/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease of the central nervous system (CNS). There is still lack of commercially viable treatment currently. Pien Tze Huang (PZH), a traditional Chinese medicine, has been proved to have anti-inflammatory, neuroprotective, and immunoregulatory effects. This study investigated the possible therapeutic effects of PZH on experimental autoimmune encephalomyelitis (EAE) rats, a classic animal model of MS. Male Lewis rats were immunized with myelin basic protein (MBP) peptide to establish an EAE model and then treated with three doses of PZH. Clinical symptoms, organ coefficient, histopathological features, levels of proinflammatory cytokines, and chemokines as well as MBP and Olig2 were analyzed. The results indicated that PZH ameliorated the clinical severity of EAE rats. It also remarkably reduced inflammatory cell infiltration in the CNS of EAE rats. Furthermore, the levels of IL-17A, IL-23, CCL3, and CCL5 in serum and the CNS were significantly decreased; the p-P65 and p-STAT3 levels were also downregulated in the CNS, while MBP and Olig2 in the CNS of EAE rats had a distinct improvement after PZH treatment. In addition, PZH has no obvious toxicity at the concentration of 0.486 g/kg/d. This study demonstrated that PZH could be used to treat MS.
Collapse
|
25
|
Age-Dependent Decline in Fate Switch from NG2 Cells to Astrocytes After Olig2 Deletion. J Neurosci 2018; 38:2359-2371. [PMID: 29382710 DOI: 10.1523/jneurosci.0712-17.2018] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 12/31/2017] [Accepted: 01/23/2018] [Indexed: 01/25/2023] Open
Abstract
NG2 cells are a resident glial progenitor cell population that is uniformly distributed throughout the developing and mature mammalian CNS. Those in the postnatal CNS generate exclusively myelinating and non-myelinating oligodendrocytes and are thus equated with oligodendrocyte precursor cells. Prenatally, NG2 cells in the ventral gray matter of the forebrain generate protoplasmic astrocytes as well as oligodendrocytes. The fate conversion from NG2 cells into protoplasmic astrocytes is dependent on downregulation of the key oligodendrocyte transcription factor Olig2. We showed previously that constitutive deletion of Olig2 in NG2 cells converts NG2 cells in the neocortex into protoplasmic astrocytes at the expense of oligodendrocytes. In this study, we show that postnatal deletion of Olig2 caused NG2 cells in the neocortex but not in other gray matter regions to become protoplasmic astrocytes. However, NG2 cells in the neocortex became more resistant to astrocyte fate switch over the first 3 postnatal weeks. Fewer NG2 cells differentiated into astrocytes and did so with longer latency after Olig2 deletion at postnatal day 18 (P18) compared with deletion at P2. The high-mobility group transcription factor Sox10 was not downregulated for at least 1 month after Olig2 deletion at P18 despite an early transient upregulation of the astrocyte transcription factor NFIA. Furthermore, inhibiting cell proliferation in slice culture reduced astrocyte differentiation from Olig2-deleted perinatal NG2 cells, suggesting that cell division might facilitate nuclear reorganization needed for astrocyte transformation.SIGNIFICANCE STATEMENT NG2 cells are glial progenitor cells that retain a certain degree of lineage plasticity. In the normal postnatal neocortex, they generate mostly oligodendrocyte lineage cells. When the oligodendrocyte transcription factor Olig2 is deleted in NG2 cells in the neocortex, they switch their fate to protoplasmic astrocytes. However, the efficiency of the fate switch decreases with age over the first 3 postnatal weeks and is reduced when cell proliferation is inhibited. As the neocortex matures, sustained expression of the oligodendrocyte lineage-specific key transcription factor Sox10 becomes less dependent on Olig2. Together, our findings suggest a gradual stabilization of the oligodendrocyte lineage genes and loss of lineage plasticity during the first 3 weeks after birth, possibly due to nuclear reorganization.
Collapse
|
26
|
Adnani L, Han S, Li S, Mattar P, Schuurmans C. Mechanisms of Cortical Differentiation. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 336:223-320. [DOI: 10.1016/bs.ircmb.2017.07.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
27
|
Motavaf M, Sadeghizadeh M, Javan M. Attempts to Overcome Remyelination Failure: Toward Opening New Therapeutic Avenues for Multiple Sclerosis. Cell Mol Neurobiol 2017; 37:1335-1348. [PMID: 28224237 DOI: 10.1007/s10571-017-0472-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Accepted: 02/12/2017] [Indexed: 01/02/2023]
Abstract
Multiple sclerosis (MS) is a chronic immune-mediated disorder of the central nervous system that results in destruction of the myelin sheath wrapped around the axons and eventual axon degeneration. The disease is pathologically heterogeneous; however, perhaps its most frustrating aspect is the lack of efficient regenerative response for remyelination. Current treatment strategies are based on anti-inflammatory or immunomodulatory medications that have the potential to reduce the numbers of newly evolving lesions. However, therapies are still required that can repair already damaged myelin for which current treatments are not effective. A prerequisite for the development of such new treatments is understanding the reasons for insufficient endogenous repair. This review briefly summarizes the currently suggested causes of remyelination failure in MS and possible solutions.
Collapse
Affiliation(s)
- Mahsa Motavaf
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Islamic Republic of Iran
| | - Majid Sadeghizadeh
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Islamic Republic of Iran
| | - Mohammad Javan
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, P.O. Box 14115-331, Tehran, Islamic Republic of Iran.
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Islamic Republic of Iran.
| |
Collapse
|
28
|
Tian Z, Zhao Q, Biswas S, Deng W. Methods of reactivation and reprogramming of neural stem cells for neural repair. Methods 2017; 133:3-20. [PMID: 28864354 DOI: 10.1016/j.ymeth.2017.08.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 07/21/2017] [Accepted: 08/24/2017] [Indexed: 12/27/2022] Open
Abstract
Research on the biology of adult neural stem cells (NSCs) and induced NSCs (iNSCs), as well as NSC-based therapies for diseases in central nervous system (CNS) has started to generate the expectation that these cells may be used for treatments in CNS injuries or disorders. Recent technological progresses in both NSCs themselves and their derivatives have brought us closer to therapeutic applications. Adult neurogenesis presents in particular regions in mammal brain, known as neurogenic niches such as the dental gyrus (DG) in hippocampus and the subventricular zone (SVZ), within which adult NSCs usually stay for long periods out of the cell cycle, in G0. The reactivation of quiescent adult NSCs needs orchestrated interactions between the extrinsic stimulis from niches and the intrinsic factors involving transcription factors (TFs), signaling pathway, epigenetics, and metabolism to start an intracellular regulatory program, which promotes the quiescent NSCs exit G0 and reenter cell cycle. Extrinsic and intrinsic mechanisms that regulate adult NSCs are interconnected and feedback on one another. Since endogenous neurogenesis only happens in restricted regions and steadily fails with disease advances, interest has evolved to apply the iNSCs converted from somatic cells to treat CNS disorders, as is also promising and preferable. To overcome the limitation of viral-based reprogramming of iNSCs, bioactive small molecules (SM) have been explored to enhance the efficiency of iNSC reprogramming or even replace TFs, making the iNSCs more amenable to clinical application. Despite intense research efforts to translate the studies of adult and induced NSCs from the bench to bedside, vital troubles remain at several steps in these processes. In this review, we examine the present status, advancement, pitfalls, and potential of the two types of NSC technologies, focusing on each aspects of reactivation of quiescent adult NSC and reprogramming of iNSC from somatic cells, as well as on progresses in cell-based regenerative strategies for neural repair and criteria for successful therapeutic applications.
Collapse
Affiliation(s)
- Zuojun Tian
- Department of Neurology, The Institute of Guangzhou Respiratory Disease, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, PR China; Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, CA 95817, USA; Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA 95817, USA
| | - Qiuge Zhao
- Department of Neurology, The Institute of Guangzhou Respiratory Disease, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, PR China
| | - Sangita Biswas
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, CA 95817, USA; Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA 95817, USA.
| | - Wenbin Deng
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, CA 95817, USA; Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA 95817, USA.
| |
Collapse
|
29
|
Tan BT, Jiang L, Liu L, Yin Y, Luo ZRX, Long ZY, Li S, Yu LH, Wu YM, Liu Y. Local injection of Lenti-Olig2 at lesion site promotes functional recovery of spinal cord injury in rats. CNS Neurosci Ther 2017; 23:475-487. [PMID: 28452182 DOI: 10.1111/cns.12694] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 03/08/2017] [Accepted: 03/10/2017] [Indexed: 12/17/2022] Open
Abstract
AIMS Olig2 is one of the most critical factors during CNS development, which belongs to b-HLH transcription factor family. Previous reports have shown that Olig2 regulates the remyelination processes in CNS demyelination diseases models. However, the role of Olig2 in contusion spinal cord injury (SCI) and the possible therapeutic effects remain obscure. This study aims to investigate the effects of overexpression Olig2 by lentivirus on adult spinal cord injury rats. METHODS Lenti-Olig2 expression and control Lenti-eGFP vectors were prepared, and virus in a total of 5 μL (108 TU/mL) was locally injected into the injured spinal cord 1.5 mm rostral and caudal near the epicenter. Immunostaining, Western blot, electron microscopy, and CatWalk analyzes were employed to investigate the effects of Olig2 on spinal cord tissue repair and functional recovery. RESULTS Injection of Lenti-Olig2 significantly increased the number of oligodendrocytes lineage cells and enhanced myelination after SCI. More importantly, the introduction of Olig2 greatly improved hindlimb locomotor performances. Other oligodendrocyte-related transcription factors, which were downregulated or upregulated after injury, were reversed by Olig2 induction. CONCLUSIONS Our findings provided the evidence that overexpression Olig2 promotes myelination and locomotor recovery of contusion SCI, which gives us more understanding of Olig2 on spinal cord injury treatment.
Collapse
Affiliation(s)
- Bo-Tao Tan
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Research Institute of Surgery, Daping Hospital, The Third Military Medical University, Chongqing, China.,Department of Rehabilitation Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Long Jiang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Research Institute of Surgery, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Li Liu
- Department of Brain, The Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Ying Yin
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ze-Ru-Xin Luo
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zai-Yun Long
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Research Institute of Surgery, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Sen Li
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Research Institute of Surgery, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Le-Hua Yu
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ya-Min Wu
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Research Institute of Surgery, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Yuan Liu
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Research Institute of Surgery, Daping Hospital, The Third Military Medical University, Chongqing, China
| |
Collapse
|
30
|
Akkermann R, Beyer F, Küry P. Heterogeneous populations of neural stem cells contribute to myelin repair. Neural Regen Res 2017; 12:509-517. [PMID: 28553319 PMCID: PMC5436337 DOI: 10.4103/1673-5374.204999] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
As ingenious as nature's invention of myelin sheaths within the mammalian nervous system is, as fatal can be damage to this specialized lipid structure. Long-term loss of electrical insulation and of further supportive functions myelin provides to axons, as seen in demyelinating diseases such as multiple sclerosis (MS), leads to neurodegeneration and results in progressive disabilities. Multiple lines of evidence have demonstrated the increasing inability of oligodendrocyte precursor cells (OPCs) to replace lost oligodendrocytes (OLs) in order to restore lost myelin. Much research has been dedicated to reveal potential reasons for this regeneration deficit but despite promising approaches no remyelination-promoting drugs have successfully been developed yet. In addition to OPCs neural stem cells of the adult central nervous system also hold a high potential to generate myelinating OLs. There are at least two neural stem cell niches in the brain, the subventricular zone lining the lateral ventricles and the subgranular zone of the dentate gyrus, and an additional source of neural stem cells has been located in the central canal of the spinal cord. While a substantial body of literature has described their neurogenic capacity, still little is known about the oligodendrogenic potential of these cells, even if some animal studies have provided proof of their contribution to remyelination. In this review, we summarize and discuss these studies, taking into account the different niches, the heterogeneity within and between stem cell niches and present current strategies of how to promote stem cell-mediated myelin repair.
Collapse
Affiliation(s)
- Rainer Akkermann
- Neuroregeneration Laboratory, Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Felix Beyer
- Neuroregeneration Laboratory, Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Patrick Küry
- Neuroregeneration Laboratory, Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| |
Collapse
|
31
|
Dulamea AO. Role of Oligodendrocyte Dysfunction in Demyelination, Remyelination and Neurodegeneration in Multiple Sclerosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 958:91-127. [PMID: 28093710 DOI: 10.1007/978-3-319-47861-6_7] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Oligodendrocytes (OLs) are the myelinating cells of the central nervous system (CNS) during development and throughout adulthood. They result from a complex and well controlled process of activation, proliferation, migration and differentiation of oligodendrocyte progenitor cells (OPCs) from the germinative niches of the CNS. In multiple sclerosis (MS), the complex pathological process produces dysfunction and apoptosis of OLs leading to demyelination and neurodegeneration. This review attempts to describe the patterns of demyelination in MS, the steps involved in oligodendrogenesis and myelination in healthy CNS, the different pathways leading to OLs and myelin loss in MS, as well as principles involved in restoration of myelin sheaths. Environmental factors and their impact on OLs and pathological mechanisms of MS are also discussed. Finally, we will present evidence about the potential therapeutic targets in re-myelination processes that can be accessed in order to develop regenerative therapies for MS.
Collapse
Affiliation(s)
- Adriana Octaviana Dulamea
- Neurology Clinic, University of Medicine and Pharmacy "Carol Davila", Fundeni Clinical Institute, Building A, Neurology Clinic, Room 201, 022328, Bucharest, Romania.
| |
Collapse
|
32
|
Akkermann R, Jadasz JJ, Azim K, Küry P. Taking Advantage of Nature's Gift: Can Endogenous Neural Stem Cells Improve Myelin Regeneration? Int J Mol Sci 2016; 17:ijms17111895. [PMID: 27854261 PMCID: PMC5133894 DOI: 10.3390/ijms17111895] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 10/28/2016] [Accepted: 11/09/2016] [Indexed: 01/18/2023] Open
Abstract
Irreversible functional deficits in multiple sclerosis (MS) are directly correlated to axonal damage and loss. Neurodegeneration results from immune-mediated destruction of myelin sheaths and subsequent axonal demyelination. Importantly, oligodendrocytes, the myelinating glial cells of the central nervous system, can be replaced to some extent to generate new myelin sheaths. This endogenous regeneration capacity has so far mainly been attributed to the activation and recruitment of resident oligodendroglial precursor cells. As this self-repair process is limited and increasingly fails while MS progresses, much interest has evolved regarding the development of remyelination-promoting strategies and the presence of alternative cell types, which can also contribute to the restoration of myelin sheaths. The adult brain comprises at least two neurogenic niches harboring life-long adult neural stem cells (NSCs). An increasing number of investigations are beginning to shed light on these cells under pathological conditions and revealed a significant potential of NSCs to contribute to myelin repair activities. In this review, these emerging investigations are discussed with respect to the importance of stimulating endogenous repair mechanisms from germinal sources. Moreover, we present key findings of NSC-derived oligodendroglial progeny, including a comprehensive overview of factors and mechanisms involved in this process.
Collapse
Affiliation(s)
- Rainer Akkermann
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, 40225 Düsseldorf, Germany.
| | - Janusz Joachim Jadasz
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, 40225 Düsseldorf, Germany.
| | - Kasum Azim
- Focus Translational Neuroscience, Institute of Physiological Chemistry, University of Mainz, 55122 Mainz, Germany.
| | - Patrick Küry
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, 40225 Düsseldorf, Germany.
| |
Collapse
|
33
|
Yu YH, Narayanan G, Sankaran S, Ramasamy S, Chan SY, Lin S, Chen J, Yang H, Srivats H, Ahmed S. Purification, Visualization, and Molecular Signature of Neural Stem Cells. Stem Cells Dev 2015; 25:189-201. [PMID: 26464067 PMCID: PMC4770853 DOI: 10.1089/scd.2015.0190] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Neural stem cells (NSCs) are isolated from primary brain tissue and propagated as a heterogeneous mix of cells, including neural progenitors. To date, NSCs have not been purified in vitro to allow study of their biology and utility in regenerative medicine. In this study, we identify C1qR1 as a novel marker for NSCs and show that it can be used along with Lewis-X (LeX) to yield a highly purified population of NSCs. Using time-lapse microscopy, we are able to follow NSCs forming neurospheres, allowing their visualization. Finally, using single-cell polymerase chain reaction (PCR), we determine the molecular signature of NSCs. The single-cell PCR data suggest that along with the Notch and Shh pathways, the Hippo pathway plays an important role in NSC activity.
Collapse
Affiliation(s)
- Yuan Hong Yu
- 1 Neural Stem Cell Laboratory, Institute of Medical Biology , Singapore, Singapore
| | - Gunaseelan Narayanan
- 1 Neural Stem Cell Laboratory, Institute of Medical Biology , Singapore, Singapore
| | - Shvetha Sankaran
- 1 Neural Stem Cell Laboratory, Institute of Medical Biology , Singapore, Singapore
| | - Srinivas Ramasamy
- 1 Neural Stem Cell Laboratory, Institute of Medical Biology , Singapore, Singapore
| | - Shi Yu Chan
- 1 Neural Stem Cell Laboratory, Institute of Medical Biology , Singapore, Singapore
| | - Shuping Lin
- 1 Neural Stem Cell Laboratory, Institute of Medical Biology , Singapore, Singapore
| | - Jinmiao Chen
- 2 Bioinformatics Laboratory , Singapore Immunology Network, Singapore, Singapore
| | - Henry Yang
- 2 Bioinformatics Laboratory , Singapore Immunology Network, Singapore, Singapore
| | - Hariharan Srivats
- 1 Neural Stem Cell Laboratory, Institute of Medical Biology , Singapore, Singapore
| | - Sohail Ahmed
- 1 Neural Stem Cell Laboratory, Institute of Medical Biology , Singapore, Singapore
| |
Collapse
|
34
|
Li N, Leung GKK. Oligodendrocyte Precursor Cells in Spinal Cord Injury: A Review and Update. BIOMED RESEARCH INTERNATIONAL 2015; 2015:235195. [PMID: 26491661 PMCID: PMC4600489 DOI: 10.1155/2015/235195] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 06/19/2015] [Accepted: 06/25/2015] [Indexed: 12/20/2022]
Abstract
Spinal cord injury (SCI) is a devastating condition to individuals, families, and society. Oligodendrocyte loss and demyelination contribute as major pathological processes of secondary damages after injury. Oligodendrocyte precursor cells (OPCs), a subpopulation that accounts for 5 to 8% of cells within the central nervous system, are potential sources of oligodendrocyte replacement after SCI. OPCs react rapidly to injuries, proliferate at a high rate, and can differentiate into myelinating oligodendrocytes. However, posttraumatic endogenous remyelination is rarely complete, and a better understanding of OPCs' characteristics and their manipulations is critical to the development of novel therapies. In this review, we summarize known characteristics of OPCs and relevant regulative factors in both health and demyelinating disorders including SCI. More importantly, we highlight current evidence on post-SCI OPCs transplantation as a potential treatment option as well as the impediments against regeneration. Our aim is to shed lights on important knowledge gaps and to provoke thoughts for further researches and the development of therapeutic strategies.
Collapse
Affiliation(s)
- Ning Li
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong
| | - Gilberto K. K. Leung
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong
| |
Collapse
|
35
|
Küspert M, Wegner M. SomethiNG 2 talk about-Transcriptional regulation in embryonic and adult oligodendrocyte precursors. Brain Res 2015; 1638:167-182. [PMID: 26232072 DOI: 10.1016/j.brainres.2015.07.024] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 07/14/2015] [Accepted: 07/18/2015] [Indexed: 12/26/2022]
Abstract
Glial cells that express the chondroitin sulfate proteoglycan NG2 represent an inherently heterogeneous population. These so-called NG2-glia are present during development and in the adult CNS, where they are referred to as embryonic oligodendrocyte precursors and adult NG2-glia, respectively. They give rise to myelinating oligodendrocytes at all times of life. Over the years much has been learnt about the transcriptional network in embryonic oligodendrocyte precursors, and several transcription factors from the HLH, HMG-domain, zinc finger and homeodomain protein families have been identified as main constituents. Much less is known about the corresponding network in adult NG2-glia. Here we summarize and discuss current knowledge on functions of each of these transcription factor families in NG2-glia, and where possible compare transcriptional regulation in embryonic oligodendrocyte precursors and adult NG2-glia. This article is part of a Special Issue entitled SI:NG2-glia (Invited only).
Collapse
Affiliation(s)
- Melanie Küspert
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Fahrstrasse 17, Erlangen D-91054, Germany.
| | - Michael Wegner
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Fahrstrasse 17, Erlangen D-91054, Germany.
| |
Collapse
|
36
|
Wegener A, Deboux C, Bachelin C, Frah M, Kerninon C, Seilhean D, Weider M, Wegner M, Nait-Oumesmar B. Gain of Olig2 function in oligodendrocyte progenitors promotes remyelination. ACTA ACUST UNITED AC 2015; 138:120-35. [PMID: 25564492 DOI: 10.1093/brain/awu375] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The basic helix-loop-helix transcription factor Olig2 is a key determinant for the specification of neural precursor cells into oligodendrocyte progenitor cells. However, the functional role of Olig2 in oligodendrocyte migration and differentiation remains elusive both during developmental myelination and under demyelinating conditions of the adult central nervous system. To decipher Olig2 functions, we generated transgenic mice (TetOlig2:Sox10(rtTA/+)) overexpressing Olig2 in Sox10(+) oligodendroglial cells in a doxycycline inducible manner. We show that Olig2 overexpression increases the generation of differentiated oligodendrocytes, leading to precocious myelination of the central nervous system. Unexpectedly, we found that gain of Olig2 function in oligodendrocyte progenitor cells enhances their migration rate. To determine whether Olig2 overexpression in adult oligodendrocyte progenitor cells promotes oligodendrocyte regeneration for myelin repair, we induced lysophosphatidylcholine demyelination in the corpus callosum of TetOlig2:Sox10(rtTA/+) and control mice. We found that Olig2 overexpression enhanced oligodendrocyte progenitor cell differentiation and remyelination. To assess the relevance of these findings in demyelinating diseases, we also examined OLIG2 expression in multiple sclerosis lesions. We demonstrate that OLIG2 displays a differential expression pattern in multiple sclerosis lesions that correlates with lesion activity. Strikingly, OLIG2 was predominantly detected in NOGO-A(+) (now known as RTN4-A) maturing oligodendrocytes, which prevailed in active lesion borders, rather than chronic silent and shadow plaques. Taken together, our data provide proof of principle indicating that OLIG2 overexpression in oligodendrocyte progenitor cells might be a possible therapeutic mechanism for enhancing myelin repair.
Collapse
Affiliation(s)
- Amélie Wegener
- 1 Inserm, U 1127, F-75013, Paris, France 1 Inserm, U 1127, F-75013, Paris, France 1 Inserm, U 1127, F-75013, Paris, France 1 Inserm, U 1127, F-75013, Paris, France 1 Inserm, U 1127, F-75013, Paris, France
| | - Cyrille Deboux
- 1 Inserm, U 1127, F-75013, Paris, France 1 Inserm, U 1127, F-75013, Paris, France 1 Inserm, U 1127, F-75013, Paris, France 1 Inserm, U 1127, F-75013, Paris, France
| | - Corinne Bachelin
- 1 Inserm, U 1127, F-75013, Paris, France 1 Inserm, U 1127, F-75013, Paris, France 1 Inserm, U 1127, F-75013, Paris, France 1 Inserm, U 1127, F-75013, Paris, France
| | - Magali Frah
- 1 Inserm, U 1127, F-75013, Paris, France 1 Inserm, U 1127, F-75013, Paris, France 1 Inserm, U 1127, F-75013, Paris, France 1 Inserm, U 1127, F-75013, Paris, France
| | - Christophe Kerninon
- 1 Inserm, U 1127, F-75013, Paris, France 1 Inserm, U 1127, F-75013, Paris, France 1 Inserm, U 1127, F-75013, Paris, France 1 Inserm, U 1127, F-75013, Paris, France
| | - Danielle Seilhean
- 1 Inserm, U 1127, F-75013, Paris, France 1 Inserm, U 1127, F-75013, Paris, France 1 Inserm, U 1127, F-75013, Paris, France 1 Inserm, U 1127, F-75013, Paris, France 1 Inserm, U 1127, F-75013, Paris, France
| | | | | | - Brahim Nait-Oumesmar
- 1 Inserm, U 1127, F-75013, Paris, France 1 Inserm, U 1127, F-75013, Paris, France 1 Inserm, U 1127, F-75013, Paris, France 1 Inserm, U 1127, F-75013, Paris, France 1 Inserm, U 1127, F-75013, Paris, France
| |
Collapse
|
37
|
Murcia-Belmonte V, Esteban PF, Martínez-Hernández J, Gruart A, Luján R, Delgado-García JM, de Castro F. Anosmin-1 over-expression regulates oligodendrocyte precursor cell proliferation, migration and myelin sheath thickness. Brain Struct Funct 2015; 221:1365-85. [PMID: 25662897 DOI: 10.1007/s00429-014-0977-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 12/22/2014] [Indexed: 12/11/2022]
Abstract
During development of the central nervous system, anosmin-1 (A1) works as a chemotropic cue contributing to axonal outgrowth and collateralization, as well as modulating the migration of different cell types, fibroblast growth factor receptor 1 (FGFR1) being the main receptor involved in all these events. To further understand the role of A1 during development, we have analysed the over-expression of human A1 in a transgenic mouse line. Compared with control mice during development and in early adulthood, A1 over-expressing transgenic mice showed an enhanced oligodendrocyte precursor cell (OPC) proliferation and a higher number of OPCs in the subventricular zone and in the corpus callosum (CC). The migratory capacity of OPCs from the transgenic mice is increased in vitro due to a higher basal activation of ERK1/2 mediated through FGFR1 and they also produced more myelin basic protein (MBP). In vivo, the over-expression of A1 resulted in an elevated number of mature oligodendrocytes with higher levels of MBP mRNA and protein, as well as increased levels of activation of the ERK1/2 proteins, while electron microscopy revealed thicker myelin sheaths around the axons of the CC in adulthood. Also in the mature CC, the nodes of Ranvier were significantly longer and the conduction velocity of the nerve impulse in vivo was significantly increased in the CC of A1 over-expressing transgenic mice. Altogether, these data confirmed the involvement of A1 in oligodendrogliogenesis and its relevance for myelination.
Collapse
Affiliation(s)
- Verónica Murcia-Belmonte
- Grupo de Neurobiología del Desarrollo-GNDe, Hospital Nacional de Parapléjicos, Finca La Peraleda, s/n, 45071, Toledo, Spain.,Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Campus San Juan de Alicante, 03550, Alicante, Spain
| | - Pedro F Esteban
- Grupo de Neurobiología del Desarrollo-GNDe, Hospital Nacional de Parapléjicos, Finca La Peraleda, s/n, 45071, Toledo, Spain
| | - José Martínez-Hernández
- Departamento de Ciencias Médicas, CRIB-Facultad de Medicina, Universidad de Castilla-La Mancha, C/Almansa 14, 02006, Albacete, Spain
| | - Agnès Gruart
- División de Neurociencias, Universidad Pablo de Olavide, Ctra. De Utrera, Km.1, 41013, Seville, Spain
| | - Rafael Luján
- Departamento de Ciencias Médicas, CRIB-Facultad de Medicina, Universidad de Castilla-La Mancha, C/Almansa 14, 02006, Albacete, Spain
| | - José María Delgado-García
- División de Neurociencias, Universidad Pablo de Olavide, Ctra. De Utrera, Km.1, 41013, Seville, Spain
| | - Fernando de Castro
- Grupo de Neurobiología del Desarrollo-GNDe, Hospital Nacional de Parapléjicos, Finca La Peraleda, s/n, 45071, Toledo, Spain.
| |
Collapse
|
38
|
Morell M, Tsan YC, O'Shea KS. Inducible expression of noggin selectively expands neural progenitors in the adult SVZ. Stem Cell Res 2015; 14:79-94. [DOI: 10.1016/j.scr.2014.11.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Revised: 10/09/2014] [Accepted: 11/06/2014] [Indexed: 12/29/2022] Open
|
39
|
Tan B, Yu J, Yin Y, Jia G, Jiang W, Yu L. The Olig family affects central nervous system development and disease. Neural Regen Res 2014; 9:329-36. [PMID: 25206819 PMCID: PMC4146145 DOI: 10.4103/1673-5374.128232] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/15/2013] [Indexed: 11/04/2022] Open
Abstract
Neural cell differentiation and maturation is a critical step during central nervous system development. The oligodendrocyte transcription family (Olig family) is known to be an important factor in regulating neural cell differentiation. Because of this, the Olig family also affects acute and chronic central nervous system diseases, including brain injury, multiple sclerosis, and even gliomas. Improved understanding about the functions of the Olig family in central nervous system development and disease will greatly aid novel breakthroughs in central nervous system diseases. This review investigates the role of the Olig family in central nervous system development and related diseases.
Collapse
Affiliation(s)
- Botao Tan
- Department of Rehabilitation Medicine, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Jing Yu
- Department of Rehabilitation Medicine, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Ying Yin
- Department of Rehabilitation Medicine, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Gongwei Jia
- Department of Rehabilitation Medicine, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Wei Jiang
- Department of Rehabilitation Medicine, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Lehua Yu
- Department of Rehabilitation Medicine, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| |
Collapse
|
40
|
Torres LH, Annoni R, Balestrin NT, Coleto PL, Duro SO, Garcia RCT, Pacheco-Neto M, Mauad T, Camarini R, Britto LRG, Marcourakis T. Environmental tobacco smoke in the early postnatal period induces impairment in brain myelination. Arch Toxicol 2014; 89:2051-8. [PMID: 25182420 DOI: 10.1007/s00204-014-1343-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 08/14/2014] [Indexed: 02/01/2023]
Abstract
Environmental tobacco smoke (ETS) is associated with high morbidity and mortality, mainly in children. However, few studies focus on the brain development effects of ETS exposure. Myelination mainly occurs in the early years of life in humans and the first three postnatal weeks in rodents and is sensitive to xenobiotics exposure. This study investigated the effects of early postnatal ETS exposure on myelination. BALB/c mice were exposed to ETS generated from 3R4F reference research cigarettes from the third to the fourteenth days of life. The myelination of nerve fibers in the optic nerve by morphometric analysis and the levels of Olig1 and myelin basic protein (MBP) were evaluated in the cerebellum, diencephalon, telencephalon, and brainstem in infancy, adolescence, and adulthood. Infant mice exposed to ETS showed a decrease in the percentage of myelinated fibers in the optic nerve, compared with controls. ETS induced a decrease in Olig1 protein levels in the cerebellum and brainstem and an increase in MBP levels in the cerebellum at infant. It was also found a decrease in MBP levels in the telencephalon and brainstem at adolescence and in the cerebellum and diencephalon at adulthood. The present study demonstrates that exposure to ETS, in a critical phase of development, affects the percentage of myelinated fibers and myelin-specific proteins in infant mice. Although we did not observe differences in the morphological analysis in adolescence and adulthood, there was a decrease in MBP levels in distinctive brain regions suggesting a delayed effect in adolescence and adulthood.
Collapse
Affiliation(s)
- Larissa H Torres
- Department of Clinical and Toxicological Analysis, Faculdade de Ciências Farmacêuticas, School of Pharmaceutical Sciences, University of São Paulo, Av Prof Lineu Prestes, 580 Bl 13B, São Paulo, SP, CEP 05508-000, Brazil
| | - Raquel Annoni
- Department of Pathology, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Natalia T Balestrin
- Department of Clinical and Toxicological Analysis, Faculdade de Ciências Farmacêuticas, School of Pharmaceutical Sciences, University of São Paulo, Av Prof Lineu Prestes, 580 Bl 13B, São Paulo, SP, CEP 05508-000, Brazil
| | - Priscila L Coleto
- Department of Clinical and Toxicological Analysis, Faculdade de Ciências Farmacêuticas, School of Pharmaceutical Sciences, University of São Paulo, Av Prof Lineu Prestes, 580 Bl 13B, São Paulo, SP, CEP 05508-000, Brazil
| | - Stephanie O Duro
- Department of Clinical and Toxicological Analysis, Faculdade de Ciências Farmacêuticas, School of Pharmaceutical Sciences, University of São Paulo, Av Prof Lineu Prestes, 580 Bl 13B, São Paulo, SP, CEP 05508-000, Brazil
| | - Raphael C T Garcia
- Department of Clinical and Toxicological Analysis, Faculdade de Ciências Farmacêuticas, School of Pharmaceutical Sciences, University of São Paulo, Av Prof Lineu Prestes, 580 Bl 13B, São Paulo, SP, CEP 05508-000, Brazil
| | - Maurílio Pacheco-Neto
- Department of Clinical Pathology, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Thais Mauad
- Department of Pathology, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Rosana Camarini
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Luiz R G Britto
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Tania Marcourakis
- Department of Clinical and Toxicological Analysis, Faculdade de Ciências Farmacêuticas, School of Pharmaceutical Sciences, University of São Paulo, Av Prof Lineu Prestes, 580 Bl 13B, São Paulo, SP, CEP 05508-000, Brazil.
| |
Collapse
|
41
|
Mitew S, Hay C, Peckham H, Xiao J, Koenning M, Emery B. Mechanisms regulating the development of oligodendrocytes and central nervous system myelin. Neuroscience 2014; 276:29-47. [DOI: 10.1016/j.neuroscience.2013.11.029] [Citation(s) in RCA: 154] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 11/13/2013] [Accepted: 11/14/2013] [Indexed: 12/29/2022]
|
42
|
El-Etr M, Rame M, Boucher C, Ghoumari AM, Kumar N, Liere P, Pianos A, Schumacher M, Sitruk-Ware R. Progesterone and nestorone promote myelin regeneration in chronic demyelinating lesions of corpus callosum and cerebral cortex. Glia 2014; 63:104-17. [PMID: 25092805 DOI: 10.1002/glia.22736] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Accepted: 07/21/2014] [Indexed: 01/09/2023]
Abstract
Multiple Sclerosis affects mainly women and consists in intermittent or chronic damages to the myelin sheaths, focal inflammation, and axonal degeneration. Current therapies are limited to immunomodulators and antiinflammatory drugs, but there is no efficient treatment for stimulating the endogenous capacity of myelin repair. Progesterone and synthetic progestins have been shown in animal models of demyelination to attenuate myelin loss, reduce clinical symptoms severity, modulate inflammatory responses and partially reverse the age-dependent decline in remyelination. Moreover, progesterone has been demonstrated to promote myelin formation in organotypic cultures of cerebellar slices. In the present study, we show that progesterone and the synthetic 19-nor-progesterone derivative Nestorone® promote the repair of severe chronic demyelinating lesions induced by feeding cuprizone to female mice for up to 12 weeks. Progesterone and Nestorone increase the density of NG2(+) oligodendrocyte progenitor cells and CA II(+) mature oligodendrocytes and enhance the formation of myelin basic protein (MBP)- and proteolipid protein (PLP)-immunoreactive myelin. However, while demyelination in response to cuprizone was less marked in corpus callosum than in cerebral cortex, remyelination appeared earlier in the former. The remyelinating effect of progesterone was progesterone receptor (PR)-dependent, as it was absent in PR-knockout mice. Progesterone and Nestorone also decreased (but did not suppress) neuroinflammatory responses, specifically astrocyte and microglial cell activation. Therefore, some progestogens are promising therapeutic candidates for promoting the regeneration of myelin.
Collapse
Affiliation(s)
- Martine El-Etr
- UMR 788 Inserm and University Paris-Sud, 94276, Kremlin-Bicêtre, France
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Transcription factor induction of human oligodendrocyte progenitor fate and differentiation. Proc Natl Acad Sci U S A 2014; 111:E2885-94. [PMID: 24982138 DOI: 10.1073/pnas.1408295111] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Human oligodendrocyte progenitor cell (OPC) specification and differentiation occurs slowly and limits the potential for cell-based treatment of demyelinating disease. In this study, using FACS-based isolation and microarray analysis, we identified a set of transcription factors expressed by human primary CD140a(+)O4(+) OPCs relative to CD133(+)CD140a(-) neural stem/progenitor cells (NPCs). Among these, lentiviral overexpression of transcription factors ASCL1, SOX10, and NKX2.2 in NPCs was sufficient to induce Sox10 enhancer activity, OPC mRNA, and protein expression consistent with OPC fate; however, unlike ASCL1 and NKX2.2, only the transcriptome of SOX10-infected NPCs was induced to a human OPC gene expression signature. Furthermore, only SOX10 promoted oligodendrocyte commitment, and did so at quantitatively equivalent levels to native OPCs. In xenografts of shiverer/rag2 animals, SOX10 increased the rate of mature oligodendrocyte differentiation and axon ensheathment. Thus, SOX10 appears to be the principle and rate-limiting regulator of myelinogenic fate from human NPCs.
Collapse
|
44
|
El Waly B, Macchi M, Cayre M, Durbec P. Oligodendrogenesis in the normal and pathological central nervous system. Front Neurosci 2014; 8:145. [PMID: 24971048 PMCID: PMC4054666 DOI: 10.3389/fnins.2014.00145] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Accepted: 05/23/2014] [Indexed: 12/26/2022] Open
Abstract
Oligodendrocytes (OLGs) are generated late in development and myelination is thus a tardive event in the brain developmental process. It is however maintained whole life long at lower rate, and myelin sheath is crucial for proper signal transmission and neuronal survival. Unfortunately, OLGs present a high susceptibility to oxidative stress, thus demyelination often takes place secondary to diverse brain lesions or pathologies. OLGs can also be the target of immune attacks, leading to primary demyelination lesions. Following oligodendrocytic death, spontaneous remyelination may occur to a certain extent. In this review, we will mainly focus on the adult brain and on the two main sources of progenitor cells that contribute to oligodendrogenesis: parenchymal oligodendrocyte precursor cells (OPCs) and subventricular zone (SVZ)-derived progenitors. We will shortly come back on the main steps of oligodendrogenesis in the postnatal and adult brain, and summarize the key factors involved in the determination of oligodendrocytic fate. We will then shed light on the main causes of demyelination in the adult brain and present the animal models that have been developed to get insight on the demyelination/remyelination process. Finally, we will synthetize the results of studies searching for factors able to modulate spontaneous myelin repair.
Collapse
Affiliation(s)
- Bilal El Waly
- CNRS, Institut de Biologie du Développement de Marseille UMR 7288, Aix Marseille Université Marseille, France
| | - Magali Macchi
- CNRS, Institut de Biologie du Développement de Marseille UMR 7288, Aix Marseille Université Marseille, France
| | - Myriam Cayre
- CNRS, Institut de Biologie du Développement de Marseille UMR 7288, Aix Marseille Université Marseille, France
| | - Pascale Durbec
- CNRS, Institut de Biologie du Développement de Marseille UMR 7288, Aix Marseille Université Marseille, France
| |
Collapse
|
45
|
Silbereis JC, Nobuta H, Tsai HH, Heine VM, McKinsey GL, Meijer DH, Howard MA, Petryniak MA, Potter GB, Alberta JA, Baraban SC, Stiles CD, Rubenstein JLR, Rowitch DH. Olig1 function is required to repress dlx1/2 and interneuron production in Mammalian brain. Neuron 2014; 81:574-87. [PMID: 24507192 DOI: 10.1016/j.neuron.2013.11.024] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/04/2013] [Indexed: 12/21/2022]
Abstract
Abnormal GABAergic interneuron density, and imbalance of excitatory versus inhibitory tone, is thought to result in epilepsy, neurodevelopmental disorders, and psychiatric disease. Recent studies indicate that interneuron cortical density is determined primarily by the size of the precursor pool in the embryonic telencephalon. However, factors essential for regulating interneuron allocation from telencephalic multipotent precursors are poorly understood. Here we report that Olig1 represses production of GABAergic interneurons throughout the mouse brain. Olig1 deletion in mutant mice results in ectopic expression and upregulation of Dlx1/2 genes in the ventral medial ganglionic eminences and adjacent regions of the septum, resulting in an ∼30% increase in adult cortical interneuron numbers. We show that Olig1 directly represses the Dlx1/2 I12b intergenic enhancer and that Dlx1/2 functions genetically downstream of Olig1. These findings establish Olig1 as an essential repressor of Dlx1/2 and interneuron production in developing mammalian brain.
Collapse
Affiliation(s)
- John C Silbereis
- Department of Pediatrics, Eli and Edythe Broad Institute for Stem Cell Research and Regeneration Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Neurosurgery, University of California, San Francisco, San Francisco, CA 94143, USA; Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Hiroko Nobuta
- Department of Pediatrics, Eli and Edythe Broad Institute for Stem Cell Research and Regeneration Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Neurosurgery, University of California, San Francisco, San Francisco, CA 94143, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Hui-Hsin Tsai
- Department of Pediatrics, Eli and Edythe Broad Institute for Stem Cell Research and Regeneration Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Neurosurgery, University of California, San Francisco, San Francisco, CA 94143, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Vivi M Heine
- Department of Pediatrics, Eli and Edythe Broad Institute for Stem Cell Research and Regeneration Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Neurosurgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Gabriel L McKinsey
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Psychiatry, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Dimphna H Meijer
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Mackenzie A Howard
- Department of Neurosurgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Magda A Petryniak
- Department of Pediatrics, Eli and Edythe Broad Institute for Stem Cell Research and Regeneration Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Gregory B Potter
- Department of Pediatrics, Eli and Edythe Broad Institute for Stem Cell Research and Regeneration Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - John A Alberta
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Scott C Baraban
- Department of Neurosurgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Charles D Stiles
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - John L R Rubenstein
- Department of Psychiatry, University of California, San Francisco, San Francisco, CA 94143, USA
| | - David H Rowitch
- Department of Pediatrics, Eli and Edythe Broad Institute for Stem Cell Research and Regeneration Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Neurosurgery, University of California, San Francisco, San Francisco, CA 94143, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| |
Collapse
|
46
|
Xapelli S, Agasse F, Grade S, Bernardino L, Ribeiro FF, Schitine CS, Heimann AS, Ferro ES, Sebastião AM, De Melo Reis RA, Malva JO. Modulation of subventricular zone oligodendrogenesis: a role for hemopressin? Front Cell Neurosci 2014; 8:59. [PMID: 24578683 PMCID: PMC3936357 DOI: 10.3389/fncel.2014.00059] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Accepted: 02/07/2014] [Indexed: 11/13/2022] Open
Abstract
Neural stem cells (NSCs) from the subventricular zone (SVZ) have been indicated as a source of new oligodendrocytes to use in regenerative medicine for myelin pathologies. Indeed, NSCs are multipotent cells that can self-renew and differentiate into all neural cell types of the central nervous system. In normal conditions, SVZ cells are poorly oligodendrogenic, nevertheless their oligodendrogenic potential is boosted following demyelination. Importantly, progressive restriction into the oligodendrocyte fate is specified by extrinsic and intrinsic factors, endocannabinoids being one of these factors. Although a role for endocannabinoids in oligodendrogenesis has already been foreseen, selective agonists and antagonists of cannabinoids receptors produce severe adverse side effects. Herein, we show that hemopressin (Hp), a modulator of CB1 receptors, increased oligodendroglial differentiation in SVZ neural stem/progenitor cell cultures derived from neonatal mice. The original results presented in this work suggest that Hp and derivates may be of potential interest for the development of future strategies to treat demyelinating diseases.
Collapse
Affiliation(s)
- Sara Xapelli
- Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra Coimbra, Portugal ; Institute of Pharmacology and Neurosciences, Faculty of Medicine, University of Lisbon Lisboa, Portugal ; Unit of Neurosciences, Institute of Molecular Medicine, University of Lisbon Lisboa, Portugal
| | - Fabienne Agasse
- Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra Coimbra, Portugal
| | - Sofia Grade
- Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra Coimbra, Portugal ; Institute for Stem Cell Research, Helmholtz Centre Munich, German Research Centre for Environmental Health Neuherberg, Germany ; Department of Physiological Genomics, Faculty of Medicine, Ludwig-Maximilians University of Munich Munich, Germany
| | - Liliana Bernardino
- Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra Coimbra, Portugal ; Health Sciences Research Center, University of Beira Interior Covilhã, Portugal
| | - Filipa F Ribeiro
- Institute of Pharmacology and Neurosciences, Faculty of Medicine, University of Lisbon Lisboa, Portugal ; Unit of Neurosciences, Institute of Molecular Medicine, University of Lisbon Lisboa, Portugal
| | - Clarissa S Schitine
- Neurochemistry Laboratory, Biophysics Institute Carlos Chagas Filho, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| | | | - Emer S Ferro
- Departamento de Biologia Celular e do Desenvolvimento, Instituto de Ciências Biomédicas São Paulo, Brazil
| | - Ana M Sebastião
- Institute of Pharmacology and Neurosciences, Faculty of Medicine, University of Lisbon Lisboa, Portugal ; Unit of Neurosciences, Institute of Molecular Medicine, University of Lisbon Lisboa, Portugal
| | - Ricardo A De Melo Reis
- Neurochemistry Laboratory, Biophysics Institute Carlos Chagas Filho, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| | - João O Malva
- Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra Coimbra, Portugal ; Center of Investigation in Environment, Genetics and Oncobiology, Institute for Biomedical Imaging and Life Sciences, Faculty of Medicine, University of Coimbra Coimbra, Portugal
| |
Collapse
|
47
|
Maki T, Liang AC, Miyamoto N, Lo EH, Arai K. Mechanisms of oligodendrocyte regeneration from ventricular-subventricular zone-derived progenitor cells in white matter diseases. Front Cell Neurosci 2013; 7:275. [PMID: 24421755 PMCID: PMC3872787 DOI: 10.3389/fncel.2013.00275] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Accepted: 12/10/2013] [Indexed: 12/31/2022] Open
Abstract
White matter dysfunction is an important part of many CNS disorders including multiple sclerosis (MS) and vascular dementia. Within injured areas, myelin loss and oligodendrocyte death may trigger endogenous attempts at regeneration. However, during disease progression, remyelination failure may eventually occur due to impaired survival/proliferation, migration/recruitment, and differentiation of oligodendrocyte precursor cells (OPCs). The ventricular-subventricular zone (V-SVZ) and the subgranular zone (SGZ) are the main sources of neural stem/progenitor cells (NSPCs), which can give rise to neurons as well as OPCs. Under normal conditions in the adult brain, the V-SVZ progenitors generate a large number of neurons with a small number of oligodendrocyte lineage cells. However, after demyelination, the fate of V-SVZ-derived progenitor cells shifts from neurons to OPCs, and these newly generated OPCs migrate to the demyelinating lesions to ease white matter damage. In this mini-review, we will summarize the recent studies on extrinsic (e.g., vasculature, extracellular matrix (ECM), cerebrospinal fluid (CSF)) and intrinsic (e.g., transcription factors, epigenetic modifiers) factors, which mediate oligodendrocyte generation from the V-SVZ progenitor cells. A deeper understanding of the mechanisms that regulate the fate of V-SVZ progenitor cells may lead to new therapeutic approaches for ameliorating white matter dysfunction and damage in CNS disorders.
Collapse
Affiliation(s)
- Takakuni Maki
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School Charlestown, MA, USA
| | - Anna C Liang
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School Charlestown, MA, USA
| | - Nobukazu Miyamoto
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School Charlestown, MA, USA
| | - Eng H Lo
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School Charlestown, MA, USA
| | - Ken Arai
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School Charlestown, MA, USA
| |
Collapse
|
48
|
Non-neuronal cell responses differ between normal and Down syndrome developing brains. Int J Dev Neurosci 2013; 31:796-803. [PMID: 24113258 DOI: 10.1016/j.ijdevneu.2013.09.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Revised: 09/27/2013] [Accepted: 09/28/2013] [Indexed: 11/23/2022] Open
Abstract
Down syndrome (DS), the most common genetic cause of mental retardation, is characterized by reduced number of neurons and delayed myelination. Though non-neuronal cells in the brain are vital for the development, survival, and function of neurons, there is a paucity of comparative studies of normal development and DS, in particular in the temporal lobe, a region of interest for cognitive decline. We evaluated immunoreactivity for CD68 (macrophage), HLA-DR (microglia), Olig2 and TPPP/p25 (oligodendroglia), and GFAP (astroglia) in the germinal matrix (GM), temporal lobe white matter (TeWM) and hippocampus from 14 weeks of gestations to newborn in 28 DS patients and 30 age-matched controls. The rate of increase of CD68 positive cells in the GM, CA1 hippocampal subregion and subiculum was significantly higher in DS. The density of Olig2 positive cells in the GM was lower in DS brains at early stages, then showed a transient increase contrasting controls. Olig2 expression increased more in the TeWM in DS, suggesting an altered pattern of oligodendrocyte progenitor generation. GFAP-immunoreactivity in DS was significantly lower in the middle pregnancy period in the TeWM and did not increase between early and middle periods in the GM compared to controls, likely reflecting a defect in astrocyte production. The altered expression of non-neuronal cell markers during normal development and DS may play a role in, or reflect, defective neurogenesis, leading to reduced number of neurons and delayed myelination in the developing DS brain. This has implications for the understanding of the mental retardation in DS patients.
Collapse
|
49
|
Hussain R, Ghoumari AM, Bielecki B, Steibel J, Boehm N, Liere P, Macklin WB, Kumar N, Habert R, Mhaouty-Kodja S, Tronche F, Sitruk-Ware R, Schumacher M, Ghandour MS. The neural androgen receptor: a therapeutic target for myelin repair in chronic demyelination. ACTA ACUST UNITED AC 2013; 136:132-46. [PMID: 23365095 DOI: 10.1093/brain/aws284] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Myelin regeneration is a major therapeutic goal in demyelinating diseases, and the failure to remyelinate rapidly has profound consequences for the health of axons and for brain function. However, there is no efficient treatment for stimulating myelin repair, and current therapies are limited to anti-inflammatory agents. Males are less likely to develop multiple sclerosis than females, but often have a more severe disease course and reach disability milestones at an earlier age than females, and these observations have spurred interest in the potential protective effects of androgens. Here, we demonstrate that testosterone treatment efficiently stimulates the formation of new myelin and reverses myelin damage in chronic demyelinated brain lesions, resulting from the long-term administration of cuprizone, which is toxic for oligodendrocytes. In addition to the strong effect of testosterone on myelin repair, the number of activated astrocytes and microglial cells returned to low control levels, indicating a reduction of neuroinflammatory responses. We also identify the neural androgen receptor as a novel therapeutic target for myelin recovery. After the acute demyelination of cerebellar slices in organotypic culture, the remyelinating actions of testosterone could be mimicked by 5α-dihydrotestosterone, a metabolite that is not converted to oestrogens, and blocked by the androgen receptor antagonist flutamide. Testosterone treatment also failed to promote remyelination after chronic cuprizone-induced demyelination in mice with a non-functional androgen receptor. Importantly, testosterone did not stimulate the formation of new myelin sheaths after specific knockout of the androgen receptor in neurons and macroglial cells. Thus, the neural brain androgen receptor is required for the remyelination effect of testosterone, whereas the presence of the receptor in microglia and in peripheral tissues is not sufficient to enhance remyelination. The potent synthetic testosterone analogue 7α-methyl-19-nortestosterone, which has been developed for long-term male contraception and androgen replacement therapy in hypogonadal males and does not stimulate prostate growth, also efficiently promoted myelin repair. These data establish the efficacy of androgens as remyelinating agents and qualify the brain androgen receptor as a promising drug target for remyelination therapy, thus providing the preclinical rationale for a novel therapeutic use of androgens in males with multiple sclerosis.
Collapse
Affiliation(s)
- Rashad Hussain
- UMR 788 INSERM and Université Paris-Sud, 94276 Kremlin-Bicêtre, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Savary K, Caglayan D, Caja L, Tzavlaki K, Bin Nayeem S, Bergström T, Jiang Y, Uhrbom L, Forsberg-Nilsson K, Westermark B, Heldin CH, Ferletta M, Moustakas A. Snail depletes the tumorigenic potential of glioblastoma. Oncogene 2013; 32:5409-20. [PMID: 23524585 PMCID: PMC3898470 DOI: 10.1038/onc.2013.67] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Revised: 12/02/2012] [Accepted: 01/05/2013] [Indexed: 12/26/2022]
Abstract
Glioblastoma multiforme (GBM) is an aggressive brain malignancy characterized by high heterogeneity and invasiveness. It is increasingly accepted that the refractory feature of GBM to current therapies stems from the existence of few tumorigenic cells that sustain tumor growth and spreading, the so-called glioma-initiating cells (GICs). Previous studies showed that cytokines of the bone morphogenetic protein (BMP) family induce differentiation of the GICs, and thus act as tumor suppressors. Molecular pathways that explain this behavior of BMP cytokines remain largely elusive. Here, we show that BMP signaling induces Smad-dependent expression of the transcriptional regulator Snail in a rapid and sustained manner. Consistent with its already established promigratory function in other cell types, we report that Snail silencing decreases GBM cell migration. Consequently, overexpression of Snail increases GBM invasiveness in a mouse xenograft model. Surprisingly, we found that Snail depletes the GBM capacity to form gliomaspheres in vitro and to grow tumors in vivo, both of which are important features shared by GICs. Thus Snail, acting downstream of BMP signaling, dissociates the invasive capacity of GBM cells from their tumorigenic potential.
Collapse
Affiliation(s)
- K Savary
- Ludwig Institute for Cancer Research, Science for Life Laboratory, Biomedical Center, Uppsala University, Uppsala, Sweden
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|