1
|
Tahmasian N, Feng MY, Arbabi K, Rusu B, Cao W, Kukreja B, Lubotzky A, Wainberg M, Tripathy SJ, Kalish BT. Neonatal Brain Injury Triggers Niche-Specific Changes to Cellular Biogeography. eNeuro 2024; 11:ENEURO.0224-24.2024. [PMID: 39681473 DOI: 10.1523/eneuro.0224-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 10/28/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024] Open
Abstract
Preterm infants are at risk for brain injury and neurodevelopmental impairment due, in part, to white matter injury following chronic hypoxia exposure. However, the precise molecular mechanisms by which neonatal hypoxia disrupts early neurodevelopment are poorly understood. Here, we constructed a brain-wide map of the regenerative response to newborn brain injury using high-resolution imaging-based spatial transcriptomics to analyze over 800,000 cells in a mouse model of chronic neonatal hypoxia. Additionally, we developed a new method for inferring condition-associated differences in cell type spatial proximity, enabling the identification of niche-specific changes in cellular architecture. We observed hypoxia-associated changes in region-specific cell states, cell type composition, and spatial organization. Importantly, our analysis revealed mechanisms underlying reparative neurogenesis and gliogenesis, while also nominating pathways that may impede circuit rewiring following neonatal hypoxia. Altogether, our work provides a comprehensive description of the molecular response to newborn brain injury.
Collapse
Affiliation(s)
- Nareh Tahmasian
- Program in Neuroscience and Mental Health, SickKids Research Institute, Toronto, Ontario M5G 1L7, Canada
- Department of Laboratory Medicine and Pathology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
- Department of Biological Sciences, Sunnybrook Research Institute, Toronto, Ontario M4N 3M5, Canada
| | - Min Yi Feng
- Program in Neuroscience and Mental Health, SickKids Research Institute, Toronto, Ontario M5G 1L7, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1A8, Canada
| | - Keon Arbabi
- Institute of Medical Science, University of Toronto, Toronto, Ontario M5G 1A8, Canada
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, Ontario M5T 1R8, Canada
| | - Bianca Rusu
- Program in Neuroscience and Mental Health, SickKids Research Institute, Toronto, Ontario M5G 1L7, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1A8, Canada
| | - Wuxinhao Cao
- Program in Neuroscience and Mental Health, SickKids Research Institute, Toronto, Ontario M5G 1L7, Canada
| | - Bharti Kukreja
- Program in Neuroscience and Mental Health, SickKids Research Institute, Toronto, Ontario M5G 1L7, Canada
| | - Asael Lubotzky
- Division of Neurology, Department of Paediatrics, Hospital for Sick Children, Toronto, Ontario M5G 1L7, Canada
| | - Michael Wainberg
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, Ontario M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario M5G 1A8, Canada
- Prosserman Centre for Population Health Research, Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario M5G 1X5, Canada
| | - Shreejoy J Tripathy
- Institute of Medical Science, University of Toronto, Toronto, Ontario M5G 1A8, Canada
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, Ontario M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario M5G 1A8, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario M5G 1A8, Canada
| | - Brian T Kalish
- Program in Neuroscience and Mental Health, SickKids Research Institute, Toronto, Ontario M5G 1L7, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1A8, Canada
- Division of Neonatology, Department of Paediatrics, Hospital for Sick Children, Toronto, Ontario M5G 1L7, Canada
| |
Collapse
|
2
|
Dansu DK, Selcen I, Sauma S, Prentice E, Huang D, Li M, Moyon S, Casaccia P. Histone H4 acetylation differentially modulates proliferation in adult oligodendrocyte progenitors. J Cell Biol 2024; 223:e202308064. [PMID: 39133301 PMCID: PMC11318668 DOI: 10.1083/jcb.202308064] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 06/18/2024] [Accepted: 07/29/2024] [Indexed: 08/13/2024] Open
Abstract
Adult oligodendrocyte progenitors (aOPCs) generate myelinating oligodendrocytes like neonatal progenitors (nOPCs), and they also display unique functional features. Here, using unbiased histone proteomics analysis and ChIP sequencing analysis of PDGFRα+ OPCs sorted from neonatal and adult Pdgfra-H2B-EGFP reporter mice, we identify the activating H4K8ac histone mark as enriched in the aOPCs. We detect increased occupancy of the H4K8ac activating mark at chromatin locations corresponding to genes related to the progenitor state (e.g., Hes5, Gpr17), metabolic processes (e.g., Txnip, Ptdgs), and myelin components (e.g., Cnp, Mog). aOPCs showed higher levels of transcripts related to lipid metabolism and myelin, and lower levels of transcripts related to cell cycle and proliferation compared with nOPCs. In addition, pharmacological inhibition of histone acetylation decreased the expression of the H4K8ac target genes in aOPCs and decreased their proliferation. Overall, this study identifies acetylation of the histone H4K8 as a regulator of the proliferative capacity of aOPCs.
Collapse
Affiliation(s)
- David K. Dansu
- Neuroscience Initiative, Advanced Science Research Center at the City University of New York, New York, NY, USA
- Graduate Program in Biochemistry, The Graduate Center of The City University of New York, New York, NY, USA
| | - Ipek Selcen
- Neuroscience Initiative, Advanced Science Research Center at the City University of New York, New York, NY, USA
- Graduate Program in Biochemistry, The Graduate Center of The City University of New York, New York, NY, USA
| | - Sami Sauma
- Neuroscience Initiative, Advanced Science Research Center at the City University of New York, New York, NY, USA
- Graduate Program in Biology, The Graduate Center of The City University of New York, New York, NY, USA
| | - Emily Prentice
- Neuroscience Initiative, Advanced Science Research Center at the City University of New York, New York, NY, USA
- Graduate Program in Biology, The Graduate Center of The City University of New York, New York, NY, USA
| | - Dennis Huang
- Neuroscience Initiative, Advanced Science Research Center at the City University of New York, New York, NY, USA
- Graduate Program in Biology, The Graduate Center of The City University of New York, New York, NY, USA
| | - Meng Li
- Norris Medical Library, University of Southern California, Los Angeles, CA, USA
| | - Sarah Moyon
- Neuroscience Initiative, Advanced Science Research Center at the City University of New York, New York, NY, USA
- Institute of NeuroPhysiopathology (INP) UMR7051, Aix-Marseille University, CNRS, Marseille, France
| | - Patrizia Casaccia
- Neuroscience Initiative, Advanced Science Research Center at the City University of New York, New York, NY, USA
- Graduate Program in Biochemistry, The Graduate Center of The City University of New York, New York, NY, USA
- Graduate Program in Biology, The Graduate Center of The City University of New York, New York, NY, USA
| |
Collapse
|
3
|
Verploegh ISC, Conidi A, El Hassnaoui H, Verhoeven FAM, Korporaal AL, Ntafoulis I, van den Hout MCGN, Brouwer RWW, Lamfers MLM, van IJcken WFJ, Huylebroeck D, Leenstra S. BMP4 and Temozolomide Synergize in the Majority of Patient-Derived Glioblastoma Cultures. Int J Mol Sci 2024; 25:10176. [PMID: 39337661 PMCID: PMC11432198 DOI: 10.3390/ijms251810176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 09/16/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024] Open
Abstract
One of the main causes of poor prognoses in patient with glioblastoma (GBM) is drug resistance to current standard treatment, which includes chemoradiation and adjuvant temozolomide (TMZ). In addition, the concept of cancer stem cells provides new insights into therapy resistance and management also in GBM and glioblastoma stem cell-like cells (GSCs), which might contribute to therapy resistance. Bone morphogenetic protein-4 (BMP4) stimulates astroglial differentiation of GSCs and thereby reduces their self-renewal capacity. Exposure of GSCs to BMP4 may also sensitize these cells to TMZ. A recent phase I trial has shown that local delivery of BMP4 is safe, but a large variation in survival is seen in these treated patients and in features of their cultured tumors. We wanted to combine TMZ and BMP4 (TMZ + BMP4) therapy and assess the inter-tumoral variability in response to TMZ + BMP4 in patient-derived GBM cultures. A phase II trial could then benefit a larger group of patients than those treated with BMP4 only. We first show that simultaneous treatment with TMZ + BMP4 is more effective than sequential treatment. Second, when applying our optimized treatment protocol, 70% of a total of 20 GBM cultures displayed TMZ + BMP4 synergy. This combination induces cellular apoptosis and does not inhibit cell proliferation. Comparative bulk RNA-sequencing indicates that treatment with TMZ + BMP4 eventually results in decreased MAPK signaling, in line with previous evidence that increased MAPK signaling is associated with resistance to TMZ. Based on these results, we advocate further clinical trial research to test patient benefit and validate pathophysiological hypothesis.
Collapse
Affiliation(s)
- Iris S. C. Verploegh
- Department of Neurosurgery, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands; (I.S.C.V.)
- Department of Cell Biology, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Andrea Conidi
- Department of Cell Biology, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
- Department of Clinical Genetics, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Hoesna El Hassnaoui
- Department of Neurosurgery, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands; (I.S.C.V.)
| | - Floor A. M. Verhoeven
- Department of Neurosurgery, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands; (I.S.C.V.)
| | - Anne L. Korporaal
- Department of Cell Biology, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Ioannis Ntafoulis
- Department of Neurosurgery, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands; (I.S.C.V.)
| | - Mirjam C. G. N. van den Hout
- Department of Cell Biology, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
- Center for Biomics, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Rutger W. W. Brouwer
- Department of Cell Biology, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
- Center for Biomics, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Martine L. M. Lamfers
- Department of Neurosurgery, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands; (I.S.C.V.)
| | - Wilfred F. J. van IJcken
- Department of Cell Biology, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
- Center for Biomics, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Danny Huylebroeck
- Department of Cell Biology, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Sieger Leenstra
- Department of Neurosurgery, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands; (I.S.C.V.)
| |
Collapse
|
4
|
Dansu DK, Sauma S, Huang D, Li M, Moyon S, Casaccia P. The epigenetic landscape of oligodendrocyte progenitors changes with time. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.06.579145. [PMID: 38501119 PMCID: PMC10946295 DOI: 10.1101/2024.02.06.579145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/20/2024]
Abstract
SUMMARY Dansu et al. identify distinct histone H4 modifications as potential mechanism underlying the functional differences between adult and neonatal progenitors. While H4K8ac favors the expression of differentiation genes, their expression is halted by H4K20me3. Adult oligodendrocyte progenitors (aOPCs) generate myelinating oligodendrocytes, like neonatal progenitors (nOPCs), but they also display unique functional features. Here, using RNA-sequencing, unbiased histone proteomics analysis and ChIP-sequencing, we define the transcripts and histone marks underlying the unique properties of aOPCs. We describe the lower proliferative capacity and higher levels of expression of oligodendrocyte specific genes in aOPCs compared to nOPCs, as well as the greater levels of H4 histone marks. We also report increased occupancy of the H4K8ac mark at chromatin locations corresponding to oligodendrocyte-specific transcription factors and lipid metabolism genes. Pharmacological inhibition of H4K8ac deposition reduces the levels of these transcripts in aOPCs, rendering their transcriptome more similar to nOPCs. The repressive H4K20me3 mark is also higher in aOPCs compared to nOPCs and pharmacological inhibition of its deposition results in increased levels of genes related to the mature oligodendrocyte state. Overall, this study identifies two histone marks which are important for the unique transcriptional and functional identity of aOPCs.
Collapse
|
5
|
Selcen I, Prentice E, Casaccia P. The epigenetic landscape of oligodendrocyte lineage cells. Ann N Y Acad Sci 2023; 1522:24-41. [PMID: 36740586 PMCID: PMC10085863 DOI: 10.1111/nyas.14959] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The epigenetic landscape of oligodendrocyte lineage cells refers to the cell-specific modifications of DNA, chromatin, and RNA that define a unique gene expression pattern of functionally specialized cells. Here, we focus on the epigenetic changes occurring as progenitors differentiate into myelin-forming cells and respond to the local environment. First, modifications of DNA, RNA, nucleosomal histones, key principles of chromatin organization, topologically associating domains, and local remodeling will be reviewed. Then, the relationship between epigenetic modulators and RNA processing will be explored. Finally, the reciprocal relationship between the epigenome as a determinant of the mechanical properties of cell nuclei and the target of mechanotransduction will be discussed. The overall goal is to provide an interpretative key on how epigenetic changes may account for the heterogeneity of the transcriptional profiles identified in this lineage.
Collapse
Affiliation(s)
- Ipek Selcen
- Graduate Program in Biochemistry, The Graduate Center of The City University of New York, New York, New York, USA.,Neuroscience Initiative, Advanced Science Research Center, The Graduate Center of The City University of New York, New York, New York, USA
| | - Emily Prentice
- Neuroscience Initiative, Advanced Science Research Center, The Graduate Center of The City University of New York, New York, New York, USA.,Graduate Program in Biology, The Graduate Center of The City University of New York, New York, New York, USA
| | - Patrizia Casaccia
- Graduate Program in Biochemistry, The Graduate Center of The City University of New York, New York, New York, USA.,Neuroscience Initiative, Advanced Science Research Center, The Graduate Center of The City University of New York, New York, New York, USA.,Graduate Program in Biology, The Graduate Center of The City University of New York, New York, New York, USA
| |
Collapse
|
6
|
Douceau S, Deutsch Guerrero T, Ferent J. Establishing Hedgehog Gradients during Neural Development. Cells 2023; 12:225. [PMID: 36672161 PMCID: PMC9856818 DOI: 10.3390/cells12020225] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/23/2022] [Accepted: 12/25/2022] [Indexed: 01/07/2023] Open
Abstract
A morphogen is a signaling molecule that induces specific cellular responses depending on its local concentration. The concept of morphogenic gradients has been a central paradigm of developmental biology for decades. Sonic Hedgehog (Shh) is one of the most important morphogens that displays pleiotropic functions during embryonic development, ranging from neuronal patterning to axon guidance. It is commonly accepted that Shh is distributed in a gradient in several tissues from different origins during development; however, how these gradients are formed and maintained at the cellular and molecular levels is still the center of a great deal of research. In this review, we first explored all of the different sources of Shh during the development of the nervous system. Then, we detailed how these sources can distribute Shh in the surrounding tissues via a variety of mechanisms. Finally, we addressed how disrupting Shh distribution and gradients can induce severe neurodevelopmental disorders and cancers. Although the concept of gradient has been central in the field of neurodevelopment since the fifties, we also describe how contemporary leading-edge techniques, such as organoids, can revisit this classical model.
Collapse
Affiliation(s)
- Sara Douceau
- INSERM UMR-S 1270, F-75005 Paris, France
- Institut du Fer à Moulin, INSERM, Sorbonne Univeristy, F-75005 Paris, France
| | - Tanya Deutsch Guerrero
- INSERM UMR-S 1270, F-75005 Paris, France
- Institut du Fer à Moulin, INSERM, Sorbonne Univeristy, F-75005 Paris, France
| | - Julien Ferent
- INSERM UMR-S 1270, F-75005 Paris, France
- Institut du Fer à Moulin, INSERM, Sorbonne Univeristy, F-75005 Paris, France
| |
Collapse
|
7
|
Manzari-Tavakoli A, Babajani A, Farjoo MH, Hajinasrollah M, Bahrami S, Niknejad H. The Cross-Talks Among Bone Morphogenetic Protein (BMP) Signaling and Other Prominent Pathways Involved in Neural Differentiation. Front Mol Neurosci 2022; 15:827275. [PMID: 35370542 PMCID: PMC8965007 DOI: 10.3389/fnmol.2022.827275] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 02/14/2022] [Indexed: 11/21/2022] Open
Abstract
The bone morphogenetic proteins (BMPs) are a group of potent morphogens which are critical for the patterning, development, and function of the central nervous system. The appropriate function of the BMP pathway depends on its interaction with other signaling pathways involved in neural differentiation, leading to synergistic or antagonistic effects and ultimately favorable biological outcomes. These opposite or cooperative effects are observed when BMP interacts with fibroblast growth factor (FGF), cytokines, Notch, Sonic Hedgehog (Shh), and Wnt pathways to regulate the impact of BMP-induced signaling in neural differentiation. Herein, we review the cross-talk between BMP signaling and the prominent signaling pathways involved in neural differentiation, emphasizing the underlying basic molecular mechanisms regarding the process of neural differentiation. Knowing these cross-talks can help us to develop new approaches in regenerative medicine and stem cell based therapy. Recently, cell therapy has received significant attention as a promising treatment for traumatic or neurodegenerative diseases. Therefore, it is important to know the signaling pathways involved in stem cell differentiation toward neural cells. Our better insight into the cross-talk of signaling pathways during neural development would improve neural differentiation within in vitro tissue engineering approaches and pre-clinical practices and develop futuristic therapeutic strategies for patients with neurological disease.
Collapse
Affiliation(s)
- Asma Manzari-Tavakoli
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Rayan Center for Neuroscience & Behavior, Department of Biology, Faculty of Science, Ferdowsi University, Mashhad, Iran
| | - Amirhesam Babajani
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Hadi Farjoo
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mostafa Hajinasrollah
- Department of Stem Cells and Developmental Biology, Cell Sciences Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Soheyl Bahrami
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology in AUVA Research Center, Vienna, Austria
| | - Hassan Niknejad
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
8
|
Han W, Pan Y, Han Z, Cheng L, Jiang L. Advanced Maternal Age Impairs Myelination in Offspring Rats. Front Pediatr 2022; 10:850213. [PMID: 35311052 PMCID: PMC8927774 DOI: 10.3389/fped.2022.850213] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 02/07/2022] [Indexed: 11/13/2022] Open
Abstract
The effects of advanced maternal age (AMA) on the neurodevelopment of offspring are becoming increasingly important. Myelination is an important aspect of brain development; however, a limited number of studies have focused on the effects of AMA on myelination in offspring. The current study aims to evaluate the association between AMA and myelin sheath development in offspring. We studied the learning and memory function of immature offspring using the novel object recognition test. Then, we investigated the expression of myelin basic protein (MBP) in the immature offspring of young (3-month-old) and old (12-month-old) female rats at different time points (14, 28, and 60 days) after birth with immunofluorescence and western blotting. The myelin sheath ultrastructure was observed with transmission electron microscopy in immature and mature offspring. Extracellular signal-regulated kinase 1 and 2 (ERK1/2) and phosphorylated ERK1/2 (p-ERK1/2) were investigated by western blot in immature offspring at the above time points. AMA impaired the memory function of offspring during early postnatal days. The MBP expression level gradually increased with postnatal development in the offspring of both the AMA and Control (Ctl) groups, but the MBP level in the offspring of the AMA group was lower than that of the Ctl group at 14 days after birth. In addition, the ultrastructure of the myelin sheath was defective in AMA offspring during the early postnatal period; however, the myelin sheath was not significantly affected in offspring during adulthood. Interestingly, ERK phosphorylation at 14 days after birth was lower in AMA offspring than in Ctl offspring. However, ERK phosphorylation at 28 days after birth was higher in AMA offspring than in Ctl offspring. The peak of ERK phosphorylation in the AMA group was abnormal and delayed. Our results indicated that AMA is associated with poor developmental myelin formation in offspring. The ERK signaling pathway may play an essential role in the adverse effects of AMA on the offspring myelin sheath development.
Collapse
Affiliation(s)
- Wei Han
- Department of Neurology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Ya'nan Pan
- Department of Neurology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Ziyao Han
- Department of Neurology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Li Cheng
- Department of Neurology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Li Jiang
- Department of Neurology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| |
Collapse
|
9
|
Dansu DK, Sauma S, Casaccia P. Oligodendrocyte progenitors as environmental biosensors. Semin Cell Dev Biol 2021; 116:38-44. [PMID: 33092959 PMCID: PMC8053729 DOI: 10.1016/j.semcdb.2020.09.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/23/2020] [Accepted: 09/27/2020] [Indexed: 01/10/2023]
Abstract
The past decade has seen an important revision of the traditional concept of the role and function of glial cells. From "passive support" for neurons, oligodendrocyte lineage cells are now recognized as metabolic exchangers with neurons, a cellular interface with blood vessels and responders to gut-derived metabolites or changes in the social environment. In the developing brain, the differentiation of neonatal oligodendrocyte progenitors (nOPCs) is required for normal brain function. In adulthood, the differentiation of adult OPCs (aOPCs) serves an important role in learning, behavioral adaptation and response to myelin injury. Here, we propose the concept of OPCs as environmental biosensors, which "sense" chemical and physical stimuli over time and adjust to the new challenges by modifying their epigenome and consequent transcriptome. Because epigenetics defines the ability of the cell to "adapt" gene expression to changes in the environment, we propose a model of OPC differentiation resulting from time-dependent changes of the epigenomic landscape in response to declining mitogens, raising hormone levels, neuronal activity, changes in space constraints or stiffness of the extracellular matrix. We propose that the intrinsically different functional properties of aOPCs compared to nOPCs result from the accrual of "epigenetic memories" of distinct events, which are "recorded" in the nuclei of OPCs as histone and DNA marks, defining a "unique epigenomic landscape" over time.
Collapse
Affiliation(s)
- David K Dansu
- Graduate Program in Biochemistry, Graduate Center of the City University of New York, New York, NY, USA; Neuroscience Initiative, Advanced Science Research Center, The Graduate Center of the City University of New York, New York, NY, USA
| | - Sami Sauma
- Graduate Program in Biology, Graduate Center of the City University of New York, New York, NY, USA; Neuroscience Initiative, Advanced Science Research Center, The Graduate Center of the City University of New York, New York, NY, USA
| | - Patrizia Casaccia
- Graduate Program in Biochemistry, Graduate Center of the City University of New York, New York, NY, USA; Graduate Program in Biology, Graduate Center of the City University of New York, New York, NY, USA; Neuroscience Initiative, Advanced Science Research Center, The Graduate Center of the City University of New York, New York, NY, USA.
| |
Collapse
|
10
|
Cui H, Yu W, Yu M, Luo Y, Yang M, Cong R, Chu X, Gao G, Zhong M. GPR126 regulates colorectal cancer cell proliferation by mediating HDAC2 and GLI2 expression. Cancer Sci 2021; 112:1798-1810. [PMID: 33629464 PMCID: PMC8088945 DOI: 10.1111/cas.14868] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 02/20/2021] [Accepted: 02/20/2021] [Indexed: 12/15/2022] Open
Abstract
The G‐protein‐coupled receptor 126 (GPR126) may play an important role in tumor development, although its role remains poorly understood. We found that GPR126 had higher expression in most colorectal cancer cell lines than in normal colon epithelial cell lines, and higher expression levels in colorectal cancer tissues than in normal adjacent colon tissues. GPR126 knockdown induced by shRNA inhibited cell viability and colony formation in HT‐29, HCT116, and LoVo cells, decreased BrdU incorporation into newly synthesized proliferating HT‐29 cells, led to an arrest of cell cycle progression at the G1 phase in HCT‐116 and HT‐29 cells, and suppressed tumorigenesis of HT‐29, HCT116, and LoVo cells in nude mouse xenograft models. GPR126 knockdown engendered decreased transcription and translation of histone deacetylase 2 (HDAC2), previously implicated in the activation of GLI1 and GLI2 in the Hedgehog signaling pathway. Ectopic expression of HDAC2 in GPR126‐silenced cells restored cell viability and proliferation, GLI2 luciferase reporter activity, partially recovered GLI2 expression, and reduced the cell cycle arrest. HDAC2 regulated GLI2 expression and, along with GLI2, it bound to the PTCH1 promoter, as evidenced by a chip assay with HT‐29 cells. Purmorphamine, a hedgehog agonist, largely restored the cell viability and expression of GLI2 proteins in GPR126‐silenced HT‐29 cells, whereas GANT61, a hedgehog inhibitor, further enhanced the GPR126 knockdown‐induced inhibitory effects. Our findings demonstrate that GPR126 regulates colorectal cancer cell proliferation by mediating the expression of HDAC2 and GLI2, therefore it may represent a suitable therapeutic target for colorectal cancer treatment.
Collapse
Affiliation(s)
- Hengxiang Cui
- Medical Research Center, Second Affiliated Hospital of Nantong University, Nantong, China.,Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Wenjie Yu
- Department of Internal Medicine, Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Minhao Yu
- Department of Gastrointestinal Surgery, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yang Luo
- Department of Gastrointestinal Surgery, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Mingming Yang
- Department of Biochemistry and Molecular Biology, School of Medicine, Nantong University, Nantong, China
| | - Ruochen Cong
- Medical Research Center, Second Affiliated Hospital of Nantong University, Nantong, China
| | - Xin Chu
- Medical Research Center, Second Affiliated Hospital of Nantong University, Nantong, China
| | - Ganglong Gao
- Department of Gastrointestinal Surgery, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Ming Zhong
- Department of Gastrointestinal Surgery, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
11
|
Pruvost M, Moyon S. Oligodendroglial Epigenetics, from Lineage Specification to Activity-Dependent Myelination. Life (Basel) 2021; 11:62. [PMID: 33467699 PMCID: PMC7830029 DOI: 10.3390/life11010062] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 01/08/2021] [Accepted: 01/10/2021] [Indexed: 12/25/2022] Open
Abstract
Oligodendroglial cells are the myelinating cells of the central nervous system. While myelination is crucial to axonal activity and conduction, oligodendrocyte progenitor cells and oligodendrocytes have also been shown to be essential for neuronal support and metabolism. Thus, a tight regulation of oligodendroglial cell specification, proliferation, and myelination is required for correct neuronal connectivity and function. Here, we review the role of epigenetic modifications in oligodendroglial lineage cells. First, we briefly describe the epigenetic modalities of gene regulation, which are known to have a role in oligodendroglial cells. We then address how epigenetic enzymes and/or marks have been associated with oligodendrocyte progenitor specification, survival and proliferation, differentiation, and finally, myelination. We finally mention how environmental cues, in particular, neuronal signals, are translated into epigenetic modifications, which can directly influence oligodendroglial biology.
Collapse
|
12
|
The Wnt Effector TCF7l2 Promotes Oligodendroglial Differentiation by Repressing Autocrine BMP4-Mediated Signaling. J Neurosci 2021; 41:1650-1664. [PMID: 33452226 DOI: 10.1523/jneurosci.2386-20.2021] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 12/02/2020] [Accepted: 01/01/2021] [Indexed: 11/21/2022] Open
Abstract
Promoting oligodendrocyte (OL) differentiation represents a promising option for remyelination therapy for treating the demyelinating disease multiple sclerosis (MS). The Wnt effector transcription factor 7-like 2 (TCF7l2) was upregulated in MS lesions and had been proposed to inhibit OL differentiation. Recent data suggest the opposite yet underlying mechanisms remain elusive. Here, we unravel a previously unappreciated function of TCF7l2 in controlling autocrine bone morphogenetic protein (BMP)4-mediated signaling. Disrupting TCF7l2 in mice of both sexes results in oligodendroglial-specific BMP4 upregulation and canonical BMP4 signaling activation in vivo Mechanistically, TCF7l2 binds to Bmp4 gene regulatory element and directly represses its transcriptional activity. Functionally, enforced TCF7l2 expression promotes OL differentiation by reducing autocrine BMP4 secretion and dampening BMP4 signaling. Importantly, compound genetic disruption demonstrates that oligodendroglial-specific BMP4 deletion rescues arrested OL differentiation elicited by TCF7l2 disruption in vivo Collectively, our study reveals a novel connection between TCF7l2 and BMP4 in oligodendroglial lineage and provides new insights into augmenting TCF7l2 for promoting remyelination in demyelinating disorders such as MS.SIGNIFICANCE STATEMENT Incomplete or failed myelin repairs, primarily resulting from the arrested differentiation of myelin-forming oligodendrocytes (OLs) from oligodendroglial progenitor cells, is one of the major reasons for neurologic progression in people affected by multiple sclerosis (MS). Using in vitro culture systems and in vivo animal models, this study unraveled a previously unrecognized autocrine regulation of bone morphogenetic protein (BMP)4-mediated signaling by the Wnt effector transcription factor 7-like 2 (TCF7l2). We showed for the first time that TCF7l2 promotes oligodendroglial differentiation by repressing BMP4-mediated activity, which is dysregulated in MS lesions. Our study suggests that elevating TCF7l2 expression may be possible in overcoming arrested oligodendroglial differentiation as observed in MS patients.
Collapse
|
13
|
LoPresti P. HDAC6 in Diseases of Cognition and of Neurons. Cells 2020; 10:E12. [PMID: 33374719 PMCID: PMC7822434 DOI: 10.3390/cells10010012] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 12/12/2022] Open
Abstract
Central nervous system (CNS) neurodegenerative diseases are characterized by faulty intracellular transport, cognition, and aggregate regulation. Traditionally, neuroprotection exerted by histone deacetylase (HDAC) inhibitors (HDACi) has been attributed to the ability of this drug class to promote histone acetylation. However, HDAC6 in the healthy CNS functions via distinct mechanisms, due largely to its cytoplasmic localization. Indeed, in healthy neurons, cytoplasmic HDAC6 regulates the acetylation of a variety of non-histone proteins that are linked to separate functions, i.e., intracellular transport, neurotransmitter release, and aggregate formation. These three HDAC6 activities could work independently or in synergy. Of particular interest, HDAC6 targets the synaptic protein Bruchpilot and neurotransmitter release. In pathological conditions, HDAC6 becomes abundant in the nucleus, with deleterious consequences for transcription regulation and synapses. Thus, HDAC6 plays a leading role in neuronal health or dysfunction. Here, we review recent findings and novel conclusions on the role of HDAC6 in neurodegeneration. Selective studies with pan-HDACi are also included. We propose that an early alteration of HDAC6 undermines synaptic transmission, while altering transport and aggregation, eventually leading to neurodegeneration.
Collapse
Affiliation(s)
- Patrizia LoPresti
- Department of Psychology, University of Illinois at Chicago, 1007 West Harrison Street, Chicago, IL 60607, USA
| |
Collapse
|
14
|
Samudyata, Castelo-Branco G, Liu J. Epigenetic regulation of oligodendrocyte differentiation: From development to demyelinating disorders. Glia 2020; 68:1619-1630. [PMID: 32154951 DOI: 10.1002/glia.23820] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 02/19/2020] [Accepted: 03/02/2020] [Indexed: 12/16/2022]
Abstract
The maintenance of progenitor states or the differentiation of progenitors into specific lineages requires epigenetic remodeling of the gene expression program. In the central nervous system, oligodendrocyte progenitors (OPCs) give rise to oligodendrocytes (OLs), whose main function has been thought to be to produce myelin, a lipid-rich structure insulating the axons. However, recent findings suggest diverse OL transcriptional states, which might imply additional functions. The differentiation of OPCs into postmitotic OLs is a highly regulated and sensitive process and requires temporal waves of gene expression through epigenetic remodeling of the genome. In this review, we will discuss recent advances in understanding the events shaping the chromatin landscape through histone modifications and long noncoding RNAs during OPC differentiation, in physiological and pathological conditions. We suggest that epigenetic regulation plays a fundamental role in governing the accessibility of transcriptional machinery to DNA sequences, which ultimately determines functional outcomes in OLs.
Collapse
Affiliation(s)
- Samudyata
- Laboratory of Molecular Neurobiology, Department Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Gonçalo Castelo-Branco
- Laboratory of Molecular Neurobiology, Department Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden.,Ming Wai Lau Centre for Reparative Medicine, Stockholm node, Karolinska Institutet, Stockholm, Sweden
| | - Jia Liu
- Advanced Science Research Center at the Graduate Center, Neuroscience Initiative, City University of New York, New York, New York, USA
| |
Collapse
|
15
|
Mizrak D, Levitin HM, Delgado AC, Crotet V, Yuan J, Chaker Z, Silva-Vargas V, Sims PA, Doetsch F. Single-Cell Analysis of Regional Differences in Adult V-SVZ Neural Stem Cell Lineages. Cell Rep 2020; 26:394-406.e5. [PMID: 30625322 PMCID: PMC6368857 DOI: 10.1016/j.celrep.2018.12.044] [Citation(s) in RCA: 162] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 11/20/2018] [Accepted: 12/11/2018] [Indexed: 12/23/2022] Open
Abstract
The ventricular-subventricular zone (V-SVZ) harbors adult neural stem cells. V-SVZ neural stem cells exhibit features of astrocytes, have a regional identity, and depending on their location in the lateral or septal wall of the lateral ventricle, generate different types of neuronal and glial progeny. We performed large-scale single-cell RNA sequencing to provide a molecular atlas of cells from the lateral and septal adult V-SVZ of male and female mice. This revealed regional and sex differences among adult V-SVZ cells. We uncovered lineage potency bias at the single-cell level among lateral and septal wall astrocytes toward neurogenesis and oligodendrogenesis, respectively. Finally, we identified transcription factor co-expression modules marking key temporal steps in neurogenic and oligodendrocyte lineage progression. Our data suggest functionally important spatial diversity in neurogenesis and oligodendrogenesis in the adult brain and reveal molecular correlates of adult NSC dormancy and lineage specialization. Mizrak et al. performed large-scale, single-cell RNA sequencing of the adult ventricular-subventricular zone neural stem cell niche. They identify regional differences between the lateral wall and septal wall, as well as sex differences in cell types and signaling pathways.
Collapse
Affiliation(s)
- Dogukan Mizrak
- Department of Systems Biology, Columbia University Medical Center, New York, NY 10032, USA; Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA
| | - Hanna Mendes Levitin
- Department of Systems Biology, Columbia University Medical Center, New York, NY 10032, USA
| | - Ana C Delgado
- Biozentrum, University of Basel, 4056 Basel, Switzerland
| | - Valerie Crotet
- Biozentrum, University of Basel, 4056 Basel, Switzerland
| | - Jinzhou Yuan
- Department of Systems Biology, Columbia University Medical Center, New York, NY 10032, USA
| | - Zayna Chaker
- Biozentrum, University of Basel, 4056 Basel, Switzerland
| | | | - Peter A Sims
- Department of Systems Biology, Columbia University Medical Center, New York, NY 10032, USA; Sulzberger Columbia Genome Center, Columbia University Medical Center, New York, NY 10032, USA; Department of Biochemistry & Molecular Biophysics, Columbia University Medical Center, New York, NY 10032, USA.
| | - Fiona Doetsch
- Biozentrum, University of Basel, 4056 Basel, Switzerland.
| |
Collapse
|
16
|
Inhibitors of Myelination and Remyelination, Bone Morphogenetic Proteins, are Upregulated in Human Neurological Disease. Neurochem Res 2020; 45:656-662. [PMID: 32030597 DOI: 10.1007/s11064-020-02980-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 01/28/2020] [Accepted: 02/01/2020] [Indexed: 12/12/2022]
Abstract
During demyelinating disease such as multiple sclerosis and stroke, myelin is destroyed and along with it, the oligodendrocytes that synthesize the myelin. Thus, recovery is limited due to both interruptions in neuronal transmission as well as lack of support for neurons. Although oligodendrocyte progenitor cells remain abundant in the central nervous system, they rarely mature and form new functional myelin in the diseased CNS. In cell culture and in experimental models of demyelinating disease, inhibitory signaling factors decrease myelination and remyelination. One of the most potent of these are the bone morphogenetic proteins (BMPs), a family of proteins that strongly inhibits oligodendrocyte progenitor differentiation and myelination in culture. BMPs are highly expressed in the dorsal CNS during pre-natal development and serve to regulate dorsal ventral patterning. Their expression decreases after birth but is significantly increased in rodent demyelination models such as experimental autoimmune encephalomyelitis, cuprizone ingestion and spinal cord injury. However, until recently, evidence for BMP upregulation in human disease has been scarce. This review discusses new human studies showing that in multiple sclerosis and other demyelinating diseases, BMPs are expressed by immune cells invading the CNS as well as resident CNS cell types, mostly astrocytes and microglia. Expression of endogenous BMP antagonists is also regulated. Identification of BMPs in the CNS is correlated with areas of demyelination and inflammation. These studies further support BMP as a potential therapeutic target.
Collapse
|
17
|
Abstract
Structural plasticity in the myelinated infrastructure of the nervous system has come to light. Although an innate program of myelin development proceeds independent of nervous system activity, a second mode of myelination exists in which activity-dependent, plastic changes in myelin-forming cells influence myelin structure and neurological function. These complementary and possibly temporally overlapping activity-independent and activity-dependent modes of myelination crystallize in a model of experience-modulated myelin development and plasticity with broad implications for neurological function. In this article, I consider the contributions of myelin to neural circuit function, the dynamic influences of experience on myelin microstructure, and the role that plasticity of myelin may play in cognition.
Collapse
Affiliation(s)
- Michelle Monje
- Department of Neurology, Stanford University, Stanford, California 94305, USA;
| |
Collapse
|
18
|
Inhibiting Bone Morphogenetic Protein 4 Type I Receptor Signaling Promotes Remyelination by Potentiating Oligodendrocyte Differentiation. eNeuro 2019; 6:ENEURO.0399-18.2019. [PMID: 31028086 PMCID: PMC6529590 DOI: 10.1523/eneuro.0399-18.2019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 03/31/2019] [Accepted: 04/18/2019] [Indexed: 12/22/2022] Open
Abstract
Blocking inhibitory factors within CNS demyelinating lesions is regarded as a promising strategy to promote remyelination. Bone morphogenetic protein 4 (BMP4) is an inhibitory factor present in demyelinating lesions. Noggin, an endogenous antagonist to BMP, has previously been shown to increase the number of oligodendrocytes and promote remyelination in vivo. However, it remains unclear how BMP4 signaling inhibits remyelination. Here we investigated the downstream signaling pathway that mediates the inhibitory effect that BMP4 exerts upon remyelination through pharmacological and transgenic approaches. Using the cuprizone mouse model of central demyelination, we demonstrate that selectively blocking BMP4 signaling via the pharmacological inhibitor LDN-193189 significantly promotes oligodendroglial differentiation and the extent of remyelination in vivo. This was accompanied by the downregulation of transcriptional targets that suppress oligodendrocyte differentiation. Further, selective deletion of BMP receptor type IA (BMPRIA) within primary mouse oligodendrocyte progenitor cells (OPCs) significantly enhanced their differentiation and subsequent myelination in vitro. Together, the results of this study identify that BMP4 signals via BMPRIA within OPCs to inhibit oligodendroglial differentiation and their capacity to myelinate axons, and suggest that blocking the BMP4/BMPRIA pathway in OPCs is a promising strategy to promote CNS remyelination.
Collapse
|
19
|
Gou X, Tang Y, Qu Y, Xiao D, Ying J, Mu D. Could the inhibitor of DNA binding 2 and 4 play a role in white matter injury? Rev Neurosci 2019; 30:625-638. [PMID: 30738015 DOI: 10.1515/revneuro-2018-0090] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 11/02/2018] [Indexed: 01/12/2023]
Abstract
Abstract
White matter injury (WMI) prevents the normal development of myelination, leading to central nervous system myelination disorders and the production of chronic sequelae associated with WMI, such as chronic dyskinesia, cognitive impairment and cerebral palsy. This results in a large emotional and socioeconomic burden. Decreased myelination in preterm infant WMI is associated with the delayed development or destruction of oligodendrocyte (OL) lineage cells, particularly oligodendrocyte precursor cells (OPCs). The development of cells from the OL lineage involves the migration, proliferation and different stages of OL differentiation, finally leading to myelination. A series of complex intrinsic, extrinsic and epigenetic factors regulate the OPC cell cycle withdrawal, OL lineage progression and myelination. We focus on the inhibitor of DNA binding 2 (ID2), because it is widely involved in the different stages of OL differentiation and genesis. ID2 is a key transcription factor for the normal development of OL lineage cells, and the pathogenesis of WMI is closely linked with OL developmental disorders. ID4, another family member of the IDs protein, also plays a similar role in OL differentiation and genesis. ID2 and ID4 belong to the helix-loop-helix family; they lack the DNA-binding sequences and inhibit oligodendrogenesis and OPC differentiation. In this review, we mainly discuss the roles of ID2 in OL development, especially during OPC differentiation, and summarize the ID2-mediated intracellular and extracellular signaling pathways that regulate these processes. We also discuss ID4 in relation to bone morphogenetic protein signaling and oligodendrogenesis. It is likely that these developmental mechanisms are also involved in the myelin repair or remyelination in human neurological diseases.
Collapse
Affiliation(s)
- Xiaoyun Gou
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu 610041, China
| | - Ying Tang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu 610041, China
| | - Yi Qu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu 610041, China
| | - Dongqiong Xiao
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu 610041, China
| | - Junjie Ying
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu 610041, China
| | - Dezhi Mu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu 610041, China
| |
Collapse
|
20
|
Tsai E, Casaccia P. Mechano-modulation of nuclear events regulating oligodendrocyte progenitor gene expression. Glia 2019; 67:1229-1239. [PMID: 30734358 DOI: 10.1002/glia.23595] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 01/03/2019] [Accepted: 01/14/2019] [Indexed: 12/20/2022]
Abstract
Oligodendrocytes differentiate from oligodendrocyte progenitor cells (OPCs) in response to distinct extracellular signals. This process requires changes in gene expression resulting from the interplay between transcription factors and epigenetic modulators. Extracellular signals include chemical and physical stimuli. This review focuses on the signaling mechanisms activated in oligodendrocyte progenitors in response to mechanical forces. Of particular interest is a better understanding on how these forces are transduced into the OPC nuclei and subsequently reshape their epigenetic landscape. Here we will introduce the concept of epigenetic regulation of gene expression, first in general and then focusing on the oligodendrocyte lineage. We will then review the current literature on mechano-transduction in distinct cell types, followed by pathways identified in myelinating oligodendrocytes and their progenitors. Overall, the reader will be provided with a comprehensive review of the signaling pathways which allow oligodendrocyte progenitors to "sense" physical forces and transduce them into patterns of gene expression.
Collapse
Affiliation(s)
- Eric Tsai
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York.,Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Patrizia Casaccia
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York.,Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York.,Department of Biology and Biochemistry, Neuroscience Initiative at the Advanced Science Research Center of the Graduate Center of The City University of New York, New York
| |
Collapse
|
21
|
Ulanska-Poutanen J, Mieczkowski J, Zhao C, Konarzewska K, Kaza B, Pohl HB, Bugajski L, Kaminska B, Franklin RJ, Zawadzka M. Injury-induced perivascular niche supports alternative differentiation of adult rodent CNS progenitor cells. eLife 2018; 7:30325. [PMID: 30222103 PMCID: PMC6141235 DOI: 10.7554/elife.30325] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 09/03/2018] [Indexed: 01/06/2023] Open
Abstract
Following CNS demyelination, oligodendrocyte progenitor cells (OPCs) are able to differentiate into either remyelinating oligodendrocytes (OLs) or remyelinating Schwann cells (SCs). However, the signals that determine which type of remyelinating cell is generated and the underlying mechanisms involved have not been identified. Here, we show that distinctive microenvironments created in discrete niches within demyelinated white matter determine fate decisions of adult OPCs. By comparative transcriptome profiling we demonstrate that an ectopic, injury-induced perivascular niche is enriched with secreted ligands of the BMP and Wnt signalling pathways, produced by activated OPCs and endothelium, whereas reactive astrocyte within non-vascular area express the dual BMP/Wnt antagonist Sostdc1. The balance of BMP/Wnt signalling network is instructive for OPCs to undertake fate decision shortly after their activation: disruption of the OPCs homeostasis during demyelination results in BMP4 upregulation, which, in the absence of Socstdc1, favours SCs differentiation.
Collapse
Affiliation(s)
- Justyna Ulanska-Poutanen
- Laboratory of Molecular Neurobiology, Neurobiology Center, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Jakub Mieczkowski
- Laboratory of Molecular Neurobiology, Neurobiology Center, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Chao Zhao
- Wellcome Trust - Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom.,Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Katarzyna Konarzewska
- Laboratory of Molecular Neurobiology, Neurobiology Center, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Beata Kaza
- Laboratory of Molecular Neurobiology, Neurobiology Center, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Hartmut Bf Pohl
- Department of Biology, Institute of Molecular Health Sciences, Zurich, Switzerland
| | - Lukasz Bugajski
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Bozena Kaminska
- Laboratory of Molecular Neurobiology, Neurobiology Center, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Robin Jm Franklin
- Wellcome Trust - Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom.,Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Malgorzata Zawadzka
- Laboratory of Molecular Neurobiology, Neurobiology Center, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
22
|
Laouarem Y, Traiffort E. Developmental and Repairing Production of Myelin: The Role of Hedgehog Signaling. Front Cell Neurosci 2018; 12:305. [PMID: 30237763 PMCID: PMC6135882 DOI: 10.3389/fncel.2018.00305] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 08/22/2018] [Indexed: 11/13/2022] Open
Abstract
Since the discovery of its role as a morphogen directing ventral patterning of the spinal cord, the secreted protein Sonic Hedgehog (Shh) has been implicated in a wide array of events contributing to the development, maintenance and repair of the central nervous system (CNS). One of these events is the generation of oligodendrocytes, the glial cells of the CNS responsible for axon myelination. In embryo, the first oligodendroglial cells arise from the ventral ventricular zone in the developing brain and spinal cord where Shh induces the basic helix-loop-helix transcription factors Olig1 and Olig2 both necessary and sufficient for oligodendrocyte production. Later on, Shh signaling participates in the production of oligodendroglial cells in the dorsal ventricular-subventricular zone in the postnatal forebrain. Finally, the modulation of Hedgehog signaling activity promotes the repair of demyelinated lesions. This mini-review article focuses on the Shh-dependent molecular mechanisms involved in the spatial and temporal control of oligodendrocyte lineage appearance. The apparent intricacy of the roles of two essential components of Shh signaling, Smoothened and Gli1, in the postnatal production of myelin and its regeneration following a demyelinating event is also highlighted. A deeper understanding of the implication of each of the components that regulate oligodendrogenesis and myelination should beneficially influence the therapeutic strategies in the field of myelin diseases.
Collapse
Affiliation(s)
| | - Elisabeth Traiffort
- Small Molecules of Neuroprotection, Neuroregeneration and Remyelination – U1195, INSERM, University Paris-Sud/Paris-Saclay, Kremlin-Bicêtre, France
| |
Collapse
|
23
|
Scaglione A, Patzig J, Liang J, Frawley R, Bok J, Mela A, Yattah C, Zhang J, Teo SX, Zhou T, Chen S, Bernstein E, Canoll P, Guccione E, Casaccia P. PRMT5-mediated regulation of developmental myelination. Nat Commun 2018; 9:2840. [PMID: 30026560 PMCID: PMC6053423 DOI: 10.1038/s41467-018-04863-9] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 06/01/2018] [Indexed: 12/16/2022] Open
Abstract
Oligodendrocytes (OLs) are the myelin-forming cells of the central nervous system. They are derived from differentiation of oligodendrocyte progenitors through a process requiring cell cycle exit and histone modifications. Here we identify the histone arginine methyl-transferase PRMT5, a molecule catalyzing symmetric methylation of histone H4R3, as critical for developmental myelination. PRMT5 pharmacological inhibition, CRISPR/cas9 targeting, or genetic ablation decrease p53-dependent survival and impair differentiation without affecting proliferation. Conditional ablation of Prmt5 in progenitors results in hypomyelination, reduced survival and differentiation. Decreased histone H4R3 symmetric methylation is followed by increased nuclear acetylation of H4K5, and is rescued by pharmacological inhibition of histone acetyltransferases. Data obtained using purified histones further validate the results obtained in mice and in cultured oligodendrocyte progenitors. Together, these results identify PRMT5 as critical for oligodendrocyte differentiation and developmental myelination by modulating the cross-talk between histone arginine methylation and lysine acetylation. Myelin-forming cells derive from oligodendrocyte progenitors. Here the authors identify histone arginine methyl-transferase PRMT5 as critical for developmental myelination by modulating the cross-talk between histone arginine methylation and lysine acetylation, to favor differentiation.
Collapse
Affiliation(s)
- Antonella Scaglione
- Neuroscience Initiative at the Advanced Science Research Center of the Graduate Center of The City University of New York, 85 St. Nicholas Terrace, New York, NY, 10031, USA.,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA
| | - Julia Patzig
- Neuroscience Initiative at the Advanced Science Research Center of the Graduate Center of The City University of New York, 85 St. Nicholas Terrace, New York, NY, 10031, USA.,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA
| | - Jialiang Liang
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA.,Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA
| | - Rebecca Frawley
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA
| | - Jabez Bok
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), 61 Biopolis Drive, Proteos Building #3-06, Singapore, 138673, Singapore
| | - Angeliki Mela
- Department of Pathology and Cell Biology, Columbia University Medical Center, 630 West 168th Street, New York, NY, 10032, USA
| | - Camila Yattah
- Neuroscience Initiative at the Advanced Science Research Center of the Graduate Center of The City University of New York, 85 St. Nicholas Terrace, New York, NY, 10031, USA.,Graduate Program in Biochemistry, The Graduate Center of The City University of New York, 365 5th Avenue, New York, NY, 10016, USA
| | - Jingxian Zhang
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), 61 Biopolis Drive, Proteos Building #3-06, Singapore, 138673, Singapore
| | - Shun Xie Teo
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), 61 Biopolis Drive, Proteos Building #3-06, Singapore, 138673, Singapore
| | - Ting Zhou
- Room A-829, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
| | - Shuibing Chen
- Room A-829, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
| | - Emily Bernstein
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA.,Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY, 10029, USA
| | - Peter Canoll
- Department of Pathology and Cell Biology, Columbia University Medical Center, 630 West 168th Street, New York, NY, 10032, USA
| | - Ernesto Guccione
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA.,Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), 61 Biopolis Drive, Proteos Building #3-06, Singapore, 138673, Singapore.,Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY, 10029, USA
| | - Patrizia Casaccia
- Neuroscience Initiative at the Advanced Science Research Center of the Graduate Center of The City University of New York, 85 St. Nicholas Terrace, New York, NY, 10031, USA. .,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA. .,Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA. .,Graduate Program in Biochemistry, The Graduate Center of The City University of New York, 365 5th Avenue, New York, NY, 10016, USA.
| |
Collapse
|
24
|
Hirsch TZ, Martin-Lannerée S, Reine F, Hernandez-Rapp J, Herzog L, Dron M, Privat N, Passet B, Halliez S, Villa-Diaz A, Lacroux C, Klein V, Haïk S, Andréoletti O, Torres JM, Vilotte JL, Béringue V, Mouillet-Richard S. Epigenetic Control of the Notch and Eph Signaling Pathways by the Prion Protein: Implications for Prion Diseases. Mol Neurobiol 2018; 56:2159-2173. [PMID: 29998397 DOI: 10.1007/s12035-018-1193-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 06/26/2018] [Indexed: 12/12/2022]
Abstract
Among the ever-growing number of self-replicating proteins involved in neurodegenerative diseases, the prion protein PrP remains the most infamous for its central role in transmissible spongiform encephalopathies (TSEs). In these diseases, pathogenic prions propagate through a seeding mechanism, where normal PrPC molecules are converted into abnormally folded scrapie isoforms termed PrPSc. Since its discovery over 30 years ago, much advance has contributed to define the host-encoded cellular prion protein PrPC as a critical relay of prion-induced neuronal cell demise. A current consensual view is that the conversion of PrPC into PrPSc in neuronal cells diverts the former from its normal function with subsequent molecular alterations affecting synaptic plasticity. Here, we report that prion infection is associated with reduced expression of key effectors of the Notch pathway in vitro and in vivo, recapitulating changes fostered by the absence of PrPC. We further show that both prion infection and PrPC depletion promote drastic alterations in the expression of a defined set of Eph receptors and their ephrin ligands, which represent important players in synaptic function. Our data indicate that defects in the Notch and Eph axes can be mitigated in response to histone deacetylase inhibition in PrPC-depleted as well as prion-infected cells. We thus conclude that infectious prions cause a loss-of-function phenotype with respect to Notch and Eph signaling and that these alterations are sustained by epigenetic mechanisms.
Collapse
Affiliation(s)
- Théo Z Hirsch
- INSERM UMR 1124, 75006, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, UMR 1124, 75006, Paris, France
- INSERM U1162, 75010, Paris, France
| | - Séverine Martin-Lannerée
- INSERM UMR 1124, 75006, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, UMR 1124, 75006, Paris, France
| | - Fabienne Reine
- INRA, Université Paris-Saclay, UR 892 Virologie Immunologie Moléculaires, 78350, Jouy-en-Josas, France
| | - Julia Hernandez-Rapp
- INSERM UMR 1124, 75006, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, UMR 1124, 75006, Paris, France
- Centre de Recherche du CHU de Québec, Université Laval, Québec, G1V4G2, Québec, Canada
| | - Laetitia Herzog
- INRA, Université Paris-Saclay, UR 892 Virologie Immunologie Moléculaires, 78350, Jouy-en-Josas, France
| | - Michel Dron
- INRA, Université Paris-Saclay, UR 892 Virologie Immunologie Moléculaires, 78350, Jouy-en-Josas, France
| | - Nicolas Privat
- INSERM UMR 1127, CNRS UMR 7225, 75013, Paris, France
- Institut du Cerveau et de la Moelle épinière, ICM, Inserm U 1127, CNRS UMR 7225, Sorbonne Université, F-75013, Paris, France
| | - Bruno Passet
- INRA UMR1313, Génétique Animale et Biologie Intégrative, 78350, Jouy-en-Josas, France
| | - Sophie Halliez
- INRA, Université Paris-Saclay, UR 892 Virologie Immunologie Moléculaires, 78350, Jouy-en-Josas, France
- INSERM, UMR-S1172, Lille University, 59045, Lille, France
| | - Ana Villa-Diaz
- Centro de Investigación en Sanidad Animal-INIA, 28130, Madrid, Spain
| | | | - Victor Klein
- INSERM UMR 1124, 75006, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, UMR 1124, 75006, Paris, France
| | - Stéphane Haïk
- INSERM UMR 1127, CNRS UMR 7225, 75013, Paris, France
- Institut du Cerveau et de la Moelle épinière, ICM, Inserm U 1127, CNRS UMR 7225, Sorbonne Université, F-75013, Paris, France
| | | | - Juan-Maria Torres
- Centro de Investigación en Sanidad Animal-INIA, 28130, Madrid, Spain
| | - Jean-Luc Vilotte
- INRA UMR1313, Génétique Animale et Biologie Intégrative, 78350, Jouy-en-Josas, France
| | | | - Sophie Mouillet-Richard
- INSERM UMR 1124, 75006, Paris, France.
- Université Paris Descartes, Sorbonne Paris Cité, UMR 1124, 75006, Paris, France.
| |
Collapse
|
25
|
Santos AK, Vieira MS, Vasconcellos R, Goulart VAM, Kihara AH, Resende RR. Decoding cell signalling and regulation of oligodendrocyte differentiation. Semin Cell Dev Biol 2018; 95:54-73. [PMID: 29782926 DOI: 10.1016/j.semcdb.2018.05.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 05/15/2018] [Accepted: 05/17/2018] [Indexed: 12/20/2022]
Abstract
Oligodendrocytes are fundamental for the functioning of the nervous system; they participate in several cellular processes, including axonal myelination and metabolic maintenance for astrocytes and neurons. In the mammalian nervous system, they are produced through waves of proliferation and differentiation, which occur during embryogenesis. However, oligodendrocytes and their precursors continue to be generated during adulthood from specific niches of stem cells that were not recruited during development. Deficiencies in the formation and maturation of these cells can generate pathologies mainly related to myelination. Understanding the mechanisms involved in oligodendrocyte development, from the precursor to mature cell level, will allow inferring therapies and treatments for associated pathologies and disorders. Such mechanisms include cell signalling pathways that involve many growth factors, small metabolic molecules, non-coding RNAs, and transcription factors, as well as specific elements of the extracellular matrix, which act in a coordinated temporal and spatial manner according to a given stimulus. Deciphering those aspects will allow researchers to replicate them in vitro in a controlled environment and thus mimic oligodendrocyte maturation to understand the role of oligodendrocytes in myelination in pathologies and normal conditions. In this study, we review these aspects, based on the most recent in vivo and in vitro data on oligodendrocyte generation and differentiation.
Collapse
Affiliation(s)
- A K Santos
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil
| | - M S Vieira
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil; Instituto Nanocell, Rua Santo Antônio, 420, 35500-041 Divinópolis, MG, Brazil
| | - R Vasconcellos
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil; Instituto Nanocell, Rua Santo Antônio, 420, 35500-041 Divinópolis, MG, Brazil
| | - V A M Goulart
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil
| | - A H Kihara
- Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil
| | - R R Resende
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil; Instituto Nanocell, Rua Santo Antônio, 420, 35500-041 Divinópolis, MG, Brazil.
| |
Collapse
|
26
|
Yang Y, Cheng Z, Tang H, Jiao H, Sun X, Cui Q, Luo F, Pan H, Ma C, Li B. Neonatal Maternal Separation Impairs Prefrontal Cortical Myelination and Cognitive Functions in Rats Through Activation of Wnt Signaling. Cereb Cortex 2018; 27:2871-2884. [PMID: 27178192 DOI: 10.1093/cercor/bhw121] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Adverse early-life experience such as depriving the relationship between parents and children induces permanent phenotypic changes, and impairs the cognitive functions associated with the prefrontal cortex (PFC). However, the underlying mechanism remains unclear. In this work, we used rat neonatal maternal separation (NMS) model to illuminate whether and how NMS in early life affects cognitive functions, and what the underlying cellular and molecular mechanism is. We showed that rat pups separated from their dam 3 h daily during the first 3 postnatal weeks alters medial prefrontal cortex (mPFC) myelination and impairs mPFC-dependent behaviors. Myelination appears necessary for mPFC-dependent behaviors, as blockade of oligodendrocytes (OLs) differentiation or lysolecithin-induced demyelination, impairs mPFC functions. We further demonstrate that histone deacetylases 1/2 (HDAC1/2) are drastically reduced in NMS rats. Inhibition of HDAC1/2 promotes Wnt activation, which negatively regulates OLs development. Conversely, selective inhibition of Wnt signaling by XAV939 partly rescue myelination arrestment and behavior deficiency caused by NMS. These findings indicate that NMS impairs mPFC cognitive functions, at least in part, through modulation of oligodendrogenesis and myelination. Understanding the mechanism of NMS on mPFC-dependent behaviors is critical for developing pharmacological and psychological interventions for child neglect and abuse.
Collapse
Affiliation(s)
- Youjun Yang
- Center for Neuropsychiatric Disorders, Institute of Life Science, Nanchang University, Nanchang 330031, P.R. China
| | - Zongyue Cheng
- Center for Neuropsychiatric Disorders, Institute of Life Science, Nanchang University, Nanchang 330031, P.R. China
| | - Hua Tang
- Center for Neuropsychiatric Disorders, Institute of Life Science, Nanchang University, Nanchang 330031, P.R. China
| | - Huifeng Jiao
- Center for Neuropsychiatric Disorders, Institute of Life Science, Nanchang University, Nanchang 330031, P.R. China
| | - Xuan Sun
- Center for Neuropsychiatric Disorders, Institute of Life Science, Nanchang University, Nanchang 330031, P.R. China
| | - Qiuzhu Cui
- Center for Neuropsychiatric Disorders, Institute of Life Science, Nanchang University, Nanchang 330031, P.R. China
| | - Fei Luo
- Center for Neuropsychiatric Disorders, Institute of Life Science, Nanchang University, Nanchang 330031, P.R. China
| | - Haili Pan
- Center for Neuropsychiatric Disorders, Institute of Life Science, Nanchang University, Nanchang 330031, P.R. China
| | - Chaolin Ma
- Center for Neuropsychiatric Disorders, Institute of Life Science, Nanchang University, Nanchang 330031, P.R. China
| | - Baoming Li
- Center for Neuropsychiatric Disorders, Institute of Life Science, Nanchang University, Nanchang 330031, P.R. China
| |
Collapse
|
27
|
Koreman E, Sun X, Lu QR. Chromatin remodeling and epigenetic regulation of oligodendrocyte myelination and myelin repair. Mol Cell Neurosci 2017; 87:18-26. [PMID: 29254827 DOI: 10.1016/j.mcn.2017.11.010] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 10/27/2017] [Accepted: 11/14/2017] [Indexed: 12/14/2022] Open
Abstract
Oligodendrocytes are essential for the development, function, and health of the vertebrate central nervous system. These cells maintain axon myelination to ensure saltatory propagation of action potentials. Oligodendrocyte develops from neural progenitor cells, in a step-wise process that involves oligodendrocyte precursor specification, proliferation, and differentiation. The lineage progression requires coordination of transcriptional and epigenetic circuits to mediate the stage-specific intricacies of oligodendrocyte development. Epigenetic mechanisms involve DNA methylation, histone modifications, ATP-dependent chromatin remodeling, and non-coding RNA modulation that regulate the chromatin state over regulatory genes, which must be expressed or repressed to establish oligodendrocyte identity and lineage progression. In this review, we will focus on epigenetic programming associated with histone modification enzymes, chromatin remodeling, and non-coding RNAs that regulate oligodendrocyte lineage progression, and discuss how these mechanisms might be harnessed to induce myelin repair for treatment of demyelinating diseases such as multiple sclerosis.
Collapse
Affiliation(s)
- Elijah Koreman
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Xiaowei Sun
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Q Richard Lu
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| |
Collapse
|
28
|
Petersen MA, Ryu JK, Chang KJ, Etxeberria A, Bardehle S, Mendiola AS, Kamau-Devers W, Fancy SPJ, Thor A, Bushong EA, Baeza-Raja B, Syme CA, Wu MD, Rios Coronado PE, Meyer-Franke A, Yahn S, Pous L, Lee JK, Schachtrup C, Lassmann H, Huang EJ, Han MH, Absinta M, Reich DS, Ellisman MH, Rowitch DH, Chan JR, Akassoglou K. Fibrinogen Activates BMP Signaling in Oligodendrocyte Progenitor Cells and Inhibits Remyelination after Vascular Damage. Neuron 2017; 96:1003-1012.e7. [PMID: 29103804 DOI: 10.1016/j.neuron.2017.10.008] [Citation(s) in RCA: 142] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 08/30/2017] [Accepted: 10/04/2017] [Indexed: 12/20/2022]
Abstract
Blood-brain barrier (BBB) disruption alters the composition of the brain microenvironment by allowing blood proteins into the CNS. However, whether blood-derived molecules serve as extrinsic inhibitors of remyelination is unknown. Here we show that the coagulation factor fibrinogen activates the bone morphogenetic protein (BMP) signaling pathway in oligodendrocyte progenitor cells (OPCs) and suppresses remyelination. Fibrinogen induces phosphorylation of Smad 1/5/8 and inhibits OPC differentiation into myelinating oligodendrocytes (OLs) while promoting an astrocytic fate in vitro. Fibrinogen effects are rescued by BMP type I receptor inhibition using dorsomorphin homolog 1 (DMH1) or CRISPR/Cas9 activin A receptor type I (ACVR1) knockout in OPCs. Fibrinogen and the BMP target Id2 are increased in demyelinated multiple sclerosis (MS) lesions. Therapeutic depletion of fibrinogen decreases BMP signaling and enhances remyelination in vivo. Targeting fibrinogen may be an upstream therapeutic strategy to promote the regenerative potential of CNS progenitors in diseases with remyelination failure.
Collapse
Affiliation(s)
- Mark A Petersen
- Gladstone Institutes, San Francisco, CA, USA; Department of Pediatrics, University of California, San Francisco, CA, USA
| | - Jae Kyu Ryu
- Gladstone Institutes, San Francisco, CA, USA
| | - Kae-Jiun Chang
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Ainhoa Etxeberria
- Department of Neurology, University of California, San Francisco, CA, USA
| | | | | | - Wanjiru Kamau-Devers
- Gladstone Institutes, San Francisco, CA, USA; Berkeley City College, Berkeley, CA, USA
| | - Stephen P J Fancy
- Department of Pediatrics, University of California, San Francisco, CA, USA; Department of Neurology, University of California, San Francisco, CA, USA; Newborn Brain Research Institute, University of California, San Francisco, CA, USA
| | - Andrea Thor
- National Center for Microscopy and Imaging Research, Center for Research in Biological Systems, University of California, San Diego, La Jolla, CA, USA
| | - Eric A Bushong
- National Center for Microscopy and Imaging Research, Center for Research in Biological Systems, University of California, San Diego, La Jolla, CA, USA
| | | | | | - Michael D Wu
- Gladstone Institutes, San Francisco, CA, USA; Department of Anesthesia, University of California, San Francisco, CA, USA
| | | | | | - Stephanie Yahn
- Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Lauriane Pous
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Jae K Lee
- Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Christian Schachtrup
- Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Hans Lassmann
- Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Eric J Huang
- Department of Pathology, University of California, San Francisco, CA, USA
| | - May H Han
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Martina Absinta
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Daniel S Reich
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Mark H Ellisman
- National Center for Microscopy and Imaging Research, Center for Research in Biological Systems, University of California, San Diego, La Jolla, CA, USA; Department of Neurosciences, University of California, San Diego, La Jolla, California, USA; Salk Institute for Biological Studies, La Jolla, San Diego, California, USA
| | - David H Rowitch
- Department of Pediatrics, University of California, San Francisco, CA, USA; Department of Neurosurgery, Eli and Edythe Broad Institute for Stem Cell Research and Regeneration Medicine, University of California, San Francisco, CA, USA; Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - Jonah R Chan
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Katerina Akassoglou
- Gladstone Institutes, San Francisco, CA, USA; Department of Neurology, University of California, San Francisco, CA, USA.
| |
Collapse
|
29
|
Leferink PS, Heine VM. The Healthy and Diseased Microenvironments Regulate Oligodendrocyte Properties: Implications for Regenerative Medicine. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 188:39-52. [PMID: 29024633 DOI: 10.1016/j.ajpath.2017.08.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 07/12/2017] [Accepted: 08/01/2017] [Indexed: 02/08/2023]
Abstract
White matter disorders are characterized by deficient myelin or myelin loss, lead to a range of neurologic dysfunctions, and can result in early death. Oligodendrocytes, which are responsible for white matter formation, are the first targets for treatment. However, many studies indicate that failure of white matter repair goes beyond the intrinsic incapacity of oligodendrocytes to (re)generate myelin and that failed interactions with neighboring cells or factors in the diseased microenvironment can underlie white matter defects. Moreover, most of the white matter disorders show specific white matter pathology caused by different disease mechanisms. Herein, we review the factors within the cellular and the extracellular microenvironment regulating oligodendrocyte properties and discuss stem cell tools to identify microenvironmental factors of importance to the development of improved regenerative medicine for patients with white matter disorders.
Collapse
Affiliation(s)
- Prisca S Leferink
- Department of Pediatrics/Child Neurology, VU University Medical Center, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Vivi M Heine
- Department of Pediatrics/Child Neurology, VU University Medical Center, Amsterdam Neuroscience, Amsterdam, the Netherlands; Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
30
|
Forbes TA, Gallo V. All Wrapped Up: Environmental Effects on Myelination. Trends Neurosci 2017; 40:572-587. [PMID: 28844283 PMCID: PMC5671205 DOI: 10.1016/j.tins.2017.06.009] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 06/26/2017] [Indexed: 12/16/2022]
Abstract
To date, studies have demonstrated the dynamic influence of exogenous environmental stimuli on multiple regions of the brain. This environmental influence positively and negatively impacts programs governing myelination, and acts on myelinating oligodendrocyte (OL) cells across the human lifespan. Developmentally, environmental manipulation of OL progenitor cells (OPCs) has profound effects on the establishment of functional cognitive, sensory, and motor programs. Furthermore, central nervous system (CNS) myelin remains an adaptive entity in adulthood, sensitive to environmentally induced structural changes. Here, we discuss the role of environmental stimuli on mechanisms governing programs of CNS myelination under normal and pathological conditions. Importantly, we highlight how these extrinsic cues can influence the intrinsic power of myelin plasticity to promote functional recovery.
Collapse
Affiliation(s)
- Thomas A Forbes
- Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, DC 20010, USA; The George Washington University School of Medicine and Health Sciences, Washington, DC 20037, USA.
| | - Vittorio Gallo
- Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, DC 20010, USA; The George Washington University School of Medicine and Health Sciences, Washington, DC 20037, USA.
| |
Collapse
|
31
|
Mount CW, Monje M. Wrapped to Adapt: Experience-Dependent Myelination. Neuron 2017; 95:743-756. [PMID: 28817797 PMCID: PMC5667660 DOI: 10.1016/j.neuron.2017.07.009] [Citation(s) in RCA: 148] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 07/05/2017] [Accepted: 07/10/2017] [Indexed: 02/03/2023]
Abstract
Activity of the nervous system has long been recognized as a critical modulator of brain structure and function. Influences of experience on the cytoarchitecture and functional connectivity of neurons have been appreciated since the classic work of Hubel and Wiesel (1963; Wiesel and Hubel, 1963a, 1963b). In recent years, a similar structural plasticity has come to light for the myelinated infrastructure of the nervous system. While an innate program of myelin development proceeds independently of nervous system activity, increasing evidence supports a role for activity-dependent, plastic changes in myelin-forming cells that influence myelin structure and neurological function. Accumulating evidence of complementary and likely temporally overlapping activity-independent and activity-dependent modes of myelination are beginning to crystallize in a model of myelin plasticity, with broad implications for neurological function in health and disease.
Collapse
Affiliation(s)
| | - Michelle Monje
- Department of Neurology, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
32
|
Meyers EA, Kessler JA. TGF-β Family Signaling in Neural and Neuronal Differentiation, Development, and Function. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a022244. [PMID: 28130363 DOI: 10.1101/cshperspect.a022244] [Citation(s) in RCA: 133] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Signaling by the transforming growth factor β (TGF-β) family is necessary for proper neural development and function throughout life. Sequential waves of activation, inhibition, and reactivation of TGF-β family members regulate numerous elements of the nervous system from the earliest stages of embryogenesis through adulthood. This review discusses the expression, regulation, and function of TGF-β family members in the central nervous system at various developmental stages, beginning with induction and patterning of the nervous system to their importance in the adult as modulators of inflammatory response and involvement in degenerative diseases.
Collapse
Affiliation(s)
- Emily A Meyers
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - John A Kessler
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| |
Collapse
|
33
|
Uemura MT, Ihara M, Maki T, Nakagomi T, Kaji S, Uemura K, Matsuyama T, Kalaria RN, Kinoshita A, Takahashi R. Pericyte-derived bone morphogenetic protein 4 underlies white matter damage after chronic hypoperfusion. Brain Pathol 2017; 28:521-535. [PMID: 28470822 PMCID: PMC6099372 DOI: 10.1111/bpa.12523] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 04/18/2017] [Indexed: 01/03/2023] Open
Abstract
Subcortical small vessel disease (SVD) is characterized by white matter damage resulting from arteriolosclerosis and chronic hypoperfusion. Transforming growth factor beta 1 (TGFB1) is dysregulated in the hereditary SVD, CARASIL (cerebral autosomal recessive arteriopathy with subcortical infarcts and leukoencephalopathy). However, very little is known about the role of the largest group in the TGFB superfamily - the bone morphogenetic proteins (BMPs) - in SVD pathogenesis. The aim of this study was to characterize signaling abnormalities of BMPs in sporadic SVD. We examined immunostaining of TGFB1 and BMPs (BMP2/BMP4/BMP6/BMP7/BMP9) in a total of 19 post-mortem human brain samples as follows: 7 SVD patients (4 males, 76-90 years old); 6 Alzheimer's disease (AD) patients (2 males, 67-93 years old) and 6 age-matched disease controls (3 males, 68-78 years old). We subsequently investigated the effects of oxygen-glucose deprivation and BMP4 addition on cultured cells. Furthermore, adult mice were subjected to chronic cerebral hypoperfusion using bilateral common carotid artery stenosis, followed by continuous intracerebroventricular infusion of the BMP antagonist, noggin. In the SVD cases, BMP4 was highly expressed in white matter pericytes. Oxygen-glucose deprivation induced BMP4 expression in cultured pericytes in vitro. Recombinant BMP4 increased the number of cultured endothelial cells and pericytes and converted oligodendrocyte precursor cells into astrocytes. Chronic cerebral hypoperfusion in vivo also upregulated BMP4 with concomitant white matter astrogliogenesis and reduced oligodendrocyte lineage cells, both of which were suppressed by intracerebroventricular noggin infusion. Our findings suggest ischemic white matter damage evolves in parallel with BMP4 upregulation in pericytes. BMP4 promotes angiogenesis, but induces astrogliogenesis at the expense of oligodendrocyte precursor cell proliferation and maturation, thereby aggravating white matter damage. This may explain white matter vulnerability to chronic hypoperfusion. The regulation of BMP4 signaling is a potential therapeutic strategy for treating SVD.
Collapse
Affiliation(s)
- Maiko T Uemura
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masafumi Ihara
- Department of Neurology, National Cerebral and Cardiovascular Center Hospital, Osaka, Japan
| | - Takakuni Maki
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takayuki Nakagomi
- Institute for Advanced Medical Sciences, Hyogo College of Medicine, Hyogo, Japan
| | - Seiji Kaji
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kengo Uemura
- Department of Neurology, Ishiki Hospital, Kagoshima, Japan
| | - Tomohiro Matsuyama
- Institute for Advanced Medical Sciences, Hyogo College of Medicine, Hyogo, Japan
| | - Raj N Kalaria
- Institute of Neuroscience, Newcastle University, Campus for Ageing and Vitality, Newcastle Upon Tyne, UK
| | - Ayae Kinoshita
- School of Human Health Sciences, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ryosuke Takahashi
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
34
|
Shultz RB, Wang Z, Nong J, Zhang Z, Zhong Y. Local delivery of thyroid hormone enhances oligodendrogenesis and myelination after spinal cord injury. J Neural Eng 2017; 14:036014. [PMID: 28358726 DOI: 10.1088/1741-2552/aa6450] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Traumatic spinal cord injury (SCI) causes apoptosis of myelin-forming oligodendrocytes (OLs) and demyelination of surviving axons, resulting in conduction failure. Remyelination of surviving denuded axons provides a promising therapeutic target for spinal cord repair. While cell transplantation has demonstrated efficacy in promoting remyelination and functional recovery, the lack of ideal cell sources presents a major obstacle to clinical application. The adult spinal cord contains oligodendrocyte precursor cells and multipotent neural stem/progenitor cells that have the capacity to differentiate into mature, myelinating OLs. However, endogenous oligodendrogenesis and remyelination processes are limited by the upregulation of remyelination-inhibitory molecules in the post-injury microenvironment. Multiple growth factors/molecules have been shown to promote OL differentiation and myelination. APPROACH In this study we screened these therapeutics and found that 3, 3', 5-triiodothyronine (T3) is the most effective in promoting oligodendrogenesis and OL maturation in vitro. However, systemic administration of T3 to achieve therapeutic doses in the injured spinal cord is likely to induce hyperthyroidism, resulting in serious side effects. MAIN RESULTS In this study we developed a novel hydrogel-based drug delivery system for local delivery of T3 to the injury site without eliciting systemic toxicity. SIGNIFICANCE Using a clinically relevant cervical contusion injury model, we demonstrate that local delivery of T3 at doses comparable to safe human doses promoted new mature OL formation and myelination after SCI.
Collapse
Affiliation(s)
- Robert B Shultz
- School of Biomedical Engineering, Science and Health Systems, Drexel University, 3141 Chestnut Street, Philadelphia, PA 19104, United States of America
| | | | | | | | | |
Collapse
|
35
|
Azim K, Angonin D, Marcy G, Pieropan F, Rivera A, Donega V, Cantù C, Williams G, Berninger B, Butt AM, Raineteau O. Pharmacogenomic identification of small molecules for lineage specific manipulation of subventricular zone germinal activity. PLoS Biol 2017; 15:e2000698. [PMID: 28350803 PMCID: PMC5370089 DOI: 10.1371/journal.pbio.2000698] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 02/21/2017] [Indexed: 11/18/2022] Open
Abstract
Strategies for promoting neural regeneration are hindered by the difficulty of manipulating desired neural fates in the brain without complex genetic methods. The subventricular zone (SVZ) is the largest germinal zone of the forebrain and is responsible for the lifelong generation of interneuron subtypes and oligodendrocytes. Here, we have performed a bioinformatics analysis of the transcriptome of dorsal and lateral SVZ in early postnatal mice, including neural stem cells (NSCs) and their immediate progenies, which generate distinct neural lineages. We identified multiple signaling pathways that trigger distinct downstream transcriptional networks to regulate the diversity of neural cells originating from the SVZ. Next, we used a novel in silico genomic analysis, searchable platform-independent expression database/connectivity map (SPIED/CMAP), to generate a catalogue of small molecules that can be used to manipulate SVZ microdomain-specific lineages. Finally, we demonstrate that compounds identified in this analysis promote the generation of specific cell lineages from NSCs in vivo, during postnatal life and adulthood, as well as in regenerative contexts. This study unravels new strategies for using small bioactive molecules to direct germinal activity in the SVZ, which has therapeutic potential in neurodegenerative diseases. The subventricular zone (SVZ) is the largest germinal zone of the postnatal and adult brain. It contains neural stem cells (NSCs) that give rise to neurons and oligodendrocytes (OLs) in a region-specific manner. Here, we use a bioinformatics approach to identify multiple signaling pathways that regulate the diversity of cell lineages that originate from different subregions of the SVZ. We further use a computational-based drug-discovery strategy to identify a catalogue of small molecules that can be used to manipulate the regionalization of the SVZ. We provide proof that, by administration of small molecules in vivo, it is possible to promote the specific generation of neurons and OLs from NSCs in both the postnatal and adult brain, as well as in regenerative contexts after lesion. This study unravels novel strategies for using small bioactive molecules to direct germinal activity in the SVZ, which has therapeutic potential in neurodegenerative diseases.
Collapse
Affiliation(s)
- Kasum Azim
- Brain Research Institute, University of Zürich/ETHZ, Zürich, Switzerland
- Adult Neurogenesis and Cellular Reprogramming, Institute of Physiological Chemistry, University Medical Centre of the Johannes Gutenberg University Mainz, Germany
- Focus Program Translational Neuroscience, Johannes Gutenberg University Mainz, Germany
- * E-mail: (KA); (OR); (AMB)
| | - Diane Angonin
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, Bron, France
| | - Guillaume Marcy
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, Bron, France
| | - Francesca Pieropan
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| | - Andrea Rivera
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| | - Vanessa Donega
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, Bron, France
| | | | - Gareth Williams
- Wolfson Centre for Age-Related Diseases, King's College London, Guy's Campus, London, United Kingdom
| | - Benedikt Berninger
- Adult Neurogenesis and Cellular Reprogramming, Institute of Physiological Chemistry, University Medical Centre of the Johannes Gutenberg University Mainz, Germany
- Focus Program Translational Neuroscience, Johannes Gutenberg University Mainz, Germany
| | - Arthur M. Butt
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
- * E-mail: (KA); (OR); (AMB)
| | - Olivier Raineteau
- Brain Research Institute, University of Zürich/ETHZ, Zürich, Switzerland
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, Bron, France
- * E-mail: (KA); (OR); (AMB)
| |
Collapse
|
36
|
Berghoff SA, Gerndt N, Winchenbach J, Stumpf SK, Hosang L, Odoardi F, Ruhwedel T, Böhler C, Barrette B, Stassart R, Liebetanz D, Dibaj P, Möbius W, Edgar JM, Saher G. Dietary cholesterol promotes repair of demyelinated lesions in the adult brain. Nat Commun 2017; 8:14241. [PMID: 28117328 PMCID: PMC5286209 DOI: 10.1038/ncomms14241] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 12/12/2016] [Indexed: 12/15/2022] Open
Abstract
Multiple Sclerosis (MS) is an inflammatory demyelinating disorder in which remyelination failure contributes to persistent disability. Cholesterol is rate-limiting for myelin biogenesis in the developing CNS; however, whether cholesterol insufficiency contributes to remyelination failure in MS, is unclear. Here, we show the relationship between cholesterol, myelination and neurological parameters in mouse models of demyelination and remyelination. In the cuprizone model, acute disease reduces serum cholesterol levels that can be restored by dietary cholesterol. Concomitant with blood-brain barrier impairment, supplemented cholesterol directly supports oligodendrocyte precursor proliferation and differentiation, and restores the balance of growth factors, creating a permissive environment for repair. This leads to attenuated axon damage, enhanced remyelination and improved motor learning. Remarkably, in experimental autoimmune encephalomyelitis, cholesterol supplementation does not exacerbate disease expression. These findings emphasize the safety of dietary cholesterol in inflammatory diseases and point to a previously unrecognized role of cholesterol in promoting repair after demyelinating episodes. Cholesterol is important for axonal myelination during development. Here the authors show that cholesterol levels are reduced in a cuprizone mouse model of multiple sclerosis and that dietary cholesterol supplementation enhances remyelination and recovery.
Collapse
Affiliation(s)
- Stefan A Berghoff
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Hermann-Rein-Str. 3, 37075 Göttingen, Germany
| | - Nina Gerndt
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Hermann-Rein-Str. 3, 37075 Göttingen, Germany
| | - Jan Winchenbach
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Hermann-Rein-Str. 3, 37075 Göttingen, Germany
| | - Sina K Stumpf
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Hermann-Rein-Str. 3, 37075 Göttingen, Germany
| | - Leon Hosang
- Institute of Neuroimmunology and Institute for Multiple Sclerosis Research, University Medical Centre Göttingen, Waldweg 33, 37073 Göttingen, Germany
| | - Francesca Odoardi
- Institute of Neuroimmunology and Institute for Multiple Sclerosis Research, University Medical Centre Göttingen, Waldweg 33, 37073 Göttingen, Germany
| | - Torben Ruhwedel
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Hermann-Rein-Str. 3, 37075 Göttingen, Germany
| | - Carolin Böhler
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Hermann-Rein-Str. 3, 37075 Göttingen, Germany
| | - Benoit Barrette
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Hermann-Rein-Str. 3, 37075 Göttingen, Germany
| | - Ruth Stassart
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Hermann-Rein-Str. 3, 37075 Göttingen, Germany.,Department of Neuropathology, University Medical Center, Georg-August-University, Robert Koch Str. 40, 37075 Göttingen, Germany
| | - David Liebetanz
- Department of Clinical Neurophysiology, Georg-August University, Robert Koch Str. 40, 37075 Göttingen, Germany
| | - Payam Dibaj
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Hermann-Rein-Str. 3, 37075 Göttingen, Germany
| | - Wiebke Möbius
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Hermann-Rein-Str. 3, 37075 Göttingen, Germany.,Center Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Wilhelmsplatz 1, 37073 Göttingen, Germany
| | - Julia M Edgar
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Hermann-Rein-Str. 3, 37075 Göttingen, Germany.,Applied Neurobiology Group, Institute of Infection, Immunity and Inflammation, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow G12-8TA, UK
| | - Gesine Saher
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Hermann-Rein-Str. 3, 37075 Göttingen, Germany
| |
Collapse
|
37
|
Patel SS, Tomar S, Sharma D, Mahindroo N, Udayabanu M. Targeting sonic hedgehog signaling in neurological disorders. Neurosci Biobehav Rev 2017; 74:76-97. [PMID: 28088536 DOI: 10.1016/j.neubiorev.2017.01.008] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 12/29/2016] [Accepted: 01/07/2017] [Indexed: 12/13/2022]
Abstract
Sonic hedgehog (Shh) signaling influences neurogenesis and neural patterning during the development of central nervous system. Dysregulation of Shh signaling in brain leads to neurological disorders like autism spectrum disorder, depression, dementia, stroke, Parkinson's diseases, Huntington's disease, locomotor deficit, epilepsy, demyelinating disease, neuropathies as well as brain tumors. The synthesis, processing and transport of Shh ligand as well as the localization of its receptors and signal transduction in the central nervous system has been carefully reviewed. Further, we summarize the regulation of small molecule modulators of Shh pathway with potential in neurological disorders. In conclusion, further studies are warranted to demonstrate the potential of positive and negative regulators of the Shh pathway in neurological disorders.
Collapse
Affiliation(s)
- Sita Sharan Patel
- Department of Pharmacy, Jaypee University of Information Technology, Waknaghat 173234, Himachal Pradesh, India
| | - Sunil Tomar
- School of Pharmaceutical Sciences, Shoolini University, Post Box 9, Solan 173212, Himachal Pradesh, India
| | - Diksha Sharma
- School of Pharmaceutical Sciences, Shoolini University, Post Box 9, Solan 173212, Himachal Pradesh, India
| | - Neeraj Mahindroo
- School of Pharmaceutical Sciences, Shoolini University, Post Box 9, Solan 173212, Himachal Pradesh, India
| | - Malairaman Udayabanu
- Department of Pharmacy, Jaypee University of Information Technology, Waknaghat 173234, Himachal Pradesh, India.
| |
Collapse
|
38
|
Traiffort E, Zakaria M, Laouarem Y, Ferent J. Hedgehog: A Key Signaling in the Development of the Oligodendrocyte Lineage. J Dev Biol 2016; 4:jdb4030028. [PMID: 29615592 PMCID: PMC5831774 DOI: 10.3390/jdb4030028] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 08/26/2016] [Accepted: 08/31/2016] [Indexed: 01/11/2023] Open
Abstract
The Hedgehog morphogen aroused an enormous interest since it was characterized as an essential signal for ventral patterning of the spinal cord two decades ago. The pathway is notably implicated in the initial appearance of the progenitors of oligodendrocytes (OPCs), the glial cells of the central nervous system which after maturation are responsible for axon myelination. In accordance with the requirement for Hedgehog signaling in ventral patterning, the earliest identifiable cells in the oligodendrocyte lineage are derived from the ventral ventricular zone of the developing spinal cord and brain. Here, we present the current knowledge about the involvement of Hedgehog signaling in the strict spatial and temporal regulation which characterizes the initiation and progression of the oligodendrocyte lineage. We notably describe the ability of the Hedgehog signaling to tightly orchestrate the appearance of specific combinations of genes in concert with other pathways. We document the molecular mechanisms controlling Hedgehog temporal activity during OPC specification. The contribution of the pathway to aspects of OPC development different from their specification is also highlighted especially in the optic nerve. Finally, we report the data demonstrating that Hedgehog signaling-dependency is not a universal situation for oligodendrocyte generation as evidenced in the dorsal spinal cord in contrast to the dorsal forebrain.
Collapse
Affiliation(s)
- Elisabeth Traiffort
- Neuroprotective, Neuroregenerative and Remyelinating Small Molecules' U1195, INSERM-Université Paris-Sud, Université Paris-Saclay, 80 rue du Général Leclerc, Kremlin-Bicêtre F-94276, France.
| | - Mary Zakaria
- Neuroprotective, Neuroregenerative and Remyelinating Small Molecules' U1195, INSERM-Université Paris-Sud, Université Paris-Saclay, 80 rue du Général Leclerc, Kremlin-Bicêtre F-94276, France.
| | - Yousra Laouarem
- Neuroprotective, Neuroregenerative and Remyelinating Small Molecules' U1195, INSERM-Université Paris-Sud, Université Paris-Saclay, 80 rue du Général Leclerc, Kremlin-Bicêtre F-94276, France.
| | - Julien Ferent
- IRCM, Molecular Biology of Neural Development, 110 Pine Avenue West, Montreal, QC H2W 1R7, Canada.
| |
Collapse
|
39
|
Jiang X, Nardelli J. Cellular and molecular introduction to brain development. Neurobiol Dis 2016; 92:3-17. [PMID: 26184894 PMCID: PMC4720585 DOI: 10.1016/j.nbd.2015.07.007] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2015] [Revised: 07/07/2015] [Accepted: 07/09/2015] [Indexed: 12/13/2022] Open
Abstract
Advances in the study of brain development over the last decades, especially recent findings regarding the evolutionary expansion of the human neocortex, and large-scale analyses of the proteome/transcriptome in the human brain, have offered novel insights into the molecular mechanisms guiding neural maturation, and the pathophysiology of multiple forms of neurological disorders. As a preamble to reviews of this issue, we provide an overview of the cellular, molecular and genetic bases of brain development with an emphasis on the major mechanisms associated with landmarks of normal neural development in the embryonic stage and early postnatal life, including neural stem/progenitor cell proliferation, cortical neuronal migration, evolution and folding of the cerebral cortex, synaptogenesis and neural circuit development, gliogenesis and myelination. We will only briefly depict developmental disorders that result from perturbations of these cellular or molecular mechanisms, and the most common perinatal brain injuries that could disturb normal brain development.
Collapse
Affiliation(s)
- Xiangning Jiang
- Department of Pediatrics, University of California, San Francisco, CA 94158, USA
| | - Jeannette Nardelli
- Inserm, U1141, Paris 75019, France; Université Paris Diderot, Sorbonne Paris Cité, UMRS 1141, Paris 75019, France.
| |
Collapse
|
40
|
Tong M, Andreani T, Krotow A, Gundogan F, de la Monte SM. Potential Contributions of the Tobacco Nicotine-Derived Nitrosamine Ketone to White Matter Molecular Pathology in Fetal Alcohol Spectrum Disorder. ACTA ACUST UNITED AC 2016; 3. [PMID: 28868525 PMCID: PMC5575815 DOI: 10.15436/2377-1348.16.729] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background Fetal alcohol spectrum disorder (FASD) is associated with long-term
deficits in cognitive and motor functions. Previous studies linked
neurodevelopmental abnormalities to increased oxidative stress and white
matter hypotrophy. However, similar effects occur with low-dose nitrosamine
exposures, alcohol abuse correlates with cigarette smoking, and tobacco
smoke contains tobacco-specific nitrosamines, including NNK. Hypothesis Tobacco smoke exposure is a co-factor in FASD. Design Long Evans rat pups were i.p. administered ethanol (2 g/kg) on
postnatal days (P) 2, 4, 6 and/or NNK (2 mg/kg) on P3, P5, and P7 to
simulate third trimester human exposures. Oligodendroglial
myelin-associated, neuroglial, and relevant transcription factor mRNA
transcripts were measured using targeted PCR arrays. Results Ethanol and NNK differentially altered the expression of immature and
mature oligodendroglial, neuronal and astrocytic structural and
plasticity-associated, and various transcription factor genes. NNK’s
effects were broader and more pronounced than ethanol’s, and
additive or synergistic effects of dual exposures impacted expression of all
four categories of genes investigated. Conclusion Developmental exposures to alcohol and NNK (via tobacco smoke)
contribute to sustained abnormalities in brain white matter structure and
function via distinct but overlapping alterations in the expression of genes
that regulate oligodendrocyte survival, maturation and function, neuroglial
structural integrity, and synaptic plasticity. The results support the
hypothesis that smoking may contribute to brain abnormalities associated
with FASD.
Collapse
Affiliation(s)
- Ming Tong
- Department of Medicine, Division of Gastroenterology, and the Liver Research Center Rhode Island Hospital, Providence, RI.,Warren Alpert Medical School of Brown University, Providence, RI
| | - Tomas Andreani
- Department of Medicine, Division of Gastroenterology, and the Liver Research Center Rhode Island Hospital, Providence, RI
| | | | - Fusun Gundogan
- Warren Alpert Medical School of Brown University, Providence, RI.,Department of Pathology, Women and Infants Hospital of Rhode Island, Providence, RI
| | - Suzanne M de la Monte
- Department of Medicine, Division of Gastroenterology, and the Liver Research Center Rhode Island Hospital, Providence, RI.,Warren Alpert Medical School of Brown University, Providence, RI.,Pathobiology Graduate Program, Brown University, Providence, RI.,Departments of Pathology and Neurology, and the Division of Neuropathology, Rhode Island Hospital, Providence, RI
| |
Collapse
|
41
|
Abstract
Oligodendrocyte progenitors respond to biophysical or mechanical signals, and it has been reported that mechanostimulation modulates cell proliferation, migration, and differentiation. Here we report the effect of three mechanical stimuli on mouse oligodendrocyte progenitor differentiation and identify the molecular components of the linker of nucleoskeleton and cytoskeleton (LINC) complex (i.e., SYNE1) as transducers of mechanical signals to the nucleus, where they modulate the deposition of repressive histone marks and heterochromatin formation. The expression levels of LINC components increased during progenitor differentiation and silencing the Syne1 gene resulted in aberrant histone marks deposition, chromatin reorganization and impaired myelination. We conclude that spatial constraints, via the actin cytoskeleton and LINC complex, mediate nuclear changes in oligodendrocyte progenitors that favor a default pathway of differentiation. Significance statement: It is recognized that oligodendrocyte progenitors are mechanosensitive cells. However, the molecular mechanisms translating mechanical stimuli into oligodendrocyte differentiation remain elusive. This study identifies components of the mechanotransduction pathway in the oligodendrocyte lineage.
Collapse
|
42
|
Cole AE, Murray SS, Xiao J. Bone Morphogenetic Protein 4 Signalling in Neural Stem and Progenitor Cells during Development and after Injury. Stem Cells Int 2016; 2016:9260592. [PMID: 27293450 PMCID: PMC4884839 DOI: 10.1155/2016/9260592] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 04/19/2016] [Accepted: 04/26/2016] [Indexed: 01/17/2023] Open
Abstract
Substantial progress has been made in identifying the extracellular signalling pathways that regulate neural stem and precursor cell biology in the central nervous system (CNS). The bone morphogenetic proteins (BMPs), in particular BMP4, are key players regulating neuronal and glial cell development from neural precursor cells in the embryonic, postnatal, and injured CNS. Here we review recent studies on BMP4 signalling in the generation of neurons, astrocytes, and oligodendroglial cells in the CNS. We also discuss putative mechanisms that BMP4 may utilise to influence glial cell development following CNS injury and highlight some questions for further research.
Collapse
Affiliation(s)
- Alistair E. Cole
- Department of Anatomy and Neuroscience, School of Biomedical Sciences, Faculty of Medicine, Dentistry & Health Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Simon S. Murray
- Department of Anatomy and Neuroscience, School of Biomedical Sciences, Faculty of Medicine, Dentistry & Health Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Junhua Xiao
- Department of Anatomy and Neuroscience, School of Biomedical Sciences, Faculty of Medicine, Dentistry & Health Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
43
|
Lopez Juarez A, He D, Richard Lu Q. Oligodendrocyte progenitor programming and reprogramming: Toward myelin regeneration. Brain Res 2016; 1638:209-220. [PMID: 26546966 PMCID: PMC5119932 DOI: 10.1016/j.brainres.2015.10.051] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Revised: 10/05/2015] [Accepted: 10/27/2015] [Indexed: 01/26/2023]
Abstract
Demyelinating diseases such as multiple sclerosis (MS) are among the most disabling and cost-intensive neurological disorders. The loss of myelin in the central nervous system, produced by oligodendrocytes (OLs), impairs saltatory nerve conduction, leading to motor and cognitive deficits. Immunosuppression therapy has a limited efficacy in MS patients, arguing for a paradigm shift to strategies that target OL lineage cells to achieve myelin repair. The inhibitory microenvironment in MS lesions abrogates the expansion and differentiation of resident OL precursor cells (OPCs) into mature myelin-forming OLs. Recent studies indicate that OPCs display a highly plastic ability to differentiate into alternative cell lineages under certain circumstances. Thus, understanding the mechanisms that maintain and control OPC fate and differentiation into mature OLs in a hostile, non-permissive lesion environment may open new opportunities for regenerative therapies. In this review, we will focus on 1) the plasticity of OPCs in terms of their developmental origins, distribution, and differentiation potentials in the normal and injured brain; 2) recent discoveries of extrinsic and intrinsic factors and small molecule compounds that control OPC specification and differentiation; and 3) therapeutic potential for motivation of neural progenitor cells and reprogramming of differentiated cells into OPCs and their likely impacts on remyelination. OL-based therapies through activating regenerative potentials of OPCs or cell replacement offer exciting opportunities for innovative strategies to promote remyelination and neuroprotection in devastating demyelinating diseases like MS. This article is part of a Special Issue entitled SI:NG2-glia(Invited only).
Collapse
Affiliation(s)
- Alejandro Lopez Juarez
- Department of Pediatrics, Divisions of Experimental Hematology and Cancer Biology & Developmental Biology, Cincinnati Children׳s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Danyang He
- Department of Pediatrics, Divisions of Experimental Hematology and Cancer Biology & Developmental Biology, Cincinnati Children׳s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Q Richard Lu
- Department of Pediatrics, Divisions of Experimental Hematology and Cancer Biology & Developmental Biology, Cincinnati Children׳s Hospital Medical Center, Cincinnati, OH 45229, USA.
| |
Collapse
|
44
|
Moyon S, Liang J, Casaccia P. Epigenetics in NG2 glia cells. Brain Res 2016; 1638:183-198. [PMID: 26092401 PMCID: PMC4683112 DOI: 10.1016/j.brainres.2015.06.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 05/11/2015] [Accepted: 06/02/2015] [Indexed: 12/16/2022]
Abstract
The interplay of transcription and epigenetic marks is essential for oligodendrocyte progenitor cell (OPC) proliferation and differentiation during development. Here, we review the recent advances in this field and highlight mechanisms of transcriptional repression and activation involved in OPC proliferation, differentiation and plasticity. We also describe how dysregulation of these epigenetic events may affect demyelinating disorders, and consider potential ways to manipulate NG2 cell behavior through modulation of the epigenome. This article is part of a Special Issue entitled SI:NG2-glia(Invited only).
Collapse
Affiliation(s)
- Sarah Moyon
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jialiang Liang
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Patrizia Casaccia
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Corinne Goldsmith Dickinson Center for Multiple Sclerosis, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
45
|
Wang LC, Almazan G. Cdon, a cell surface protein, mediates oligodendrocyte differentiation and myelination. Glia 2016; 64:1021-33. [PMID: 26988125 DOI: 10.1002/glia.22980] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 02/11/2016] [Indexed: 12/13/2022]
Abstract
During central nervous system development, oligodendrocyte progenitors (OLPs) establish multiple branched processes and axonal contacts to initiate myelination. A complete understanding of the molecular signals implicated in cell surface interaction to initiate myelination/remyelination is currently lacking. The objective of our study was to assess whether Cdon, a cell surface protein that was shown to participate in muscle and neuron cell development, is involved in oligodendrocyte (OLG) differentiation and myelination. Here, we demonstrate that endogenous Cdon protein is expressed in OLPs, increasing in the early differentiation stages and decreasing in mature OLGs. Immunocytochemistry of endogenous Cdon showed localization on both OLG cell membranes and cellular processes exhibiting puncta- or varicosity-like structures. Cdon knockdown with siRNA decreased protein levels by 62% as well as two myelin-specific proteins, MBP and MAG. Conversely, overexpression of full-length rat Cdon increased myelin proteins in OLGs. The complexity of OLGs branching and contact point numbers with axons were also increased in Cdon overexpressing cells growing alone or in coculture with dorsal root ganglion neurons (DRGNs). Furthermore, myelination of DRGNs was decreased when OLPs were transfected with Cdon siRNA. Altogether, our results suggest that Cdon participates in OLG differentiation and myelination, most likely in the initial stages of development.
Collapse
Affiliation(s)
- Li-Chun Wang
- Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir William Osler, Montreal, Quebec, Canada, H3G 1Y6
| | - Guillermina Almazan
- Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir William Osler, Montreal, Quebec, Canada, H3G 1Y6
| |
Collapse
|
46
|
Abstract
In the nervous system, axons transmit information in the form of electrical impulses over long distances. The speed of impulse conduction is enhanced by myelin, a lipid-rich membrane that wraps around axons. Myelin also is required for the long-term health of axons by providing metabolic support. Accordingly, myelin deficiencies are implicated in a wide range of neurodevelopmental and neuropsychiatric disorders, intellectual disabilities, and neurodegenerative conditions. Central nervous system myelin is formed by glial cells called oligodendrocytes. During development, oligodendrocyte precursor cells migrate from their origins to their target axons, extend long membrane processes that wrap axons, and produce the proteins and lipids that provide myelin membrane with its unique characteristics. Myelination is a dynamic process that involves intricate interactions between multiple cell types. Therefore, an in vivo myelination model, such as the zebrafish, which allows for live observation of cell dynamics and cell-to-cell interactions, is well suited for investigating oligodendrocyte development. Zebrafish offer several advantages to investigating myelination, including the use of transgenic reporter lines, live imaging, forward genetic screens, chemical screens, and reverse genetic approaches. This chapter will describe how these tools and approaches have provided new insights into the regulatory mechanisms that guide myelination.
Collapse
Affiliation(s)
- E S Mathews
- University of Colorado School of Medicine, Aurora, CO, United States
| | - B Appel
- University of Colorado School of Medicine, Aurora, CO, United States
| |
Collapse
|
47
|
van Tilborg E, Heijnen CJ, Benders MJ, van Bel F, Fleiss B, Gressens P, Nijboer CH. Impaired oligodendrocyte maturation in preterm infants: Potential therapeutic targets. Prog Neurobiol 2015; 136:28-49. [PMID: 26655283 DOI: 10.1016/j.pneurobio.2015.11.002] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 11/02/2015] [Accepted: 11/18/2015] [Indexed: 12/20/2022]
Abstract
Preterm birth is an evolving challenge in neonatal health care. Despite declining mortality rates among extremely premature neonates, morbidity rates remain very high. Currently, perinatal diffuse white matter injury (WMI) is the most commonly observed type of brain injury in preterm infants and has become an important research area. Diffuse WMI is associated with impaired cognitive, sensory and psychological functioning and is increasingly being recognized as a risk factor for autism-spectrum disorders, ADHD, and other psychological disturbances. No treatment options are currently available for diffuse WMI and the underlying pathophysiological mechanisms are far from being completely understood. Preterm birth is associated with maternal inflammation, perinatal infections and disrupted oxygen supply which can affect the cerebral microenvironment by causing activation of microglia, astrogliosis, excitotoxicity, and oxidative stress. This intricate interplay of events negatively influences oligodendrocyte development, causing arrested oligodendrocyte maturation or oligodendrocyte cell death, which ultimately results in myelination failure in the developing white matter. This review discusses the current state in perinatal WMI research, ranging from a clinical perspective to basic molecular pathophysiology. The complex regulation of oligodendrocyte development in healthy and pathological conditions is described, with a specific focus on signaling cascades that may play a role in WMI. Furthermore, emerging concepts in the field of WMI and issues regarding currently available animal models are put forward. Novel insights into the molecular mechanisms underlying impeded oligodendrocyte maturation in diffuse WMI may aid the development of novel treatment options which are desperately needed to improve the quality-of-life of preterm neonates.
Collapse
Affiliation(s)
- Erik van Tilborg
- Laboratory of Neuroimmunology and Developmental Origins of Disease, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Cobi J Heijnen
- Laboratory of Neuroimmunology, Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Manon J Benders
- Department of Neonatology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Frank van Bel
- Department of Neonatology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Bobbi Fleiss
- Inserm, Paris U1141, France; Université Paris Diderot, Sorbonne Paris Cité, UMRS, Paris 1141, France; Centre for the Developing Brain, Department of Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' Hospital, London, United Kingdom
| | - Pierre Gressens
- Inserm, Paris U1141, France; Université Paris Diderot, Sorbonne Paris Cité, UMRS, Paris 1141, France; Centre for the Developing Brain, Department of Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' Hospital, London, United Kingdom
| | - Cora H Nijboer
- Laboratory of Neuroimmunology and Developmental Origins of Disease, University Medical Center Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
48
|
The Autotaxin-Lysophosphatidic Acid Axis Modulates Histone Acetylation and Gene Expression during Oligodendrocyte Differentiation. J Neurosci 2015; 35:11399-414. [PMID: 26269646 DOI: 10.1523/jneurosci.0345-15.2015] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
UNLABELLED During development, oligodendrocytes (OLGs), the myelinating cells of the CNS, undergo a stepwise progression during which OLG progenitors, specified from neural stem/progenitor cells, differentiate into fully mature myelinating OLGs. This progression along the OLG lineage is characterized by well synchronized changes in morphology and gene expression patterns. The latter have been found to be particularly critical during the early stages of the lineage, and they have been well described to be regulated by epigenetic mechanisms, especially by the activity of the histone deacetylases HDAC1 and HDAC2. The data presented here identify the extracellular factor autotaxin (ATX) as a novel upstream signal modulating HDAC1/2 activity and gene expression in cells of the OLG lineage. Using the zebrafish as an in vivo model system as well as rodent primary OLG cultures, this functional property of ATX was found to be mediated by its lysophospholipase D (lysoPLD) activity, which has been well characterized to generate the lipid signaling molecule lysophosphatidic acid (LPA). More specifically, the lysoPLD activity of ATX was found to modulate HDAC1/2 regulated gene expression during a time window coinciding with the transition from OLG progenitor to early differentiating OLG. In contrast, HDAC1/2 regulated gene expression during the transition from neural stem/progenitor to OLG progenitor appeared unaffected by ATX and its lysoPLD activity. Thus, together, our data suggest that an ATX-LPA-HDAC1/2 axis regulates OLG differentiation specifically during the transition from OLG progenitor to early differentiating OLG and via a molecular mechanism that is evolutionarily conserved from at least zebrafish to rodent. SIGNIFICANCE STATEMENT The formation of the axon insulating and supporting myelin sheath by differentiating oligodendrocytes (OLGs) in the CNS is considered an essential step during vertebrate development. In addition, loss and/or dysfunction of the myelin sheath has been associated with a variety of neurologic diseases in which repair is limited, despite the presence of progenitor cells with the potential to differentiate into myelinating OLGs. This study characterizes the autotaxin-lysophosphatidic acid signaling axis as a modulator of OLG differentiation in vivo in the developing zebrafish and in vitro in rodent OLGs in culture. These findings provide novel insight into the regulation of developmental myelination, and they are likely to lead to advancing studies related to the stimulation of myelin repair under pathologic conditions.
Collapse
|
49
|
Intracellular Protein Shuttling: A Mechanism Relevant for Myelin Repair in Multiple Sclerosis? Int J Mol Sci 2015; 16:15057-85. [PMID: 26151843 PMCID: PMC4519887 DOI: 10.3390/ijms160715057] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 06/23/2015] [Accepted: 06/25/2015] [Indexed: 12/15/2022] Open
Abstract
A prominent feature of demyelinating diseases such as multiple sclerosis (MS) is the degeneration and loss of previously established functional myelin sheaths, which results in impaired signal propagation and axonal damage. However, at least in early disease stages, partial replacement of lost oligodendrocytes and thus remyelination occur as a result of resident oligodendroglial precursor cell (OPC) activation. These cells represent a widespread cell population within the adult central nervous system (CNS) that can differentiate into functional myelinating glial cells to restore axonal functions. Nevertheless, the spontaneous remyelination capacity in the adult CNS is inefficient because OPCs often fail to generate new oligodendrocytes due to the lack of stimulatory cues and the presence of inhibitory factors. Recent studies have provided evidence that regulated intracellular protein shuttling is functionally involved in oligodendroglial differentiation and remyelination activities. In this review we shed light on the role of the subcellular localization of differentiation-associated factors within oligodendroglial cells and show that regulation of intracellular localization of regulatory factors represents a crucial process to modulate oligodendroglial maturation and myelin repair in the CNS.
Collapse
|
50
|
Huang N, Chen D, Wu X, Chen X, Zhang X, Niu J, Shen HY, Xiao L. Aspirin Promotes Oligodendroglial Differentiation Through Inhibition of Wnt Signaling Pathway. Mol Neurobiol 2015; 53:3258-3266. [PMID: 26059811 DOI: 10.1007/s12035-015-9241-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 05/26/2015] [Indexed: 12/31/2022]
|