1,267 results on '"oligodendrocyte differentiation"'
Search Results
2. OPALIN is an LGI1 receptor promoting oligodendrocyte differentiation.
- Author
-
Xiao-Yu Teng, Ping Hu, Cai-Ming Zhang, Qin-Xin Zhang, Guolin Yang, Yan-Yu Zang, Zhi-Xiong Liu, Guiquan Chen, and Yun Stone Shi
- Subjects
- *
MEMBRANE proteins , *CARRIER proteins , *TRANSCRIPTION factors , *AFFINITY chromatography , *WHITE matter (Nerve tissue) - Abstract
Leucine-rich glioma-inactivated protein 1 (LGI1), a secretory protein in the brain, plays a critical role in myelination; dysfunction of this protein leads to hypomyelination and white matter abnormalities (WMAs). Here, we hypothesized that LGI1 may regulate myelination through binding to an unidentified receptor on the membrane of oligodendrocytes (OLs). To search for this hypothetic receptor, we analyzed LGI1 binding proteins through LGI1-3 × FLAG affinity chromatography with mouse brain lysates followed by mass spectrometry. An OL-specific membrane protein, the oligodendrocytic myelin paranodal and inner loop protein (OPALIN), was identified. Conditional knockout (cKO) of OPALIN in the OL lineage caused hypomyelination and WMAs, phenocopying LGI1 deficiency in mice. Biochemical analysis revealed the downregulation of Sox10 and Olig2, transcription factors critical for OL differentiation, further confirming the impaired OL maturation in Opalin cKO mice. Moreover, virus-mediated re-expression of OPALIN successfully restored myelination in Opalin cKO mice. In contrast, re-expression of LGI1-unbound OPALIN_K23A/D26A failed to reverse the hypomyelination phenotype. In conclusion, our study demonstrated that OPALIN on the OL membrane serves as an LGI1 receptor, highlighting the importance of the LGI1/OPALIN complex in orchestrating OL differentiation and myelination. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
3. Trametinib, an anti-tumor drug, promotes oligodendrocytes generation and myelin formation
- Author
-
Yang, Ying, Suo, Na, Cui, Shi-hao, Wu, Xuan, Ren, Xin-yue, Liu, Yin, Guo, Ren, and Xie, Xin
- Published
- 2024
- Full Text
- View/download PDF
4. S100B actions on glial and neuronal cells in the developing brain: an overview.
- Author
-
Hernández-Ortega, Karina, Alejandro Canul-Euan, Arturo, Mario Solis-Paredes, Juan, Borboa-Olivares, Héctor, Reyes-Muñoz, Enrique, Estrada-Gutierrez, Guadalupe, and Camacho-Arroyo, Ignacio
- Subjects
NEUROGLIA ,COCAINE ,OLIGODENDROGLIA ,NEURAL stem cells ,CELL morphology ,DOWN syndrome ,CALCIUM-binding proteins ,WEIGHT gain - Abstract
The S100B is a member of the S100 family of "E" helix-loop-"F" helix structure (EF) hand calcium-binding proteins expressed in diverse glial, selected neuronal, and various peripheral cells, exerting differential effects. In particular, this review compiles descriptions of the detection of S100B in different brain cells localized in specific regions during the development of humans, mice, and rats. Then, it summarizes S100B's actions on the differentiation, growth, and maturation of glial and neuronal cells in humans and rodents. Particular emphasis is placed on S100B regulation of the differentiation and maturation of astrocytes, oligodendrocytes (OL), and the stimulation of dendritic development in serotoninergic and cerebellar neurons during embryogenesis. We also summarized reports that associate morphological alterations (impaired neurite outgrowth, neuronal migration, altered radial glial cell morphology) of specific neural cell groups during neurodevelopment and functional disturbances (slower rate of weight gain, impaired spatial learning) with changes in the expression of S100B caused by different conditions and stimuli as exposure to stress, ethanol, cocaine and congenital conditions such as Down's Syndrome. Taken together, this evidence highlights the impact of the expression and early actions of S100B in astrocytes, OL, and neurons during brain development, which is reflected in the alterations in differentiation, growth, and maturation of these cells. This allows the integration of a spatiotemporal panorama of S100B actions in glial and neuronal cells in the developing brain. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
5. Synaptic vesicle release regulates pre-myelinating oligodendrocyte-axon interactions in a neuron subtype-specific manner.
- Author
-
Gronseth, James R., Nelson, Heather N., Johnson, Taylor L., Mallon, Taryn A., Martell, Madeline R., Pfaffenbach, Katrina A., Duxbury, Bailey B., Henke, John T., Treichel, Anthony J., and Hines, Jacob H.
- Subjects
SYNAPTIC vesicles ,OLIGODENDROGLIA ,CENTRAL nervous system ,NEURAL circuitry ,NEURONS ,CURIOSITY - Abstract
Oligodendrocyte-lineage cells are central nervous system (CNS) glia that perform multiple functions including the selective myelination of some but not all axons. During myelination, synaptic vesicle release from axons promotes sheath stabilization and growth on a subset of neuron subtypes. In comparison, it is unknown if pre-myelinating oligodendrocyte process extensions selectively interact with specific neural circuits or axon subtypes, and whether the formation and stabilization of these neuron-glia interactions involves synaptic vesicle release. In this study, we used fluorescent reporters in the larval zebrafish model to track pre-myelinating oligodendrocyte process extensions interacting with spinal axons utilizing in vivo imaging. Monitoring motile oligodendrocyte processes and their interactions with individually labeled axons revealed that synaptic vesicle release regulates the behavior of subsets of process extensions. Specifically, blocking synaptic vesicle release decreased the longevity of oligodendrocyte process extensions interacting with reticulospinal axons. Furthermore, blocking synaptic vesicle release increased the frequency that new interactions formed and retracted. In contrast, tracking the movements of all process extensions of singly-labeled oligodendrocytes revealed that synaptic vesicle release does not regulate overall process motility or exploratory behavior. Blocking synaptic vesicle release influenced the density of oligodendrocyte process extensions interacting with reticulospinal and serotonergic axons, but not commissural interneuron or dopaminergic axons. Taken together, these data indicate that alterations to synaptic vesicle release cause changes to oligodendrocyte-axon interactions that are neuron subtype specific. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
6. MicroRNAs dysregulated in multiple sclerosis affect the differentiation of CG-4 cells, an oligodendrocyte progenitor cell line.
- Author
-
Perdaens, Océane, Bottemanne, Pauline, and van Pesch, Vincent
- Subjects
CELL differentiation ,PROGENITOR cells ,MULTIPLE sclerosis ,OLIGODENDROGLIA ,CELL lines ,MICRORNA ,RHINORRHEA - Abstract
Introduction: Demyelination is one of the hallmarks of multiple sclerosis (MS). While remyelination occurs during the disease, it is incomplete from the start and strongly decreases with its progression, mainly due to the harm to oligodendrocyte progenitor cells (OPCs), causing irreversible neurological deficits and contributing to neurodegeneration. Therapeutic strategies promoting remyelination are still very preliminary and lacking within the current treatment panel for MS. Methods: In a previous study, we identified 21 microRNAs dysregulated mostly in the CSF of relapsing and/or remitting MS patients. In this study we transfected the mimics/inhibitors of several of these microRNAs separately in an OPC cell line, called CG-4. We aimed (1) to phenotypically characterize their effect on OPC differentiation and (2) to identify corroborating potential mRNA targets via immunocytochemistry, RT-qPCR analysis, RNA sequencing, and Gene Ontology enrichment analysis. Results: We observed that the majority of 13 transfected microRNA mimics decreased the differentiation of CG-4 cells. We demonstrate, by RNA sequencing and independent RT-qPCR analyses, that miR-33-3p, miR-34c-5p, and miR-124-5p arrest OPC differentiation at a late progenitor stage and miR-145-5p at a premyelinating stage as evidenced by the downregulation of premyelinating oligodendrocyte (OL) [Tcf7l2, Cnp (except for miR-145-5p)] and mature OL (Plp1, Mbp, and Mobp) markers, whereas only miR-214-3p promotes OPC differentiation. We further propose a comprehensive exploration of their change in cell fate through Gene Ontology enrichment analysis. We finally confirm by RT-qPCR analyses the downregulation of several predicted mRNA targets for each microRNA that possibly support their effect on OPC differentiation by very distinctive mechanisms, of which some are still unexplored in OPC/OL physiology. Conclusion: miR-33-3p, miR-34c-5p, and miR-124-5p arrest OPC differentiation at a late progenitor stage and miR-145-5p at a premyelinating stage, whereas miR-214-3p promotes the differentiation of CG-4 cells. We propose several potential mRNA targets and hypothetical mechanisms by which each microRNA exerts its effect. We hereby open new perspectives in the research on OPC differentiation and the pathophysiology of demyelination/remyelination, and possibly even in the search for new remyelinating therapeutic strategies in the scope of MS. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
7. S100B actions on glial and neuronal cells in the developing brain: an overview
- Author
-
Karina Hernández-Ortega, Arturo Alejandro Canul-Euan, Juan Mario Solis-Paredes, Héctor Borboa-Olivares, Enrique Reyes-Muñoz, Guadalupe Estrada-Gutierrez, and Ignacio Camacho-Arroyo
- Subjects
S100B ,astrocyte differentiation ,oligodendrocyte differentiation ,neurite outgrowth ,neurodevelopment ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
The S100B is a member of the S100 family of “E” helix–loop- “F” helix structure (EF) hand calcium-binding proteins expressed in diverse glial, selected neuronal, and various peripheral cells, exerting differential effects. In particular, this review compiles descriptions of the detection of S100B in different brain cells localized in specific regions during the development of humans, mice, and rats. Then, it summarizes S100B’s actions on the differentiation, growth, and maturation of glial and neuronal cells in humans and rodents. Particular emphasis is placed on S100B regulation of the differentiation and maturation of astrocytes, oligodendrocytes (OL), and the stimulation of dendritic development in serotoninergic and cerebellar neurons during embryogenesis. We also summarized reports that associate morphological alterations (impaired neurite outgrowth, neuronal migration, altered radial glial cell morphology) of specific neural cell groups during neurodevelopment and functional disturbances (slower rate of weight gain, impaired spatial learning) with changes in the expression of S100B caused by different conditions and stimuli as exposure to stress, ethanol, cocaine and congenital conditions such as Down’s Syndrome. Taken together, this evidence highlights the impact of the expression and early actions of S100B in astrocytes, OL, and neurons during brain development, which is reflected in the alterations in differentiation, growth, and maturation of these cells. This allows the integration of a spatiotemporal panorama of S100B actions in glial and neuronal cells in the developing brain.
- Published
- 2024
- Full Text
- View/download PDF
8. Synaptic vesicle release regulates pre-myelinating oligodendrocyte-axon interactions in a neuron subtype-specific manner
- Author
-
James R. Gronseth, Heather N. Nelson, Taylor L. Johnson, Taryn A. Mallon, Madeline R. Martell, Katrina A. Pfaffenbach, Bailey B. Duxbury, John T. Henke, Anthony J. Treichel, and Jacob H. Hines
- Subjects
oligodendrocyte ,synaptic vesicle release ,oligodendrocyte differentiation ,myelination ,myelin ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Oligodendrocyte-lineage cells are central nervous system (CNS) glia that perform multiple functions including the selective myelination of some but not all axons. During myelination, synaptic vesicle release from axons promotes sheath stabilization and growth on a subset of neuron subtypes. In comparison, it is unknown if pre-myelinating oligodendrocyte process extensions selectively interact with specific neural circuits or axon subtypes, and whether the formation and stabilization of these neuron–glia interactions involves synaptic vesicle release. In this study, we used fluorescent reporters in the larval zebrafish model to track pre-myelinating oligodendrocyte process extensions interacting with spinal axons utilizing in vivo imaging. Monitoring motile oligodendrocyte processes and their interactions with individually labeled axons revealed that synaptic vesicle release regulates the behavior of subsets of process extensions. Specifically, blocking synaptic vesicle release decreased the longevity of oligodendrocyte process extensions interacting with reticulospinal axons. Furthermore, blocking synaptic vesicle release increased the frequency that new interactions formed and retracted. In contrast, tracking the movements of all process extensions of singly-labeled oligodendrocytes revealed that synaptic vesicle release does not regulate overall process motility or exploratory behavior. Blocking synaptic vesicle release influenced the density of oligodendrocyte process extensions interacting with reticulospinal and serotonergic axons, but not commissural interneuron or dopaminergic axons. Taken together, these data indicate that alterations to synaptic vesicle release cause changes to oligodendrocyte-axon interactions that are neuron subtype specific.
- Published
- 2024
- Full Text
- View/download PDF
9. Transient regulation of focal adhesion via Tensin3 is required for nascent oligodendrocyte differentiation
- Author
-
Merour, Emeric, Hmidan, Hatem, Marie, Corentine, Helou, Pierre-Henri, Lu, Haiyang, Potel, Antoine, Hure, Jean-Baptiste, Clavairoly, Adrien, Shih, Yi Ping, Goudarzi, Salman, Dussaud, Sebastien, Ravassard, Philippe, Hafizi, Sassan, Lo, Su Hao, Hassan, Bassem A, and Parras, Carlos
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Stem Cell Research - Nonembryonic - Non-Human ,Neurosciences ,Regenerative Medicine ,Stem Cell Research ,Genetics ,Humans ,Animals ,Mice ,Focal Adhesions ,Tumor Suppressor Protein p53 ,Oligodendroglia ,Cell Differentiation ,Mice ,Knockout ,Transcription Factors ,Chromatin ,Integrins ,oligodendrocyte differentiation ,tensin ,oligodendroglial survival ,immature oligodedrocyte marker ,Human ,Mouse ,developmental biology ,human ,mouse ,neuroscience ,Biological sciences ,Biomedical and clinical sciences ,Health sciences - Abstract
The differentiation of oligodendroglia from oligodendrocyte precursor cells (OPCs) to complex and extensive myelinating oligodendrocytes (OLs) is a multistep process that involves large-scale morphological changes with significant strain on the cytoskeleton. While key chromatin and transcriptional regulators of differentiation have been identified, their target genes responsible for the morphological changes occurring during OL myelination are still largely unknown. Here, we show that the regulator of focal adhesion, Tensin3 (Tns3), is a direct target gene of Olig2, Chd7, and Chd8, transcriptional regulators of OL differentiation. Tns3 is transiently upregulated and localized to cell processes of immature OLs, together with integrin-β1, a key mediator of survival at this transient stage. Constitutive Tns3 loss of function leads to reduced viability in mouse and humans, with surviving knockout mice still expressing Tns3 in oligodendroglia. Acute deletion of Tns3 in vivo, either in postnatal neural stem cells (NSCs) or in OPCs, leads to a twofold reduction in OL numbers. We find that the transient upregulation of Tns3 is required to protect differentiating OPCs and immature OLs from cell death by preventing the upregulation of p53, a key regulator of apoptosis. Altogether, our findings reveal a specific time window during which transcriptional upregulation of Tns3 in immature OLs is required for OL differentiation likely by mediating integrin-β1 survival signaling to the actin cytoskeleton as OL undergo the large morphological changes required for their terminal differentiation.
- Published
- 2022
10. Prenatal lipopolysaccharide exposure induces anxiety-like behaviour in male mouse offspring and aberrant glial differentiation of embryonic neural stem cells
- Author
-
Chie-Pein Chen, Pei-Chun Chen, Yu-Ling Pan, and Yi-Chao Hsu
- Subjects
Lipopolysaccharide ,Prenatal infection ,Embryonic stem cell ,Neural stem cell ,Oligodendrocyte differentiation ,ApoB ,Cytology ,QH573-671 - Abstract
Abstract Background Prenatal infection has been implicated in the development of neuropsychiatric disorders in children. We hypothesised that exposure to lipopolysaccharide during prenatal development could induce anxiety-like behaviour and sensorineural hearing loss in offspring, as well as disrupt neural differentiation during embryonic neural development. Methods We simulated prenatal infection in FVB mice and mouse embryonic stem cell (ESC) lines, specifically 46C and E14Tg2a, through lipopolysaccharide treatment. Gene expression profiling analyses and behavioural tests were utilized to study the effects of lipopolysaccharide on the offspring and alterations in toll-like receptor (TLR) 2-positive and TLR4-positive cells during neural differentiation in the ESCs. Results Exposure to lipopolysaccharide (25 µg/kg) on gestation day 9 resulted in anxiety-like behaviour specifically in male offspring, while no effects were detected in female offspring. We also found significant increases in the expression of GFAP and CNPase, as well as higher numbers of GFAP + astrocytes and O4+ oligodendrocytes in the prefrontal cortex of male offspring. Furthermore, increased scores for genes related to oligodendrocyte and lipid metabolism, particularly ApoE, were observed in the prefrontal cortex regions. Upon exposure to lipopolysaccharide during the ESC-to-neural stem cell (NSC) transition, Tuj1, Map2, Gfap, O4, and Oligo2 mRNA levels increased in the differentiated neural cells on day 14. In vitro experiments demonstrated that lipopolysaccharide exposure induced inflammatory responses, as evidenced by increased expression of IL1b and ApoB mRNA. Conclusions Our findings suggest that prenatal infection at different stages of neural differentiation may result in distinct disturbances in neural differentiation during ESC—NSC transitions. Furthermore, early prenatal challenges with lipopolysaccharide selectively induce anxiety-like behaviour in male offspring. This behaviour may be attributed to the abnormal differentiation of astrocytes and oligodendrocytes in the brain, potentially mediated by ApoB/E signalling pathways in response to inflammatory stimuli.
- Published
- 2023
- Full Text
- View/download PDF
11. A2B Adenosine Receptor as a New and Attractive Target to Treat Brain Ischemia or Demyelination
- Author
-
Cherchi, Federica, Venturini, Martina, Dettori, Ilaria, Pedata, Felicita, Coppi, Elisabetta, Pugliese, Anna Maria, Bernstein, Peter R., Series Editor, Garner, Amanda L., Series Editor, Georg, Gunda I., Series Editor, Laufer, Stefan, Series Editor, Lowe, John A., Series Editor, Meanwell, Nicholas A., Series Editor, Saxena, Anil Kumar, Series Editor, Supuran, Claudiu T., Series Editor, Zhang, Ao, Series Editor, Tschammer, Nuska, Series Editor, Poulsen, Sally-Ann, Series Editor, and Colotta, Vittoria, editor
- Published
- 2023
- Full Text
- View/download PDF
12. The Wnt Effector TCF7l2 Promotes Oligodendroglial Differentiation by Repressing Autocrine BMP4-Mediated Signaling
- Author
-
Zhang, Sheng, Wang, Yan, Zhu, Xiaoqing, Song, Lanying, Zhan, Xinhua, Ma, Edric, McDonough, Jennifer, Fu, Hui, Cambi, Franca, Grinspan, Judith, and Guo, Fuzheng
- Subjects
Genetics ,Regenerative Medicine ,Neurodegenerative ,Brain Disorders ,Neurosciences ,Autoimmune Disease ,Multiple Sclerosis ,Aetiology ,2.1 Biological and endogenous factors ,Neurological ,Animals ,Autocrine Communication ,Bone Morphogenetic Protein 4 ,Brain ,Cell Differentiation ,Gene Expression Regulation ,Mice ,Mice ,Inbred C57BL ,Mice ,Transgenic ,Neurogenesis ,Oligodendrocyte Precursor Cells ,Oligodendroglia ,Transcription Factor 7-Like 2 Protein ,BMP4 repression ,canonical Wnt/beta-catenin ,myelination ,oligodendrocyte differentiation ,TCF7l2/TCF4 ,Wnt effector ,canonical Wnt/β-catenin ,Medical and Health Sciences ,Psychology and Cognitive Sciences ,Neurology & Neurosurgery - 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.
- Published
- 2021
13. Prenatal lipopolysaccharide exposure induces anxiety-like behaviour in male mouse offspring and aberrant glial differentiation of embryonic neural stem cells.
- Author
-
Chen, Chie-Pein, Chen, Pei-Chun, Pan, Yu-Ling, and Hsu, Yi-Chao
- Abstract
Background: Prenatal infection has been implicated in the development of neuropsychiatric disorders in children. We hypothesised that exposure to lipopolysaccharide during prenatal development could induce anxiety-like behaviour and sensorineural hearing loss in offspring, as well as disrupt neural differentiation during embryonic neural development. Methods: We simulated prenatal infection in FVB mice and mouse embryonic stem cell (ESC) lines, specifically 46C and E14Tg2a, through lipopolysaccharide treatment. Gene expression profiling analyses and behavioural tests were utilized to study the effects of lipopolysaccharide on the offspring and alterations in toll-like receptor (TLR) 2-positive and TLR4-positive cells during neural differentiation in the ESCs. Results: Exposure to lipopolysaccharide (25 µg/kg) on gestation day 9 resulted in anxiety-like behaviour specifically in male offspring, while no effects were detected in female offspring. We also found significant increases in the expression of GFAP and CNPase, as well as higher numbers of GFAP + astrocytes and O4+ oligodendrocytes in the prefrontal cortex of male offspring. Furthermore, increased scores for genes related to oligodendrocyte and lipid metabolism, particularly ApoE, were observed in the prefrontal cortex regions. Upon exposure to lipopolysaccharide during the ESC-to-neural stem cell (NSC) transition, Tuj1, Map2, Gfap, O4, and Oligo2 mRNA levels increased in the differentiated neural cells on day 14. In vitro experiments demonstrated that lipopolysaccharide exposure induced inflammatory responses, as evidenced by increased expression of IL1b and ApoB mRNA. Conclusions: Our findings suggest that prenatal infection at different stages of neural differentiation may result in distinct disturbances in neural differentiation during ESC—NSC transitions. Furthermore, early prenatal challenges with lipopolysaccharide selectively induce anxiety-like behaviour in male offspring. This behaviour may be attributed to the abnormal differentiation of astrocytes and oligodendrocytes in the brain, potentially mediated by ApoB/E signalling pathways in response to inflammatory stimuli. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
14. Phototherapy Alters the Plasma Metabolite Profile in Infants Born Preterm with Hyperbilirubinemia.
- Author
-
Satrom, Katherine M., Wang, Jiuzhou, Lock, Eric F., Snook, Kirsten, Lund, Troy C., and Rao, Raghavendra B.
- Published
- 2024
- Full Text
- View/download PDF
15. The Stem Cell Factor Sox2 Is a Positive Timer of Oligodendrocyte Development in the Postnatal Murine Spinal Cord.
- Author
-
Zhang, Sheng, Rasai, Abeer, Wang, Yan, Xu, Jie, Bannerman, Peter, Erol, Daffcar, Tsegaye, Danayit, Wang, Aijun, Soulika, Athena, Zhan, Xiangjiang, and Guo, Fuzheng
- Subjects
Spinal Cord ,Oligodendroglia ,Myelin Sheath ,Stem Cells ,Animals ,Animals ,Newborn ,Mice ,Transgenic ,Tamoxifen ,Integrases ,Cell Differentiation ,Cell Proliferation ,Cell Movement ,Gene Expression Regulation ,Gene Deletion ,Mutation ,Time Factors ,SOXB1 Transcription Factors ,Myelination ,Neural stem cells ,Oligodendrocyte differentiation ,Oligodendrocyte progenitor cells ,Proliferation ,Sox2 ,Neurosciences ,Multiple Sclerosis ,Brain Disorders ,Pediatric ,Neurodegenerative ,Regenerative Medicine ,Stem Cell Research ,Stem Cell Research - Nonembryonic - Non-Human ,Neurological ,Psychology ,Cognitive Sciences ,Neurology & Neurosurgery - Abstract
Myelination in the central nervous system takes place predominantly during the postnatal development of humans and rodents by myelinating oligodendrocytes (OLs), which are differentiated from oligodendrocyte progenitor cells (OPCs). We recently reported that Sox2 is essential for developmental myelination in the murine brain and spinal cord. It is still controversial regarding the role of Sox2 in oligodendroglial lineage progression in the postnatal murine spinal cord. Analyses of a series of cell- and stage-specific Sox2 mutants reveal that Sox2 plays a biphasic role in regulating oligodendroglial lineage progression in the postnatal murine spinal cord. Sox2 controls the number of OPCs for subsequent differentiation through regulating their proliferation. In addition, Sox2 regulates the timing of OL differentiation and modulates the rate of oligodendrogenesis. Our experimental data prove that Sox2 is an intrinsic positive timer of oligodendroglial lineage progression and suggest that interventions affecting oligodendroglial Sox2 expression may be therapeutic for overcoming OPC differentiation arrest in dysmyelinating and demyelinating disorders.
- Published
- 2018
16. Sox2 Is Essential for Oligodendroglial Proliferation and Differentiation during Postnatal Brain Myelination and CNS Remyelination
- Author
-
Zhang, Sheng, Zhu, Xiaoqing, Gui, Xuehong, Croteau, Christopher, Song, Lanying, Xu, Jie, Wang, Aijun, Bannerman, Peter, and Guo, Fuzheng
- Subjects
Biomedical and Clinical Sciences ,Neurosciences ,Stem Cell Research ,Multiple Sclerosis ,Stem Cell Research - Nonembryonic - Non-Human ,Regenerative Medicine ,Autoimmune Disease ,Pediatric ,Brain Disorders ,Neurodegenerative ,Genetics ,1.1 Normal biological development and functioning ,2.1 Biological and endogenous factors ,Neurological ,Animals ,Brain ,Cell Differentiation ,Cell Proliferation ,Central Nervous System ,Demyelinating Diseases ,Female ,Male ,Mice ,Mice ,Inbred C57BL ,Mice ,Knockout ,Mice ,Transgenic ,Movement Disorders ,Myelin Sheath ,Nerve Regeneration ,Oligodendroglia ,SOXB1 Transcription Factors ,Spinal Cord ,Transcriptome ,myelination and remyelination ,oligodendrocyte differentiation ,oligodendrocyte regeneration ,oligodendroglial lineage progression ,oligodendroglial progenitor cells ,Sox2 ,Medical and Health Sciences ,Psychology and Cognitive Sciences ,Neurology & Neurosurgery - Abstract
In the CNS, myelination and remyelination depend on the successful progression and maturation of oligodendroglial lineage cells, including proliferation and differentiation of oligodendroglial progenitor cells (OPCs). Previous studies have reported that Sox2 transiently regulates oligodendrocyte (OL) differentiation in the embryonic and perinatal spinal cord and appears dispensable for myelination in the postnatal spinal cord. However, the role of Sox2 in OL development in the brain has yet to be defined. We now report that Sox2 is an essential positive regulator of developmental myelination in the postnatal murine brain of both sexes. Stage-specific paradigms of genetic disruption demonstrated that Sox2 regulated brain myelination by coordinating upstream OPC population supply and downstream OL differentiation. Transcriptomic analyses further supported a crucial role of Sox2 in brain developmental myelination. Consistently, oligodendroglial Sox2-deficient mice developed severe tremors and ataxia, typical phenotypes indicative of hypomyelination, and displayed severe impairment of motor function and prominent deficits of brain OL differentiation and myelination persisting into the later CNS developmental stages. We also found that Sox2 was required for efficient OPC proliferation and expansion and OL regeneration during remyelination in the adult brain and spinal cord. Together, our genetic evidence reveals an essential role of Sox2 in brain myelination and CNS remyelination, and suggests that manipulation of Sox2 and/or Sox2-mediated downstream pathways may be therapeutic in promoting CNS myelin repair.SIGNIFICANCE STATEMENT Promoting myelin formation and repair has translational significance in treating myelin-related neurological disorders, such as periventricular leukomalacia and multiple sclerosis in which brain developmental myelin formation and myelin repair are severely affected, respectively. In this report, analyses of a series of genetic conditional knock-out systems targeting different oligodendrocyte stages reveal a previously unappreciated role of Sox2 in coordinating upstream proliferation and downstream differentiation of oligodendroglial lineage cells in the mouse brain during developmental myelination and CNS remyelination. Our study points to the potential of manipulating Sox2 and its downstream pathways to promote oligodendrocyte regeneration and CNS myelin repair.
- Published
- 2018
17. High Dose Pharmaceutical Grade Biotin (MD1003) Accelerates Differentiation of Murine and Grafted Human Oligodendrocyte Progenitor Cells In Vivo.
- Author
-
Levy, Marion J. F., Garcia-Diaz, Beatriz, Sedel, Frédéric, Baron-Van Evercooren, Anne, and Mozafari, Sabah
- Subjects
- *
OLIGODENDROGLIA , *PROGENITOR cells , *BIOTIN , *BREAST milk , *AMYOTROPHIC lateral sclerosis , *DEMYELINATION , *MYELINATION , *GRAFTING (Horticulture) - Abstract
Accumulating evidences suggest a strong correlation between metabolic changes and neurodegeneration in CNS demyelinating diseases such as multiple sclerosis (MS). Biotin, an essential cofactor for five carboxylases, is expressed by oligodendrocytes and involved in fatty acid synthesis and energy production. The metabolic effect of biotin or high-dose-biotin (MD1003) has been reported on rodent oligodendrocytes in vitro, and in neurodegenerative or demyelinating animal models. However, clinical studies, showed mild or no beneficial effect of MD1003 in amyotrophic lateral sclerosis (ALS) or MS. Here, we took advantage of a mouse model of myelin deficiency to study the effects of MD1003 on the behavior of murine and grafted human oligodendrocytes in vivo. We show that MD1003 increases the number and the differentiation potential of endogenous murine oligodendroglia over time. Moreover, the levels of MD1003 are increased in the plasma and brain of pups born to treated mothers, indicating that MD1003 can pass through the mother's milk. The histological analysis of the grafted animals shows that MD1003 increased proliferation and accelerates differentiation of human oligodendroglia, but without enhancing their myelination potential. These findings provide important insights into the role of MD1003 on murine and human oligodendrocyte maturation/myelination that may explain the mitigated outcome of ALS/MS clinical trials. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
18. Transient regulation of focal adhesion via Tensin3 is required for nascent oligodendrocyte differentiation
- Author
-
Emeric Merour, Hatem Hmidan, Corentine Marie, Pierre-Henri Helou, Haiyang Lu, Antoine Potel, Jean-Baptiste Hure, Adrien Clavairoly, Yi Ping Shih, Salman Goudarzi, Sebastien Dussaud, Philippe Ravassard, Sassan Hafizi, Su Hao Lo, Bassem A Hassan, and Carlos Parras
- Subjects
oligodendrocyte differentiation ,tensin ,oligodendroglial survival ,immature oligodedrocyte marker ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
The differentiation of oligodendroglia from oligodendrocyte precursor cells (OPCs) to complex and extensive myelinating oligodendrocytes (OLs) is a multistep process that involves large-scale morphological changes with significant strain on the cytoskeleton. While key chromatin and transcriptional regulators of differentiation have been identified, their target genes responsible for the morphological changes occurring during OL myelination are still largely unknown. Here, we show that the regulator of focal adhesion, Tensin3 (Tns3), is a direct target gene of Olig2, Chd7, and Chd8, transcriptional regulators of OL differentiation. Tns3 is transiently upregulated and localized to cell processes of immature OLs, together with integrin-β1, a key mediator of survival at this transient stage. Constitutive Tns3 loss of function leads to reduced viability in mouse and humans, with surviving knockout mice still expressing Tns3 in oligodendroglia. Acute deletion of Tns3 in vivo, either in postnatal neural stem cells (NSCs) or in OPCs, leads to a twofold reduction in OL numbers. We find that the transient upregulation of Tns3 is required to protect differentiating OPCs and immature OLs from cell death by preventing the upregulation of p53, a key regulator of apoptosis. Altogether, our findings reveal a specific time window during which transcriptional upregulation of Tns3 in immature OLs is required for OL differentiation likely by mediating integrin-β1 survival signaling to the actin cytoskeleton as OL undergo the large morphological changes required for their terminal differentiation.
- Published
- 2022
- Full Text
- View/download PDF
19. Oligodendrocyte precursor cell maturation: role of adenosine receptors
- Author
-
Federica Cherchi, Anna Maria Pugliese, and Elisabetta Coppi
- Subjects
adenosine receptors ,k+ channels ,oligodendrocyte differentiation ,oligodendrocyte progenitor cells ,remyelination ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Oligodendrocyte-formed myelin sheaths allow fast synaptic transmission in the brain and their degeneration leads to demyelinating diseases such as multiple sclerosis. Remyelination requires the differentiation of oligodendrocyte progenitor cells into mature oligodendrocytes but, in chronic neurodegenerative disorders, remyelination fails due to adverse environment. Therefore, a strategy to prompt oligodendrocyte progenitor cell differentiation towards myelinating oligodendrocytes is required. The neuromodulator adenosine, and its receptors (A1, A2A, A2B and A3 receptors: A1R, A2AR, A2BR and A3R), are crucial mediators in remyelination processes. It is known that A1Rs facilitate oligodendrocyte progenitor cell maturation and migration whereas the A3Rs initiates apoptosis in oligodendrocyte progenitor cells. Our group of research contributed to the field by demonstrating that A2AR and A2BR inhibit oligodendrocyte progenitor cell maturation by reducing voltage-dependent K+ currents necessary for cell differentiation. The present review summarizes the possible role of adenosine receptor ligands as potential therapeutic targets in demyelinating pathologies such as multiple sclerosis.
- Published
- 2021
- Full Text
- View/download PDF
20. A novel Lgi1 mutation causes white matter abnormalities and impairs motor coordination in mice.
- Author
-
Teng, Xiao‐Yu, Hu, Ping, Chen, Yangyang, Zang, Yanyu, Ye, Xiaolian, Ou, Jingmin, Chen, Guiquan, and Shi, Yun Stone
- Abstract
Leucine‐rich glioma‐inactivated protein 1 (LGI1) is known to play a key role in autosomal dominant lateral temporal lobe epilepsy (ADLTE). The ADLTE is an inherited disease characterized by focal seizures with distinctive auditory or aphasic symptoms. A large number of mutations on the Lgi1 gene have been reported and are believed to be the genetic cause for ADLTE. We identified a novel missense mutation, c.152A>G (p.Asp51Gly), on Lgi1 from a Chinese ADLTE patient who manifests locomotor imbalance and white matter reduction. However, it remains unknown how mutant LGI1 causes white matter abnormalities at molecular and cellular levels. Here, we generated a knock‐in mouse bearing this Lgi1 mutation. We found that Lgi1D51G/D51G mice exhibited impaired defective white matter and motor coordination. We observed that Lgi1D51G/D51G mice displayed a reduced number of mature oligodendrocytes (OLs) and deficient OL differentiation in the white matter. However, the population of oligodendrocyte precursor cells was not affected in Lgi1D51G/D51G mice. Mechanistically, we showed that the Lgi1D51G mutation resulted in altered mTOR signaling and led to decreased levels of Sox10. Given that Sox10 is a key transcriptional factor to control OL differentiation, our results strongly suggest that the Lgi1D51G mutation may cause white matter abnormalities via inhibiting Sox10‐dependent OL differentiation and myelination in the central nervous system. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
21. Clemastine Rescues Chemotherapy-Induced Cognitive Impairment by Improving White Matter Integrity.
- Author
-
Chen, Yingxi, Sheng, Jie, Tang, Xiuying, Zhao, Yuhong, Zhu, Shujuan, and Liu, Qian
- Subjects
- *
WHITE matter (Nerve tissue) , *CHEMOTHERAPY complications , *COGNITION disorders , *CORPUS callosum - Abstract
• Chemotherapy damages cognitive function by injuring white matter integrity. • Clemastine can rescue chemotherapy-induced cognitive impairment of mice. • Clemastine has ability of rescuing white matter injury induced by chemotherapy. With the improvement of cancer treatment techniques, increasing attention has been given to chemotherapy-induced cognitive impairment through white matter injury. Clemastine fumarate has been shown to enhance white matter integrity in cuprizone- or hypoxia-induced demyelination mouse models. However, whether clemastine can be beneficial for reversing chemotherapy-induced cognitive impairment remains unexplored. In this study, the mice received oral administration of clemastine after chemotherapy. The open-field test and Morris water maze test were used to evaluate their anxiety, locomotor activity and cognitive function. Luxol Fast Blue staining and transmission electron microscopy were used to detect the morphological damage to the myelin. Demyelination and damage to the mature oligodendrocytes and axons were observed by immunofluorescence and western blotting. Clemastine significantly improved their cognitive function and ameliorated white matter injury in the chemotherapy-treated mice. Clemastine enhanced myelination, promoted oligodendrocyte precursor cell differentiation and increased the neurofilament 200 protein levels in the corpus callosum and hippocampus. We concluded that clemastine rescues cognitive function damage caused by chemotherapy through improving white matter integrity. Remyelination, oligodendrocyte differentiation and the increase of neurofilament protein promoted by clemastine are potential strategies for reversing the cognitive dysfunction caused by chemotherapy. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
22. Ethanol effects on cerebellar myelination in a postnatal mouse model of fetal alcohol spectrum disorders.
- Author
-
Niedzwiedz-Massey, Victoria M., Douglas, James C., Rafferty, Tonya, Kane, Cynthia J.M., and Drew, Paul D.
- Subjects
- *
FETAL alcohol syndrome , *LABORATORY mice , *MYELINATION , *ANIMAL disease models , *ETHANOL , *ANIMAL experimentation , *CEREBELLUM , *NERVE tissue , *RESEARCH funding , *CENTRAL nervous system , *MICE - Abstract
Fetal alcohol spectrum disorders (FASD) are alarmingly common, result in significant personal and societal loss, and there are no effective treatments for these disorders. Cerebellar neuropathology is common in FASD and can cause impaired cognitive and motor function. The current study evaluates the effects of ethanol on oligodendrocyte-lineage cells, as well as molecules that modulate oligodendrocyte differentiation and function in the cerebellum in a postnatal mouse model of FASD. Neonatal mice were treated with ethanol from P4-P9 (postnatal day), the cerebellum was isolated at P10, and mRNAs encoding oligodendrocyte-associated molecules were quantitated by qRT-PCR. Our studies demonstrated that ethanol significantly reduced the expression of markers for multiple stages of oligodendrocyte maturation, including oligodendrocyte precursor cells, pre-myelinating oligodendrocytes, and mature myelinating oligodendrocytes. Additionally, we determined that ethanol significantly decreased the expression of molecules that play critical roles in oligodendrocyte differentiation. Interestingly, we also observed that ethanol significantly reduced the expression of myelin-associated inhibitors, which may act as a compensatory mechanism to ethanol toxicity. Furthermore, we demonstrate that ethanol alters the expression of a variety of molecules important in oligodendrocyte function and myelination. Collectively, our studies increase our understanding of specific mechanisms by which ethanol modulates myelination in the developing cerebellum, and potentially identify novel targets for FASD therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
23. The Wnt effector transcription factor 7-like 2 positively regulates oligodendrocyte differentiation in a manner independent of Wnt/β-catenin signaling.
- Author
-
Hammond, Elizabeth, Lang, Jordan, Maeda, Yoshiko, Pleasure, David, Angus-Hill, Melinda, Xu, Jie, Horiuchi, Makoto, Deng, Wenbin, and Guo, Fuzheng
- Subjects
Oligodendroglia ,Myelin Sheath ,Animals ,Mice ,Transgenic ,Mice ,Bone Morphogenetic Proteins ,Signal Transduction ,Cell Differentiation ,Gene Expression Regulation ,Developmental ,beta Catenin ,Gene Knockdown Techniques ,Transcription Factor 7-Like 2 Protein ,Wnt Signaling Pathway ,BMP signaling ,TCF7l2(TCF4) ,canonical Wnt/beta-catenin signaling ,myelination ,oligodendrocyte differentiation ,remyelination ,Brain Disorders ,Stem Cell Research ,Pediatric ,Multiple Sclerosis ,Autoimmune Disease ,Neurosciences ,Genetics ,Perinatal Period - Conditions Originating in Perinatal Period ,Neurodegenerative ,Infant Mortality ,Underpinning research ,1.1 Normal biological development and functioning ,Medical and Health Sciences ,Psychology and Cognitive Sciences ,Neurology & Neurosurgery - Abstract
Genetic or pharmacological activation of canonical Wnt/β-catenin signaling inhibits oligodendrocyte differentiation. Transcription factor 7-like 2 (TCF7l2), also known as TCF4, is a Wnt effector induced transiently in the oligodendroglial lineage. A well accepted dogma is that TCF7l2 inhibits oligodendrocyte differentiation through activation of Wnt/β-catenin signaling. We report that TCF7l2 is upregulated transiently in postmitotic, newly differentiated oligodendrocytes. Using in vivo gene conditional ablation, we found surprisingly that TCF7l2 positively regulates neonatal and postnatal mouse oligodendrocyte differentiation during developmental myelination and remyelination in a manner independent of the Wnt/β-catenin signaling pathway. We also reveal a novel role of TCF7l2 in repressing a bone morphogenetic protein signaling pathway that is known to inhibit oligodendrocyte differentiation. Thus, our study provides novel data justifying therapeutic attempts to enhance, rather than inhibit, TCF7l2 signaling to overcome arrested oligodendroglial differentiation in multiple sclerosis and other demyelinating diseases.
- Published
- 2015
24. Akt Regulates Sox10 Expression to Control Oligodendrocyte Differentiation via Phosphorylating FoxO1.
- Author
-
He Wang, Mengjia Liu, Zhuoyang Ye, Cuihua Zhou, Huiru Bi, Long Wang, Chen Zhang, Hui Fu, Ying Shen, Jian-Jun Yang, Yimin Hu, and Guiquan Chen
- Subjects
- *
LEUKOENCEPHALOPATHIES , *PROTEIN kinases , *PHOSPHORYLATION , *CELL death - Abstract
Sox10 is a well known factor to control oligodendrocyte (OL) differentiation, and its expression is regulated by Olig2. As an important protein kinase, Akt has been implicated in diseases with white matter abnormalities. To study whether and how Akt may regulate OL development, we generated OL lineage cell-specific Akt1/Akt2/Akt3 triple conditional knock-out (Akt cTKO) mice. Both male and female mice were used. These mutants exhibit a complete loss of mature OLs and unchanged apoptotic cell death in the CNS. We show that the deletion of Akt three isoforms causes downregulation of Sox10 and decreased levels of phosphorylated FoxO1 in the brain. In vitro analysis reveals that the expression of FoxO1 with mutations on phosphorylation sites for Akt significantly represses the Sox10 promoter activity, suggesting that phosphorylation of FoxO1 by Akt is important for Sox10 expression. We further demonstrate that mutant FoxO1 without Akt phosphorylation epitopes is enriched in the Sox10 promoter. Together, this study identifies a novel FoxO1 phosphorylation-dependent mechanism for Sox10 expression and OL differentiation. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
25. The combined administration of LNC-encapsulated retinoic acid and calcitriol stimulates oligodendrocyte progenitor cell differentiation in vitro and in vivo after intranasal administration.
- Author
-
Labrak, Y., Alhouayek, M., Mwema, A., d'Auria, L., Ucakar, B., van Pesch, V., Muccioli, G.G., and des Rieux, A.
- Subjects
- *
INTRANASAL administration , *TRETINOIN , *CALCITRIOL , *PROGENITOR cells , *CELL differentiation , *POLYMERSOMES , *OLIGODENDROGLIA - Abstract
[Display omitted] • Encapsulation in lipid nanocapsules stabilizes retinoic acid. • Combining calcitriol and retinoic acid is more effective than each molecule alone. • Nose-to-brain delivery of calcitriol and retinoic acid enhances myelin repair. Intranasal administration is an efficient strategy for bypassing the BBB, favoring drug accumulation in the brain, and improving its efficiency. Lipid nanocapsules (LNC) are suitable nanocarriers for the delivery of lipophilic drugs via this route and can be used to encapsulate lipophilic molecules such as retinoic acid (RA) and calcitriol (Cal). As the hallmarks of multiple sclerosis (MS) are neuroinflammation and oligodendrocyte loss, our hypothesis was that by combining two molecules known for their pro-differentiating properties, encapsulated in LNC, and delivered by intranasal administration, we would stimulate oligodendrocyte progenitor cells (OPC) differentiation into oligodendrocytes and provide a new pro-remyelinating therapy. LNC loaded with RA (LNC-RA) and Cal (LNC-Cal) were stable for at least 8 weeks. The combination of RA and Cal was more efficient than the molecules alone, encapsulated or not, on OPC differentiation in vitro and decreased microglia cell activation in a dose-dependent manner. After the combined intranasal administration of LNC-RA and LNC-Cal in a mouse cuprizone model of demyelination, increased MBP staining was observed in the corpus callosum. In conclusion, intranasal delivery of lipophilic drugs encapsulated in LNC is a promising strategy for myelinating therapies. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
26. A2B Adenosine Receptors and Sphingosine 1-Phosphate Signaling Cross-Talk in Oligodendrogliogenesis
- Author
-
Elisabetta Coppi, Francesca Cencetti, Federica Cherchi, Martina Venturini, Chiara Donati, Paola Bruni, Felicita Pedata, and Anna Maria Pugliese
- Subjects
adenosine ,sphingosine kinase (SphK) ,remyelination ,K+ channels ,oligodendrocyte differentiation ,sphingosine-1-phosphate ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Oligodendrocyte-formed myelin sheaths allow fast synaptic transmission in the brain. Impairments in the process of myelination, or demyelinating insults, might cause chronic diseases such as multiple sclerosis (MS). Under physiological conditions, remyelination is an ongoing process throughout adult life consisting in the differentiation of oligodendrocyte progenitor cells (OPCs) into mature oligodendrocytes (OLs). During pathological events, this process fails due to unfavorable environment. Adenosine and sphingosine kinase/sphingosine 1-phosphate signaling axes (SphK/S1P) play important roles in remyelination processes. Remarkably, fingolimod (FTY720), a sphingosine analog recently approved for MS treatment, plays important roles in OPC maturation. We recently demonstrated that the selective stimulation of A2B adenosine receptors (A2BRs) inhibit OPC differentiation in vitro and reduce voltage-dependent outward K+ currents (IK) necessary to OPC maturation, whereas specific SphK1 or SphK2 inhibition exerts the opposite effect. During OPC differentiation A2BR expression increases, this effect being prevented by SphK1/2 blockade. Furthermore, selective silencing of A2BR in OPC cultures prompts maturation and, intriguingly, enhances the expression of S1P lyase, the enzyme responsible for irreversible S1P catabolism. Finally, the existence of an interplay between SphK1/S1P pathway and A2BRs in OPCs was confirmed since acute stimulation of A2BRs activates SphK1 by increasing its phosphorylation. Here the role of A2BR and SphK/S1P signaling during oligodendrogenesis is reviewed in detail, with the purpose to shed new light on the interaction between A2BRs and S1P signaling, as eventual innovative targets for the treatment of demyelinating disorders.
- Published
- 2021
- Full Text
- View/download PDF
27. Pen-2 Negatively Regulates the Differentiation of Oligodendrocyte Precursor Cells into Astrocytes in the Central Nervous System.
- Author
-
Jinxing Hou, Huiru Bi, Zhuoyang Ye, Wenhui Huang, Gang Zou, Xiaochuan Zou, Yun Stone Shi, Ying Shen, Quanhong Ma, Frank Kirchhoff, Yimin Hu, and Guiquan Chen
- Subjects
- *
CENTRAL nervous system , *ASTROCYTES , *NEUROLOGICAL disorders - Abstract
Mutations on c-secretase subunits are associated with neurologic diseases. Whereas the role of c-secretase in neurogenesis has been intensively studied, little is known about its role in astrogliogenesis. Recent evidence has demonstrated that astrocytes can be generated from oligodendrocyte precursor cells (OPCs). However, it is not well understood what mechanism may control OPCs to differentiate into astrocytes. To address the above questions, we generated two independent lines of oligodendrocyte lineage-specific presenilin enhancer 2 (Pen-2) conditional KO mice. Both male and female mice were used. Here we demonstrate that conditional inactivation of Pen-2 mediated by Olig1-Cre or NG2-CreERT2 causes enhanced generation of astrocytes. Lineage-tracing experiments indicate that abnormally generated astrocytes are derived from Cre-expressing OPCs in the CNS in Pen-2 conditional KO mice. Mechanistic analysis reveals that deletion of Pen-2 inhibits the Notch signaling to upregulate signal transducer and activator of transcription 3, which triggers activation of GFAP to promote astrocyte differentiation. Together, these novel findings indicate that Pen-2 regulates the specification of astrocytes from OPCs through the signal transducer and activator of transcription 3 signaling. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
28. A2 B Adenosine Receptors and Sphingosine 1-Phosphate Signaling Cross-Talk in Oligodendrogliogenesis.
- Author
-
Coppi, Elisabetta, Cencetti, Francesca, Cherchi, Federica, Venturini, Martina, Donati, Chiara, Bruni, Paola, Pedata, Felicita, and Pugliese, Anna Maria
- Subjects
FINGOLIMOD ,ADENOSINES ,SPHINGOSINE kinase ,SPHINGOSINE ,MYELIN sheath ,DEMYELINATION - Abstract
Oligodendrocyte-formed myelin sheaths allow fast synaptic transmission in the brain. Impairments in the process of myelination, or demyelinating insults, might cause chronic diseases such as multiple sclerosis (MS). Under physiological conditions, remyelination is an ongoing process throughout adult life consisting in the differentiation of oligodendrocyte progenitor cells (OPCs) into mature oligodendrocytes (OLs). During pathological events, this process fails due to unfavorable environment. Adenosine and sphingosine kinase/sphingosine 1-phosphate signaling axes (SphK/S1P) play important roles in remyelination processes. Remarkably, fingolimod (FTY720), a sphingosine analog recently approved for MS treatment, plays important roles in OPC maturation. We recently demonstrated that the selective stimulation of A
2 B adenosine receptors (A2 B Rs) inhibit OPC differentiation in vitro and reduce voltage-dependent outward K+ currents (IK ) necessary to OPC maturation, whereas specific SphK1 or SphK2 inhibition exerts the opposite effect. During OPC differentiation A2 B R expression increases, this effect being prevented by SphK1/2 blockade. Furthermore, selective silencing of A2 B R in OPC cultures prompts maturation and, intriguingly, enhances the expression of S1P lyase, the enzyme responsible for irreversible S1P catabolism. Finally, the existence of an interplay between SphK1/S1P pathway and A2 B Rs in OPCs was confirmed since acute stimulation of A2 B Rs activates SphK1 by increasing its phosphorylation. Here the role of A2 B R and SphK/S1P signaling during oligodendrogenesis is reviewed in detail, with the purpose to shed new light on the interaction between A2 B Rs and S1P signaling, as eventual innovative targets for the treatment of demyelinating disorders. [ABSTRACT FROM AUTHOR]- Published
- 2021
- Full Text
- View/download PDF
29. The Wnt Effector TCF712 Promotes Oligodendroglial Differentiation by Repressing Autocrine BMP4-Mediated Signaling.
- Author
-
Sheng Zhang, Yan Wang, Xiaoqing Zhu, Lanying Song, Xinhua Zhan, Ma, Edric, McDonough, Jennifer, Hui Fu, Cambi, Franca, Grinspan, Judith, and Fuzheng Guo
- Subjects
- *
BONE morphogenetic proteins , *MYELIN oligodendrocyte glycoprotein , *DEMYELINATION , *MYELIN sheath diseases , *TRANSCRIPTION factors , *MULTIPLE sclerosis , *PROTHROMBIN - 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. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
30. MiRNA‐145‐5p prevents differentiation of oligodendrocyte progenitor cells by regulating expression of myelin gene regulatory factor.
- Author
-
Kornfeld, Samantha F., Cummings, Sarah E., Fathi, Samaneh, Bonin, Sawyer R., and Kothary, Rashmi
- Subjects
- *
REGULATOR genes , *PROGENITOR cells , *GENE expression , *DEMYELINATION , *MICRORNA , *MYELIN proteins - Abstract
The roles of specific microRNAs (miRNA) in oligodendrocyte (OL) differentiation have been studied in depth. However, miRNAs in OL precursors and oligodendrocyte progenitor cells (OPCs) have been less extensively investigated. MiR‐145‐5p is highly expressed in OPCs relative to differentiating OLs, suggesting this miRNA may serve a function specifically in OPCs. Knockdown of miR‐145‐5p in primary OPCs led to spontaneous differentiation, as evidenced by an increased proportion of MAG+ cells, increased cell ramification, and upregulation of multiple myelin genes including MYRF, TPPP, and MAG, and OL cell cycle exit marker Cdkn1c. Supporting this transition to a differentiating state, proliferation was reduced in miR‐145‐5p knockdown OPCs. Further, knockdown of miR‐145‐5p in differentiating OLs showed enhanced differentiation, with increased branching, myelin membrane production, and myelin gene expression. We identified several OL‐specific genes targeted by miR‐145‐5p that exhibited upregulation with miR‐145‐5p knockdown, including myelin gene regulatory factor (MYRF), that could be regulating the prodifferentiation phenotype in both miR‐145 knockdown OPCs and OLs. Indeed, spontaneous differentiation with knockdown of miR‐145‐5p was fully rescued by concurrent knockdown of MYRF. However, proliferation rate was only partially rescued with MYRF knockdown, and overexpression of miR‐145‐5p in OPCs increased proliferation rate without affecting expression of already lowly expressed differentiation genes. Taken together, these data suggest that in OPCs miR‐145‐5p both prevents differentiation at least in part by preventing expression of MYRF and promotes proliferation via as‐yet‐unidentified mechanisms. These findings clarify the need for differential regulation of miR‐145‐5p between OPCs and OLs and may have further implications in demyelinating diseases such as multiple sclerosis where miR‐145‐5p is dysregulated. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
31. New Insight into the Role of Adenosine in Demyelination, Stroke and Neuropathic Pain
- Author
-
Elisabetta Coppi, Ilaria Dettori, Federica Cherchi, Irene Bulli, Martina Venturini, Felicita Pedata, and Anna Maria Pugliese
- Subjects
cerebral ischemia ,oxygen-glucose deprivation ,adenosine receptors ,oligodendrocyte differentiation ,neuropathic pain ,dorsal root ganglion neurons ,Therapeutics. Pharmacology ,RM1-950 - Published
- 2021
- Full Text
- View/download PDF
32. Progesterone effects on the oligodendrocyte linage: all roads lead to the progesterone receptor
- Author
-
Ignacio Jure, Alejandro F De Nicola, and Florencia Labombarda
- Subjects
progesterone ,progesterone receptor ,oligodendrocyte differentiation ,myelination ,remyelination ,neuroinflammation ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
A new role has emerged for progesterone after discovering its potent actions away from reproduction in both the central and the peripheral nervous system. The aim of the present report is to discuss progesterone’s mechanisms of action involved in myelination, remyelination and neuroinflammation. The pivotal role of the classic progesterone receptor is described and evidence is compiled about progesterone’s direct effects on oligodendrocyte linage and its indirect effects on oligodendrocyte precursor cell differentiation by decreasing the neuroinflammatory environment.
- Published
- 2019
- Full Text
- View/download PDF
33. Genetic Evidence that Dorsal Spinal Oligodendrocyte Progenitor Cells are Capable of Myelinating Ventral Axons Effectively in Mice.
- Author
-
Fang, Minxi, Yu, Qian, Ou, Baiyan, Huang, Hao, Yi, Min, Xie, Binghua, Yang, Aifen, Qiu, Mengsheng, and Xu, Xiaofeng
- Abstract
In the developing spinal cord, the majority of oligodendrocyte progenitor cells (OPCs) are induced in the ventral neuroepithelium under the control of the Sonic Hedgehog (Shh) signaling pathway, whereas a small subset of OPCs are generated from the dorsal neuroepithelial cells independent of the Shh pathway. Although dorsally-derived OPCs (dOPCs) have been shown to participate in local axonal myelination in the dorsolateral regions during development, it is not known whether they are capable of migrating into the ventral region and myelinating ventral axons. In this study, we confirmed and extended the previous study on the developmental potential of dOPCs in the absence of ventrally-derived OPCs (vOPCs). In Nestin-Smo conditional knockout (cKO) mice, when ventral oligodendrogenesis was blocked, dOPCs were found to undergo rapid amplification, spread to ventral spinal tissue, and eventually differentiated into myelinating OLs in the ventral white matter with a temporal delay, providing genetic evidence that dOPCs are capable of myelinating ventral axons in the mouse spinal cord. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
34. Pushing myelination – developmental regulation of myosin expression drives oligodendrocyte morphological differentiation.
- Author
-
Domingues, Helena Sofia, Urbanski, Mateusz M., Macedo-Ribeiro, Sandra, Almaktari, Amr, Irfan, Azka, Hernandez, Yamely, Wang, Haibo, Relvas, João Bettencourt, Rubinstein, Boris, Melendez-Vasquez, Carmen V., and Pinto, Inês Mendes
- Subjects
- *
OLIGODENDROGLIA , *MYOSIN , *NEURAL circuitry , *CENTRAL nervous system , *MYELINATION , *ELECTRIC insulators & insulation - Abstract
Oligodendrocytes are the central nervous system myelin-forming cells providing axonal electrical insulation and higher-order neuronal circuitry. The mechanical forces driving the differentiation of oligodendrocyte precursor cells into myelinating oligodendrocytes are largely unknown, but likely require the spatiotemporal regulation of the architecture and dynamics of the actin and actomyosin cytoskeletons. In this study, we analyzed the expression pattern of myosin motors during oligodendrocyte development. We report that oligodendrocyte differentiation is regulated by the synchronized expression and nonuniform distribution of several members of the myosin network, particularly non-muscle myosins 2B and 2C, which potentially operate as nanomechanical modulators of cell tension and myelin membrane expansion at different cell stages. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
35. eIF2B Mutations Cause Mitochondrial Malfunction in Oligodendrocytes.
- Author
-
Herrero, Melisa, Mandelboum, Shir, and Elroy-Stein, Orna
- Abstract
Vanishing white matter (VWM) disease (OMIM#306896) is an autosomal recessive neurodegenerative leukodystrophy caused by hypomorphic mutations in any of the five genes encoding the subunits of eukaryotic translation initiation factor 2B (eIF2B). The disease is manifested by loss of cerebral white matter and progressive deterioration upon exposure to environmental and physiological stressors. "Foamy" oligodendrocytes (OLG), increased numbers of oligodendrocytes precursor cells (OPC), and immature defective astrocytes are major neuropathological denominators. Our recent work using Eif2b5
R132H/R132H mice uncovered a fundamental link between eIF2B and mitochondrial function. A decrease in oxidative phosphorylation capacity was observed in mutant astrocytes and fibroblasts. While an adaptive increase in mitochondria abundance corrects the phenotype of mutant fibroblasts, it is not sufficient to compensate for the high-energy demand of astrocytes, explaining their involvement in the disease. To date, astrocytes are marked as central for the disease while eIF2B-mutant OLG are currently assumed to lack a cellular phenotype on their own. Here we show a reduced capacity of eIF2B-mutant OPC isolated from Eif2b5R132H/R132H mice to conduct oxidative respiration despite the adaptive increase in their mitochondrial abundance. We also show their impaired ability to efficiently complete critical differentiation steps towards mature OLG. The concept that defective differentiation of eIF2B-mutant OPC could be a consequence of mitochondrial malfunction is in agreement with numerous studies indicating high dependency of differentiating OLG on accurate mitochondrial performance and ATP availability. [ABSTRACT FROM AUTHOR]- Published
- 2019
- Full Text
- View/download PDF
36. Leonurine suppresses neuroinflammation through promoting oligodendrocyte maturation.
- Author
-
Jin, Min, Li, Qian, Gu, Yuting, Wan, Bing, Huang, Jiefang, Xu, Xuanbai, Huang, Rui, and Zhang, Yanyun
- Subjects
INFLAMMATION ,OLIGODENDROGLIA ,APOPTOSIS ,ENCEPHALOMYELITIS ,AUTOIMMUNE diseases - Abstract
Focal inflammation and remyelination failure are major hallmarks of multiple sclerosis and its animal model, experimental autoimmune encephalomyelitis (EAE). In this study, we found that leonurine, a bioactive alkaloid, alleviated EAE disease severity along with reduced central nervous system inflammation and myelin damage. During the pathogenesis of EAE, leonurine dramatically suppressed the recruitment of encephalitogenic T cells into the central nervous system, whereas did not impair periphery immune responses and microglia activation. Mechanistically, leonurine protected mice against demyelination along with enhanced remyelination through promoting the maturation of oligodendrocytes in both EAE and cuprizone‐induced demyelination mouse models. Moreover, we identified that the expression of demethylase jumonji domain‐containing protein D3 was significantly enhanced upon treatment of leonurine, which suppressed the trimethylation of histone H3 lysine‐27 and enhanced oligodendrocyte maturation accordingly. Collectively, our study identified the therapeutic effect of leonurine on EAE model, which potentially represents a promising therapeutic strategy for multiple sclerosis, even other demyelination disorders. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
37. The Infantile Leukoencephalopathy-Associated Mutation of C11ORF73/HIKESHI Proteins Generates De Novo Interactive Activity with Filamin A, Inhibiting Oligodendroglial Cell Morphological Differentiation
- Author
-
Kohei Hattori, Kenji Tago, Shiori Memezawa, Arisa Ochiai, Sui Sawaguchi, Yukino Kato, Takanari Sato, Kazuma Tomizuka, Hiroaki Ooizumi, Katsuya Ohbuchi, Kazushige Mizoguchi, Yuki Miyamoto, and Junji Yamauchi
- Subjects
leukoencephalopathy ,HLD13 ,C11orf73 ,Filamin A ,oligodendrocyte differentiation ,Medicine - Abstract
Background: Genetic hypomyelinating diseases are a heterogeneous group of disorders involving the white matter. One infantile hypomyelinating leukoencephalopathy is associated with the homozygous variant (Cys4-to-Ser (C4S)) of the c11orf73 gene. Methods: We observed that in mouse oligodendroglial FBD-102b cells, the C4S mutant proteins but not the wild type ones of C11orf73 are microscopically localized in the lysosome. And, they downregulate lysosome-related signaling in an immunoblotting technique. Results: The C4S mutant proteins specifically interact with Filamin A, which is known to anchor transmembrane proteins to the actin cytoskeleton; the C4S mutant proteins and Filamin A are also observed in the lysosome fraction. While parental FBD-102b cells and cells harboring the wild type constructs exhibit morphological differentiation, cells harboring C4S mutant constructs do not. It may be that morphological differentiation is inhibited because expression of these C4S mutant proteins leads to defects in the actin cytoskeletal network involving Filamin A. Conclusions: The findings that leukoencephalopathy-associated C11ORF73 mutant proteins specifically interact with Filamin A, are localized in the lysosome, and inhibit morphological differentiation shed light on the molecular and cellular pathological mechanisms that underlie infantile hypomyelinating leukoencephalopathy.
- Published
- 2021
- Full Text
- View/download PDF
38. UDP-Gal: Ceramide Galactosyltransferase (UGT8)
- Author
-
Honke, Koichi, Taniguchi, Naoyuki, editor, Honke, Koichi, editor, Fukuda, Minoru, editor, Narimatsu, Hisashi, editor, Yamaguchi, Yoshiki, editor, and Angata, Takashi, editor
- Published
- 2014
- Full Text
- View/download PDF
39. Mechanical Strain Alters Cellular and Nuclear Dynamics at Early Stages of Oligodendrocyte Differentiation
- Author
-
Ekta Makhija, Anna Jagielska, Lena Zhu, Alexander C. Bost, William Ong, Sing Y. Chew, G. V. Shivashankar, and Krystyn J. Van Vliet
- Subjects
oligodendrocyte differentiation ,strain ,nuclear dynamics ,cell migration ,microtubules ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Mechanical and physical stimuli including material stiffness and topography or applied mechanical strain have been demonstrated to modulate differentiation of glial progenitor and neural stem cells. Recent studies probing such mechanotransduction in oligodendrocytes have focused chiefly on the biomolecular components. However, the cell-level biophysical changes associated with such responses remain largely unknown. Here, we explored mechanotransduction in oligodendrocyte progenitor cells (OPCs) during the first 48 h of differentiation induction by quantifying the biophysical state in terms of nuclear dynamics, cytoskeleton organization, and cell migration. We compared these mechanophenotypic changes in OPCs exposed to both chemical cues (differentiation factors) and mechanical cues (static tensile strain of 10%) with those exposed to only those chemical cues. We observed that mechanical strain significantly hastened the dampening of nuclear fluctuations and decreased OPC migration, consistent with the progression of differentiation. Those biophysical changes were accompanied by increased production of the intracellular microtubule network. These observations provide insights into mechanisms by which mechanical strain of physiological magnitude could promote differentiation of progenitor cells to oligodendrocytes via inducing intracellular biophysical responses over hours to days post induction.
- Published
- 2018
- Full Text
- View/download PDF
40. Differentiation of Induced Pluripotent Stem Cells into Oligodendrocytes: Increased Efficiency of Selecting Oligodendrocyte Precursor Cells Using A2B5 Monoclonal Antibody
- Author
-
Tokumoto, Yasuhito M. and Hayat, M.A., editor
- Published
- 2013
- Full Text
- View/download PDF
41. Development of Oligodendrocytes in the Vertebrate CNS
- Author
-
Miller, Robert H., Duncan, Ian D., editor, and Franklin, Robin J M, editor
- Published
- 2013
- Full Text
- View/download PDF
42. Oligodendrocyte pathology in fetal alcohol spectrum disorders
- Author
-
Nune Darbinian and Michael E. Selzer
- Subjects
Pathology ,medicine.medical_specialty ,Fetal alcohol syndrome ,oligodendrocyte precursor cells ,Review ,Neuroprotection ,Developmental Neuroscience ,fetal brain ,medicine ,RC346-429 ,development ,alcohol ,dysmyelination ,ethanol ,fetal alcohol syndrome ,myelin basic protein ,neurodegeneration ,oligodendrocyte injury ,Fetus ,biology ,business.industry ,Multiple sclerosis ,Neurodegeneration ,Oligodendrocyte differentiation ,medicine.disease ,Oligodendrocyte ,Myelin basic protein ,medicine.anatomical_structure ,biology.protein ,Neurology. Diseases of the nervous system ,business - Abstract
The pathology of fetal alcohol syndrome and the less severe fetal alcohol spectrum disorders includes brain dysmyelination. Recent studies have shed light on the molecular mechanisms underlying these white matter abnormalities. Rodent models of fetal alcohol syndrome and human studies have shown suppressed oligodendrocyte differentiation and apoptosis of oligodendrocyte precursor cells. Ethanol exposure led to reduced expression of myelin basic protein and delayed myelin basic protein expression in rat and mouse models of fetal alcohol syndrome and in human histopathological specimens. Several studies have reported increased expression of many chemokines in dysmyelinating disorders in central nervous system, including multiple sclerosis and fetal alcohol syndrome. Acute ethanol exposure reduced levels of the neuroprotective insulin-like growth factor-1 in fetal and maternal sheep and in human fetal brain tissues, while ethanol increased the expression of tumor necrosis factor α in mouse and human neurons. White matter lesions have been induced in the developing sheep brain by alcohol exposure in early gestation. Rat fetal alcohol syndrome models have shown reduced axon diameters, with thinner myelin sheaths, as well as reduced numbers of oligodendrocytes, which were also morphologically aberrant oligodendrocytes. Expressions of markers for mature myelination, including myelin basic protein, also were reduced. The accumulating knowledge concerning the mechanisms of ethanol-induced dysmyelination could lead to the development of strategies to prevent dysmyelination in children exposed to ethanol during fetal development. Future studies using fetal oligodendrocyte- and oligodendrocyte precursor cell-derived exosomes isolated from the mother’s blood may identify biomarkers for fetal alcohol syndrome and even implicate epigenetic changes in early development that affect oligodendrocyte precursor cell and oligodendrocyte function in adulthood. By combining various imaging modalities with molecular studies, it may be possible to determine which fetuses are at risk and to intervene therapeutically early in the pregnancy.
- Published
- 2022
43. Endothelial Nitric Oxide Synthase–Deficient Mice
- Author
-
Ling Chen, Rajendra Raghow, Xing-Lin Tan, Geng Lin, Fu-Ming Zhou, Xingyong Chen, Andy Y. Shih, Francesca-Fang Liao, and Wei Zheng
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,biology ,business.industry ,Neurodegeneration ,Oligodendrocyte differentiation ,medicine.disease ,biology.organism_classification ,Pathology and Forensic Medicine ,Astrogliosis ,Nitric oxide ,White matter ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,chemistry ,Enos ,Aging brain ,Medicine ,Cerebral amyloid angiopathy ,business ,030217 neurology & neurosurgery - Abstract
Age-related cerebral small-vessel disease (CSVD) is a major cause of stroke and dementia. Despite a widespread acceptance of small-vessel arteriopathy, lacunar infarction, diffuse white matter injury, and cognitive impairment as four cardinal features of CSVD, a unifying pathologic mechanism of CSVD remains elusive. Herein, we introduce partial endothelial nitric oxide synthase (eNOS)-deficient mice as a model of age-dependent, spontaneous CSVD. These mice developed cerebral hypoperfusion and blood-brain barrier leakage at a young age, which progressively worsened with advanced age. Their brains exhibited elevated oxidative stress, astrogliosis, cerebral amyloid angiopathy, microbleeds, microinfarction, and white matter pathology. Partial eNOS-deficient mice developed gait disturbances at middle age, and hippocampus-dependent memory deficits at older ages. These mice also showed enhanced expression of bone morphogenetic protein 4 (BMP4) in brain pericytes before myelin loss and white matter pathology. Because BMP4 signaling not only promotes astrogliogenesis but also blocks oligodendrocyte differentiation, we posit that paracrine actions of BMP4, localized within the neurovascular unit, promote white matter disorganization and neurodegeneration. These observations point to BMP4 signaling pathway in the aging brain vasculature as a potential therapeutic target. Finally, because studies in partial eNOS-deficient mice corroborated recent clinical evidence that blood-brain barrier disruption is a primary cause of white matter pathology, the mechanism of impaired nitric oxide signaling-mediated CSVD warrants further investigation.
- Published
- 2021
- Full Text
- View/download PDF
44. Fate Specification of Neural Stem Cells
- Author
-
Namihira, Masakazu, Nakashima, Kinichi, Seki, Tatsunori, editor, Sawamoto, Kazunobu, editor, Parent, Jack M., editor, and Alvarez-Buylla, Arturo, editor
- Published
- 2011
- Full Text
- View/download PDF
45. Physiological Functions of TR2 and TR4 Orphan Nuclear Receptor
- Author
-
Liu, Su, Xie, Shaozhen, Lee, Yi-fen, Chang, Chawnshang, Bunce, Chris M., editor, and Campbell, Moray J., editor
- Published
- 2010
- Full Text
- View/download PDF
46. Interactive Repression of MYRF Self-Cleavage and Activity in Oligodendrocyte Differentiation by TMEM98 Protein.
- Author
-
Hao Huang, Peng Teng, Junqing Du, Jun Meng, Xuemei Hu, Tao Tang, Zunyi Zhang, Qi, Yingchuan B., and Mengsheng Qiu
- Subjects
- *
MEMBRANE proteins , *MYELIN sheath , *SPINAL cord , *TRANSCRIPTION factors , *ENDOPLASMIC reticulum - Abstract
Myelin sheath formed by oligodendrocytes (OLs) is essential for the rapid propagation of action potentials in the vertebrate CNS. Myelin regulatory factor (MYRF) is one of the critical factors that control OL differentiation and myelin maintenance. Previous studies showed that MYRF is a membrane-bound transcription factor associated with the endoplasmic reticulum (HR). After self-cleavage, the N-fragment of MYRF is released from the ER and translocated into the nucleus where it functions as a transcription factor to activate myelin gene expression. At present, it remains unknown whether MYRF self-cleavage and functional activation can be regulated during OL differentiation. Here, we report that TMEM98, an ER-associated transmembrane protein, is capable of binding to the C-terminal of MYRF and inhibiting its self-cleavage and N-fragment nuclear translocation. In the developing CNS, TMEM98 is selectively expressed in early maturing OLs in mouse pups of either sex. Forced expression of TMEM98 in embryonic chicken spinal cord of either sex suppresses endogenous OL differentiation and MYRF-induced ectopic expression of myelin genes. These results suggest that TMEM98, through inhibiting the self-cleavage of MYRF, functions as a negative feedback regulator of MYRF in oligodendrocyte differentiation and myelination. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
47. Redox Signaling Mechanisms in Nervous System Development.
- Author
-
Olguín-Albuerne, Mauricio and Morán, Julio
- Subjects
- *
NERVOUS system development , *NEURAL stem cells , *NADPH oxidase , *CELL metabolism , *AXONS - Abstract
Numerous studies have demonstrated the actions of reactive oxygen species (ROS) as regulators of several physiological processes. In this study, we discuss how redox signaling mechanisms operate to control different processes such as neuronal differentiation, oligodendrocyte differentiation, dendritic growth, and axonal growth.Significance: Redox homeostasis regulates the physiology of neural stem cells (NSCs). Notably, the neuronal differentiation process of NSCs is determined by a change toward oxidative metabolism, increased levels of mitochondrial ROS, increased activity of NADPH oxidase (NOX) enzymes, decreased levels of Nrf2, and differential regulation of different redoxins. Furthermore, during the neuronal maturation processes, NOX and MICAL produce ROS to regulate cytoskeletal dynamics, which control the dendritic and axonal growth, as well as the axonal guidance.Recent Advances: The redox homeostasis changes are, in part, attributed to cell metabolism and compartmentalized production of ROS, which is regulated, sensed, and transduced by different molecules such as thioredoxins, glutaredoxins, peroxiredoxins, and nucleoredoxin to control different signaling pathways in different subcellular regions. The study of how these elements cooperatively act is essential for the understanding of nervous system development, as well as the application of regenerative therapies that recapitulate these processes.Critical Issues: The information about these topics in the last two decades leads us to the conclusion that the role of ROS signaling in development of the nervous system is more important than it was previously believed and makes clear the importance of exploring in more detail the mechanisms of redox signaling.Future Directions: Antioxid. Redox Signal . 28, 1603–1625. [ABSTRACT FROM AUTHOR]- Published
- 2018
- Full Text
- View/download PDF
48. Mechanical Strain Alters Cellular and Nuclear Dynamics at Early Stages of Oligodendrocyte Differentiation.
- Author
-
Makhija, Ekta, Jagielska, Anna, Zhu, Lena, Bost, Alexander C., Ong, William, Chew, Sing Y., Shivashankar, G. V., and Van Vliet, Krystyn J.
- Subjects
STIMULUS & response (Psychology) ,OLIGODENDROGLIA ,NEURAL stem cells ,PROGENITOR cells ,CELL differentiation - Abstract
Mechanical and physical stimuli including material stiffness and topography or applied mechanical strain have been demonstrated to modulate differentiation of glial progenitor and neural stem cells. Recent studies probing such mechanotransduction in oligodendrocytes have focused chiefly on the biomolecular components. However, the cell-level biophysical changes associated with such responses remain largely unknown. Here, we explored mechanotransduction in oligodendrocyte progenitor cells (OPCs) during the first 48 h of differentiation induction by quantifying the biophysical state in terms of nuclear dynamics, cytoskeleton organization, and cell migration. We compared these mechanophenotypic changes in OPCs exposed to both chemical cues (differentiation factors) and mechanical cues (static tensile strain of 10%) with those exposed to only those chemical cues. We observed that mechanical strain significantly hastened the dampening of nuclear fluctuations and decreased OPC migration, consistent with the progression of differentiation. Those biophysical changes were accompanied by increased production of the intracellular microtubule network. These observations provide insights into mechanisms by which mechanical strain of physiological magnitude could promote differentiation of progenitor cells to oligodendrocytes via inducing intracellular biophysical responses over hours to days post induction. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
49. Sox2 Is Essential for Oligodendroglial Proliferation and Differentiation during Postnatal Brain Myelination and CNS Remyelination.
- Author
-
Sheng Zhang, Xiaoqing Zhu, Xuehong Gui, Croteau, Christopher, Lanying Song, Jie Xu, Aijun Wang, Bannerman, Peter, and Fuzheng Guo
- Subjects
- *
MYELINATION , *OLIGODENDROGLIA , *PROGENITOR cells , *TRANSCRIPTION factors , *MYELIN , *CELL proliferation - Abstract
In the CNS, myelination and remyelination depend on the successful progression and maturation of oligodendroglial lineage cells, including proliferation and differentiation of oligodendroglial progenitor cells (OPCs). Previous studies have reported that Sox2 transiently regulates oligodendrocyte (OL) differentiation in the embryonicandperinatal spinal cordandappears dispensable for myelination in the postnatal spinal cord. However, the role of Sox2 in OL development in the brain has yet to be defined. We now report that Sox2 is an essential positive regulator of developmental myelination in the postnatal murine brain of both sexes. Stage-specific paradigms of genetic disruption demonstrated that Sox2 regulated brain myelination by coordinating upstream OPC population supply and downstream OL differentiation. Transcriptomic analyses further supported a crucial role of Sox2 in brain developmental myelination. Consistently, oligodendroglial Sox2-deficient mice developed severe tremors and ataxia, typical phenotypes indicative of hypomyelination, and displayed severe impairment of motor function and prominent deficits of brain OL differentiation and myelination persisting into the later CNS developmental stages. We also found that Sox2 was required for efficient OPC proliferation and expansion and OL regeneration during remyelination in the adult brain and spinal cord. Together, ourgeneticevidencerevealsanessential role ofSox2inbrainmyelinationandCNSremyelination, andsuggeststhatmanipulationofSox2and/or Sox2-mediated downstream pathways may be therapeutic in promoting CNS myelin repair. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
50. Akt Regulates Sox10 Expression to Control Oligodendrocyte Differentiation via Phosphorylating FoxO1
- Author
-
Yimin Hu, Ying Shen, Jian-Jun Yang, Hui Fu, Zhuoyang Ye, Chen Zhang, Guiquan Chen, He Wang, Mengjia Liu, Cuihua Zhou, Long Wang, and Huiru Bi
- Subjects
Male ,AKT1 ,Apoptosis ,AKT2 ,FOXO1 ,Biology ,AKT3 ,OLIG2 ,Mice ,Animals ,Phosphorylation ,Protein kinase B ,Research Articles ,Mice, Knockout ,Forkhead Box Protein O1 ,SOXE Transcription Factors ,General Neuroscience ,Oligodendrocyte differentiation ,Brain ,Cell Differentiation ,White Matter ,Cell biology ,Oligodendroglia ,Spinal Cord ,embryonic structures ,Female ,Proto-Oncogene Proteins c-akt - Abstract
Sox10 is a well-known factor to control oligodendrocyte (OL) differentiation and its expression is regulated by Olig2. As an important protein kinase, Akt has been implicated in diseases with white matter (WM) abnormalities. To study whether and how Akt may regulate OL development, we generated OL lineage cells-specific Akt1/Akt2/Akt3 triple conditional knockout (Akt cTKO) mice. Both male and female mice were used. These mutants exhibit complete loss of mature OLs and unchanged apoptotic cell death in the central nervous system. We show that deletion of Akt three isoforms causes down-regulation of Sox10 and decreased levels of phosphorylated FoxO1 (pFoxO1) in the brain. In vitro analysis reveals that expression of FoxO1 with mutations on phosphorylation sites for Akt significantly represses the Sox10 promoter activity, suggesting that phosphorylation of FoxO1 by Akt is important for Sox10 expression. We further demonstrate that mutant FoxO1 without Akt phosphorylation epitopes is enriched in the Sox10 promoter. Together, this study identifies a novel FoxO1 phosphorylation-dependent mechanism for Sox10 expression and OL differentiation. SIGNIFICANCE STATEMENT Dysfunction of Akt is associated with white matter diseases including the agenesis of the corpus callosum. However, it remains unknown whether Akt plays an important role in oligodendrocyte differentiation. To address this question, we generated oligodendrocyte lineage cells-specific Akt1/Akt2/Akt3 triple conditional knockout mice. Akt mutants exhibit deficient white matter development, loss of mature oligodendrocytes, absence of myelination and unchanged apoptotic cell death in the central nervous system. We demonstrate that deletion of Akt three isoforms leads to down-regulation of Sox10, and that phosphorylation of FoxO1 by Akt is critical for Sox10 expression. Together, these findings reveal a novel mechanism to regulate Sox10 expression. This study may provide insights on molecular mechanisms for neurodevelopmental diseases caused by dysfunctions of protein kinases.
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.