1
|
Peters C, Aberle T, Sock E, Brunner J, Küspert M, Hillgärtner S, Wüst HM, Wegner M. Voltage-Gated Ion Channels Are Transcriptional Targets of Sox10 during Oligodendrocyte Development. Cells 2024; 13:1159. [PMID: 38995010 PMCID: PMC11240802 DOI: 10.3390/cells13131159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 07/02/2024] [Accepted: 07/04/2024] [Indexed: 07/13/2024] Open
Abstract
The transcription factor Sox10 is an important determinant of oligodendroglial identity and influences oligodendroglial development and characteristics at various stages. Starting from RNA-seq data, we here show that the expression of several voltage-gated ion channels with known expression and important function in oligodendroglial cells depends upon Sox10. These include the Nav1.1, Cav2.2, Kv1.1, and Kir4.1 channels. For each of the four encoding genes, we found at least one regulatory region that is activated by Sox10 in vitro and at the same time bound by Sox10 in vivo. Cell-specific deletion of Sox10 in oligodendroglial cells furthermore led to a strong downregulation of all four ion channels in a mouse model and thus in vivo. Our study provides a clear functional link between voltage-gated ion channels and the transcriptional regulatory network in oligodendroglial cells. Furthermore, our study argues that Sox10 exerts at least some of its functions in oligodendrocyte progenitor cells, in myelinating oligodendrocytes, or throughout lineage development via these ion channels. By doing so, we present one way in which oligodendroglial development and properties can be linked to neuronal activity to ensure crosstalk between cell types during the development and function of the central nervous system.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Hannah M. Wüst
- Institut für Biochemie, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (C.P.); (T.A.); (E.S.); (J.B.); (M.K.); (S.H.)
| | - Michael Wegner
- Institut für Biochemie, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (C.P.); (T.A.); (E.S.); (J.B.); (M.K.); (S.H.)
| |
Collapse
|
2
|
Cui SH, Suo N, Yang Y, Wu X, Guo SM, Xie X. The aminosteroid U73122 promotes oligodendrocytes generation and myelin formation. Acta Pharmacol Sin 2024; 45:490-501. [PMID: 37935896 PMCID: PMC10834981 DOI: 10.1038/s41401-023-01183-7] [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: 06/29/2023] [Accepted: 10/13/2023] [Indexed: 11/09/2023] Open
Abstract
Oligodendrocytes (OLs) are glial cells that ensheath neuronal axons and form myelin in the central nervous system (CNS). OLs are differentiated from oligodendrocyte precursor cells (OPCs) during development and myelin repair, which is often insufficient in the latter case in demyelinating diseases such as multiple sclerosis (MS). Many factors have been reported to regulate OPC-to-OL differentiation, including a number of G protein-coupled receptors (GPCRs). In an effort to search pathways downstream of GPCRs that might be involved in OPC differentiation, we discover that U73122, a phosphoinositide specific phospholipase C (PI-PLC) inhibitor, dramatically promotes OPC-to-OL differentiation and myelin regeneration in experimental autoimmune encephalomyelitis model. Unexpectedly, U73343, a close analog of U73122 which lacks PI-PLC inhibitory activity also promotes OL differentiation, while another reported PI-PLC inhibitor edelfosine does not have such effect, suggesting that U73122 and U73343 enhance OPC differentiation independent of PLC. Although the structures of U73122 and U73343 closely resemble 17β-estradiol, and both compounds do activate estrogen receptors Erα and Erβ with low efficacy and potency, further study indicates that these compounds do not act through Erα and/or Erβ to promote OPC differentiation. RNA-Seq and bioinformatic analysis indicate that U73122 and U73343 may regulate cholesterol biosynthesis. Further study shows both compounds increase 14-dehydrozymostenol, a steroid reported to promote OPC differentiation, in OPC culture. In conclusion, the aminosteroids U73122 and U73343 promote OPC-to-OL generation and myelin formation by regulating cholesterol biosynthesis pathway.
Collapse
Affiliation(s)
- Shi-Hao Cui
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Na Suo
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Ying Yang
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Xuan Wu
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Shi-Meng Guo
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xin Xie
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China.
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China.
| |
Collapse
|
3
|
Benarroch E. What Are the Roles of Oligodendrocyte Precursor Cells in Normal and Pathologic Conditions? Neurology 2023; 101:958-965. [PMID: 37985182 PMCID: PMC10663025 DOI: 10.1212/wnl.0000000000208000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 09/25/2023] [Indexed: 11/22/2023] Open
|
4
|
Muttathukunnel P, Wälti M, Aboouf MA, Köster-Hegmann C, Haenggi T, Gassmann M, Pannzanelli P, Fritschy JM, Schneider Gasser EM. Erythropoietin regulates developmental myelination in the brain stimulating postnatal oligodendrocyte maturation. Sci Rep 2023; 13:19522. [PMID: 37945644 PMCID: PMC10636124 DOI: 10.1038/s41598-023-46783-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 11/05/2023] [Indexed: 11/12/2023] Open
Abstract
Myelination is a process tightly regulated by a variety of neurotrophic factors. Here, we show-by analyzing two transgenic mouse lines, one overexpressing EPO selectively in the brain Tg21(PDGFB-rhEPO) and another with targeted removal of EPO receptors (EPORs) from oligodendrocyte progenitor cells (OPC)s (Sox10-cre;EpoRfx/fx mice)-a key function for EPO in regulating developmental brain myelination. Overexpression of EPO resulted in faster postnatal brain growth and myelination, an increased number of myelinating oligodendrocytes, faster axonal myelin ensheathment, and improved motor coordination. Conversely, targeted ablation of EPORs from OPCs reduced the number of mature oligodendrocytes and impaired motor coordination during the second postnatal week. Furthermore, we found that EPORs are transiently expressed in the subventricular zone (SVZ) during the second postnatal week and EPO increases the postnatal expression of essential oligodendrocyte pro-differentiation and pro-maturation (Nkx6.2 and Myrf) transcripts, and the Nfatc2/calcineurin pathway. In contrast, ablation of EPORs from OPCs inactivated the Erk1/2 pathway and reduced the postnatal expression of the transcripts. Our results reveal developmental time windows in which EPO therapies could be highly effective for stimulating oligodendrocyte maturation and myelination.
Collapse
Affiliation(s)
- Paola Muttathukunnel
- Institute of Pharmacology and Toxicology, University of Zürich, 8057, Zürich, Switzerland
- Center for Neuroscience Zurich (ZNZ), Zurich, Switzerland
| | - Michael Wälti
- Institute of Pharmacology and Toxicology, University of Zürich, 8057, Zürich, Switzerland
| | - Mostafa A Aboouf
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zürich, 8057, Zurich, Switzerland
- Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, 8057, Zurich, Switzerland
- Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt
| | - Christina Köster-Hegmann
- Institute of Pharmacology and Toxicology, University of Zürich, 8057, Zürich, Switzerland
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zürich, 8057, Zurich, Switzerland
| | - Tatjana Haenggi
- Institute of Pharmacology and Toxicology, University of Zürich, 8057, Zürich, Switzerland
| | - Max Gassmann
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zürich, 8057, Zurich, Switzerland
- Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, 8057, Zurich, Switzerland
| | - Patrizia Pannzanelli
- Rita Levi Montalcini Center for Brain Repair, University of Turin, 10126, Turin, Italy
| | - Jean-Marc Fritschy
- Institute of Pharmacology and Toxicology, University of Zürich, 8057, Zürich, Switzerland
- Center for Neuroscience Zurich (ZNZ), Zurich, Switzerland
| | - Edith M Schneider Gasser
- Institute of Pharmacology and Toxicology, University of Zürich, 8057, Zürich, Switzerland.
- Center for Neuroscience Zurich (ZNZ), Zurich, Switzerland.
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zürich, 8057, Zurich, Switzerland.
| |
Collapse
|
5
|
Yoshida K, Kato D, Sugio S, Takeda I, Wake H. Activity-dependent oligodendrocyte calcium dynamics and their changes in Alzheimer's disease. Front Cell Neurosci 2023; 17:1154196. [PMID: 38026691 PMCID: PMC10644703 DOI: 10.3389/fncel.2023.1154196] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 10/11/2023] [Indexed: 12/01/2023] Open
Abstract
Oligodendrocytes (OCs) form myelin around axons, which is dependent on neuronal activity. This activity-dependent myelination plays a crucial role in training and learning. Previous studies have suggested that neuronal activity regulates proliferation and differentiation of oligodendrocyte precursor cells (OPCs) and myelination. In addition, deficient activity-dependent myelination results in impaired motor learning. However, the functional response of OC responsible for neuronal activity and their pathological changes is not fully elucidated. In this research, we aimed to understand the activity-dependent OC responses and their different properties by observing OCs using in vivo two-photon microscopy. We clarified that the Ca2+ activity in OCs is neuronal activity dependent and differentially regulated by neurotransmitters such as glutamate or adenosine triphosphate (ATP). Furthermore, in 5-month-old mice models of Alzheimer's disease, a period before the appearance of behavioral abnormalities, the elevated Ca2+ responses in OCs are ATP dependent, suggesting that OCs receive ATP from damaged tissue. We anticipate that our research will help in determining the correct therapeutic strategy for neurodegenerative diseases beyond the synapse.
Collapse
Affiliation(s)
- Kenji Yoshida
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Daisuke Kato
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Division of Multicellular Circuit Dynamics, National Institute for Physiological Sciences, National Institute of Natural Sciences, Okazaki, Japan
| | - Shouta Sugio
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Division of Multicellular Circuit Dynamics, National Institute for Physiological Sciences, National Institute of Natural Sciences, Okazaki, Japan
| | - Ikuko Takeda
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Division of Multicellular Circuit Dynamics, National Institute for Physiological Sciences, National Institute of Natural Sciences, Okazaki, Japan
| | - Hiroaki Wake
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Division of Multicellular Circuit Dynamics, National Institute for Physiological Sciences, National Institute of Natural Sciences, Okazaki, Japan
- Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Saitama, Japan
| |
Collapse
|
6
|
Hyung S, Park JH, Jung K. Application of optogenetic glial cells to neuron-glial communication. Front Cell Neurosci 2023; 17:1249043. [PMID: 37868193 PMCID: PMC10585272 DOI: 10.3389/fncel.2023.1249043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 09/15/2023] [Indexed: 10/24/2023] Open
Abstract
Optogenetic techniques combine optics and genetics to enable cell-specific targeting and precise spatiotemporal control of excitable cells, and they are increasingly being employed. One of the most significant advantages of the optogenetic approach is that it allows for the modulation of nearby cells or circuits with millisecond precision, enabling researchers to gain a better understanding of the complex nervous system. Furthermore, optogenetic neuron activation permits the regulation of information processing in the brain, including synaptic activity and transmission, and also promotes nerve structure development. However, the optimal conditions remain unclear, and further research is required to identify the types of cells that can most effectively and precisely control nerve function. Recent studies have described optogenetic glial manipulation for coordinating the reciprocal communication between neurons and glia. Optogenetically stimulated glial cells can modulate information processing in the central nervous system and provide structural support for nerve fibers in the peripheral nervous system. These advances promote the effective use of optogenetics, although further experiments are needed. This review describes the critical role of glial cells in the nervous system and reviews the optogenetic applications of several types of glial cells, as well as their significance in neuron-glia interactions. Together, it briefly discusses the therapeutic potential and feasibility of optogenetics.
Collapse
Affiliation(s)
- Sujin Hyung
- Precision Medicine Research Institute, Samsung Medical Center, Seoul, Republic of Korea
- Division of Hematology-Oncology, Department of Medicine, Sungkyunkwan University, Samsung Medical Center, Seoul, Republic of Korea
| | - Ji-Hye Park
- Graduate School of Cancer Science and Policy, Cancer Biomedical Science, National Cancer Center, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Kyuhwan Jung
- DAWINBIO Inc., Hanam-si, Gyeonggi-do, Republic of Korea
| |
Collapse
|
7
|
Cheli VT, Santiago González DA, Wan R, Rosenblum SL, Denaroso GE, Angeliu CG, Smith Z, Wang C, Paez PM. Transferrin Receptor Is Necessary for Proper Oligodendrocyte Iron Homeostasis and Development. J Neurosci 2023; 43:3614-3629. [PMID: 36977582 PMCID: PMC10198458 DOI: 10.1523/jneurosci.1383-22.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 03/15/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023] Open
Abstract
To test the hypothesis that the transferrin (Tf) cycle has unique importance for oligodendrocyte development and function, we disrupted the expression of the Tf receptor (Tfr) gene in oligodendrocyte progenitor cells (OPCs) on mice of either sex using the Cre/lox system. This ablation results in the elimination of iron incorporation via the Tf cycle but leaves other Tf functions intact. Mice lacking Tfr, specifically in NG2 or Sox10-positive OPCs, developed a hypomyelination phenotype. Both OPC differentiation and myelination were affected, and Tfr deletion resulted in impaired OPC iron absorption. Specifically, the brains of Tfr cKO animals presented a reduction in the quantity of myelinated axons, as well as fewer mature oligodendrocytes. In contrast, the ablation of Tfr in adult mice affected neither mature oligodendrocytes nor myelin synthesis. RNA-seq analysis performed in Tfr cKO OPCs revealed misregulated genes involved in OPC maturation, myelination, and mitochondrial activity. Tfr deletion in cortical OPCs also disrupted the activity of the mTORC1 signaling pathway, epigenetic mechanisms critical for gene transcription and the expression of structural mitochondrial genes. RNA-seq studies were additionally conducted in OPCs in which iron storage was disrupted by deleting the ferritin heavy chain. These OPCs display abnormal regulation of genes associated with iron transport, antioxidant activity, and mitochondrial activity. Thus, our results indicate that the Tf cycle is central for iron homeostasis in OPCs during postnatal development and suggest that both iron uptake via Tfr and iron storage in ferritin are critical for energy production, mitochondrial activity, and maturation of postnatal OPCs.SIGNIFICANCE STATEMENT By knocking-out transferrin receptor (Tfr) specifically in oligodendrocyte progenitor cells (OPCs), we have established that iron incorporation via the Tf cycle is key for OPC iron homeostasis and for the normal function of these cells during the postnatal development of the CNS. Moreover, RNA-seq analysis indicated that both Tfr iron uptake and ferritin iron storage are critical for proper OPC mitochondrial activity, energy production, and maturation.
Collapse
Affiliation(s)
- Veronica T Cheli
- Institute for Myelin and Glia Exploration, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, State University of New York, University at Buffalo, Buffalo, New York 14203
| | - Diara A Santiago González
- Institute for Myelin and Glia Exploration, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, State University of New York, University at Buffalo, Buffalo, New York 14203
| | - Rensheng Wan
- Institute for Myelin and Glia Exploration, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, State University of New York, University at Buffalo, Buffalo, New York 14203
| | - Shaina L Rosenblum
- Institute for Myelin and Glia Exploration, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, State University of New York, University at Buffalo, Buffalo, New York 14203
| | - Giancarlo E Denaroso
- Institute for Myelin and Glia Exploration, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, State University of New York, University at Buffalo, Buffalo, New York 14203
| | - Christina G Angeliu
- Institute for Myelin and Glia Exploration, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, State University of New York, University at Buffalo, Buffalo, New York 14203
| | - Zachary Smith
- Institute for Myelin and Glia Exploration, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, State University of New York, University at Buffalo, Buffalo, New York 14203
| | - Congying Wang
- Institute for Myelin and Glia Exploration, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, State University of New York, University at Buffalo, Buffalo, New York 14203
| | - Pablo M Paez
- Institute for Myelin and Glia Exploration, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, State University of New York, University at Buffalo, Buffalo, New York 14203
| |
Collapse
|
8
|
Al-Griw MA, Alghazeer R, Ratemi HW, Ben-Othman ME, Tabagah R, Shamlan G, Habibullah MM, Alnajeebi AM, Babteen NA, Eskandrani AA, Al-Farga A, Alansari WS. Blockade of L-Type Ca 2+ Channel Activity Alleviates Oligodendrocyte Pathology following Brain Injury in Male Rats. Curr Issues Mol Biol 2023; 45:3953-3964. [PMID: 37232721 DOI: 10.3390/cimb45050252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/09/2023] [Accepted: 04/16/2023] [Indexed: 05/27/2023] Open
Abstract
A growing body of studies suggests that Ca2+ signaling controls a variety of biological processes in brain elements. Activation of L-type voltage-operated Ca2+ channels (VOCCs) plays a role in the development of oligodendrocyte (OL) lineage loss, and indicates that the blocking of these channels may be an effective way to inhibit OL lineage cell loss. For this study, 10.5-day-old male Sprague-Dawley rats were used to generate cerebellar tissue slices. The slice tissues were cultured and randomly allocated to one of four groups (six each) and treated as follows: Group I, (sham control); Group II, 0.1% dimethyl sulfoxide (DMSO) only (vehicle control); Group III, injury (INJ); Group IV, (INJ and treatment with NIF). The injury was simulated by exposing the slice tissues to 20 min of oxygen-glucose deprivation (OGD). At 3 days post-treatment, the survival, apoptosis, and proliferation of the OL lineages were measured and compared. Results: In the INJ group, there was a decrease in mature myelin basic protein+ OLs (MBP+ OLs) and their precursors, NG2+ OPCs (Nerve-glia antigen 2+ oligodendrocyte precursor cell), compared with controls. A significant elevation was observed in the NG2+ OPCs and apoptotic MBP+ OLs as confirmed by a TUNEL assay. However, the cell proliferation rate was decreased in NG2+ OPCs. NIF increased OL survival as measured by apoptosis rate in both OL lineages and preserved the rate of proliferation in the NG2+ OPCs. Conclusions: Activation of L-type VOCCs may contribute to OL pathology in association with reduced mitosis of OPCs following brain injury as a strategy to treat demyelinating diseases.
Collapse
Affiliation(s)
- Mohamed A Al-Griw
- Department of Histology and Genetics, Faculty of Medicine, University of Tripoli, Tripoli 13203, Libya
| | - Rabia Alghazeer
- Department of Chemistry, Faculty of Science, University of Tripoli, Tripoli 50676, Libya
| | - Haithm W Ratemi
- Department of Genetic Engineering, Biotechnology Research Center (BTRC), Tripoli 30313, Libya
| | - Mohamed E Ben-Othman
- Department of Clinical Pathology, Faculty of Veterinary Medicine, University of Tripoli, Tripoli 13662, Libya
| | - Refaat Tabagah
- Division Developmental Biology, Zoology Department, Faculty of Sciences, University of Tripoli, Tripoli 13662, Libya
| | - Ghalia Shamlan
- Department of Food Science and Nutrition, College of Food and Agriculture Sciences, King Saud University, Riyadh 11362, Saudi Arabia
| | - Mahmmoud M Habibullah
- Medical Laboratory Technology Department, Faculty of Applied Medical Sciences, Jazan University, Jazan 45142, Saudi Arabia
| | - Afnan M Alnajeebi
- Biochemistry Department, Faculty of Science, University of Jeddah, Jeddah 21577, Saudi Arabia
| | - Nouf A Babteen
- Biochemistry Department, Faculty of Science, University of Jeddah, Jeddah 21577, Saudi Arabia
| | - Areej A Eskandrani
- Chemistry Department, Faculty of Science, Taibah University, Medina 30002, Saudi Arabia
| | - Ammar Al-Farga
- Biochemistry Department, Faculty of Science, University of Jeddah, Jeddah 21577, Saudi Arabia
| | - Wafa S Alansari
- Biochemistry Department, Faculty of Science, University of Jeddah, Jeddah 21577, Saudi Arabia
| |
Collapse
|
9
|
Glial Cell Metabolic Profile Upon Iron Deficiency: Oligodendroglial and Astroglial Casualties of Bioenergetic Adjustments. Mol Neurobiol 2023; 60:1949-1963. [PMID: 36595194 DOI: 10.1007/s12035-022-03149-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 11/24/2022] [Indexed: 01/04/2023]
Abstract
Iron deficiency (ID) represents one of the most prevalent nutritional deficits, affecting almost two billion people worldwide. Gestational iron deprivation induces hypomyelination due to oligodendroglial maturation deficiencies and is thus a useful experimental model to analyze oligodendrocyte (OLG) requirements to progress to a mature myelinating state. A previous proteomic study in the adult ID brain by our group demonstrated a pattern of dysregulated proteins involved in the tricarboxylic acid cycle and mitochondrial dysfunction. The aim of the present report was to assess bioenergetics metabolism in primary cultures of OLGs and astrocytes (ASTs) from control and ID newborns, on the hypothesis that the regulation of cell metabolism correlates with cell maturation. Oxygen consumption and extracellular acidification rates were measured using a Seahorse extracellular flux analyzer. ID OLGs and ASTs both exhibited decreased spare respiratory capacity, which indicates that ID effectively induces mitochondrial dysfunction. A decrease in glycogen granules was observed in ID ASTs, and an increase in ROS production was detected in ID OLGs. Immunolabeling of structural proteins showed that mitochondrial number and size were increased in ID OLGs, while an increased number of smaller mitochondria was observed in ID ASTs. These results reflect an unfavorable bioenergetic scenario in which ID OLGs fail to progress to a myelinating state, and indicate that the regulation of cell metabolism may impact cell fate decisions and maturation.
Collapse
|
10
|
Impact of the Voltage-Gated Calcium Channel Antagonist Nimodipine on the Development of Oligodendrocyte Precursor Cells. Int J Mol Sci 2023; 24:ijms24043716. [PMID: 36835129 PMCID: PMC9960570 DOI: 10.3390/ijms24043716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 01/31/2023] [Accepted: 02/06/2023] [Indexed: 02/17/2023] Open
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system (CNS). While most of the current treatment strategies focus on immune cell regulation, except for the drug siponimod, there is no therapeutic intervention that primarily aims at neuroprotection and remyelination. Recently, nimodipine showed a beneficial and remyelinating effect in experimental autoimmune encephalomyelitis (EAE), a mouse model of MS. Nimodipine also positively affected astrocytes, neurons, and mature oligodendrocytes. Here we investigated the effects of nimodipine, an L-type voltage-gated calcium channel antagonist, on the expression profile of myelin genes and proteins in the oligodendrocyte precursor cell (OPC) line Oli-Neu and in primary OPCs. Our data indicate that nimodipine does not have any effect on myelin-related gene and protein expression. Furthermore, nimodipine treatment did not result in any morphological changes in these cells. However, RNA sequencing and bioinformatic analyses identified potential micro (mi)RNA that could support myelination after nimodipine treatment compared to a dimethyl sulfoxide (DMSO) control. Additionally, we treated zebrafish with nimodipine and observed a significant increase in the number of mature oligodendrocytes (* p≤ 0.05). Taken together, nimodipine seems to have different positive effects on OPCs and mature oligodendrocytes.
Collapse
|
11
|
Arjun McKinney A, Petrova R, Panagiotakos G. Calcium and activity-dependent signaling in the developing cerebral cortex. Development 2022; 149:dev198853. [PMID: 36102617 PMCID: PMC9578689 DOI: 10.1242/dev.198853] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Calcium influx can be stimulated by various intra- and extracellular signals to set coordinated gene expression programs into motion. As such, the precise regulation of intracellular calcium represents a nexus between environmental cues and intrinsic genetic programs. Mounting genetic evidence points to a role for the deregulation of intracellular calcium signaling in neuropsychiatric disorders of developmental origin. These findings have prompted renewed enthusiasm for understanding the roles of calcium during normal and dysfunctional prenatal development. In this Review, we describe the fundamental mechanisms through which calcium is spatiotemporally regulated and directs early neurodevelopmental events. We also discuss unanswered questions about intracellular calcium regulation during the emergence of neurodevelopmental disease, and provide evidence that disruption of cell-specific calcium homeostasis and/or redeployment of developmental calcium signaling mechanisms may contribute to adult neurological disorders. We propose that understanding the normal developmental events that build the nervous system will rely on gaining insights into cell type-specific calcium signaling mechanisms. Such an understanding will enable therapeutic strategies targeting calcium-dependent mechanisms to mitigate disease.
Collapse
Affiliation(s)
- Arpana Arjun McKinney
- Graduate Program in Developmental and Stem Cell Biology, University of California, San Francisco, CA 94143, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA 94143, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94143, USA
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, CA 94143, USA
| | - Ralitsa Petrova
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA 94143, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94143, USA
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, CA 94143, USA
| | - Georgia Panagiotakos
- Graduate Program in Developmental and Stem Cell Biology, University of California, San Francisco, CA 94143, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA 94143, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94143, USA
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
12
|
Nimodipine Exerts Beneficial Effects on the Rat Oligodendrocyte Cell Line OLN-93. Brain Sci 2022; 12:brainsci12040476. [PMID: 35448007 PMCID: PMC9029615 DOI: 10.3390/brainsci12040476] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 03/29/2022] [Accepted: 03/30/2022] [Indexed: 02/04/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic autoimmune disease of the central nervous system (CNS). Therapy is currently limited to drugs that interfere with the immune system; treatment options that primarily mediate neuroprotection and prevent neurodegeneration are not available. Here, we studied the effects of nimodipine on the rat cell line OLN-93, which resembles young mature oligodendrocytes. Nimodipine is a dihydropyridine that blocks the voltage-gated L-type calcium channel family members Cav1.2 and Cav1.3. Our data show that the treatment of OLN-93 cells with nimodipine induced the upregulation of myelin genes, in particular of proteolipid protein 1 (Plp1), which was confirmed by a significantly greater expression of PLP1 in immunofluorescence analysis and the presence of myelin structures in the cytoplasm at the ultrastructural level. Whole-genome RNA sequencing additionally revealed the upregulation of genes that are involved in neuroprotection, remyelination, and antioxidation pathways. Interestingly, the observed effects were independent of Cav1.2 and Cav1.3 because OLN-93 cells do not express these channels, and there was no measurable response pattern in patch-clamp analysis. Taking into consideration previous studies that demonstrated a beneficial effect of nimodipine on microglia, our data support the notion that nimodipine is an interesting drug candidate for the treatment of MS and other demyelinating diseases.
Collapse
|
13
|
Moura DMS, Brennan EJ, Brock R, Cocas LA. Neuron to Oligodendrocyte Precursor Cell Synapses: Protagonists in Oligodendrocyte Development and Myelination, and Targets for Therapeutics. Front Neurosci 2022; 15:779125. [PMID: 35115904 PMCID: PMC8804499 DOI: 10.3389/fnins.2021.779125] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 12/17/2021] [Indexed: 12/17/2022] Open
Abstract
The development of neuronal circuitry required for cognition, complex motor behaviors, and sensory integration requires myelination. The role of glial cells such as astrocytes and microglia in shaping synapses and circuits have been covered in other reviews in this journal and elsewhere. This review summarizes the role of another glial cell type, oligodendrocytes, in shaping synapse formation, neuronal circuit development, and myelination in both normal development and in demyelinating disease. Oligodendrocytes ensheath and insulate neuronal axons with myelin, and this facilitates fast conduction of electrical nerve impulses via saltatory conduction. Oligodendrocytes also proliferate during postnatal development, and defects in their maturation have been linked to abnormal myelination. Myelination also regulates the timing of activity in neural circuits and is important for maintaining the health of axons and providing nutritional support. Recent studies have shown that dysfunction in oligodendrocyte development and in myelination can contribute to defects in neuronal synapse formation and circuit development. We discuss glutamatergic and GABAergic receptors and voltage gated ion channel expression and function in oligodendrocyte development and myelination. We explain the role of excitatory and inhibitory neurotransmission on oligodendrocyte proliferation, migration, differentiation, and myelination. We then focus on how our understanding of the synaptic connectivity between neurons and OPCs can inform future therapeutics in demyelinating disease, and discuss gaps in the literature that would inform new therapies for remyelination.
Collapse
Affiliation(s)
- Daniela M. S. Moura
- Department of Biology, Santa Clara University, Santa Clara, CA, United States
| | - Emma J. Brennan
- Department of Biology, Santa Clara University, Santa Clara, CA, United States
| | - Robert Brock
- Department of Biology, Santa Clara University, Santa Clara, CA, United States
| | - Laura A. Cocas
- Department of Biology, Santa Clara University, Santa Clara, CA, United States
- Department of Neurology, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
14
|
Milbocker KA, Campbell TS, Collins N, Kim S, Smith IF, Roth TL, Klintsova AY. Glia-Driven Brain Circuit Refinement Is Altered by Early-Life Adversity: Behavioral Outcomes. Front Behav Neurosci 2021; 15:786234. [PMID: 34924972 PMCID: PMC8678604 DOI: 10.3389/fnbeh.2021.786234] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/12/2021] [Indexed: 12/12/2022] Open
Abstract
Early-life adversity (ELA), often clinically referred to as "adverse childhood experiences (ACE)," is the exposure to stress-inducing events in childhood that can result in poor health outcomes. ELA negatively affects neurodevelopment in children and adolescents resulting in several behavioral deficits and increasing the risk of developing a myriad of neuropsychiatric disorders later in life. The neurobiological mechanisms by which ELA alters neurodevelopment in childhood have been the focus of numerous reviews. However, a comprehensive review of the mechanisms affecting adolescent neurodevelopment (i.e., synaptic pruning and myelination) is lacking. Synaptic pruning and myelination are glia-driven processes that are imperative for brain circuit refinement during the transition from adolescence to adulthood. Failure to optimize brain circuitry between key brain structures involved in learning and memory, such as the hippocampus and prefrontal cortex, leads to the emergence of maladaptive behaviors including increased anxiety or reduced executive function. As such, we review preclinical and clinical literature to explore the immediate and lasting effects of ELA on brain circuit development and refinement. Finally, we describe a number of therapeutic interventions best-suited to support adolescent neurodevelopment in children with a history of ELA.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Anna Y. Klintsova
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE, United States
| |
Collapse
|
15
|
Ten-eleven translocation 1 mediated-DNA hydroxymethylation is required for myelination and remyelination in the mouse brain. Nat Commun 2021; 12:5091. [PMID: 34429415 PMCID: PMC8385008 DOI: 10.1038/s41467-021-25353-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 08/05/2021] [Indexed: 11/08/2022] Open
Abstract
Ten-eleven translocation (TET) proteins, the dioxygenase for DNA hydroxymethylation, are important players in nervous system development and diseases. However, their role in myelination and remyelination after injury remains elusive. Here, we identify a genome-wide and locus-specific DNA hydroxymethylation landscape shift during differentiation of oligodendrocyte-progenitor cells (OPC). Ablation of Tet1 results in stage-dependent defects in oligodendrocyte (OL) development and myelination in the mouse brain. The mice lacking Tet1 in the oligodendrocyte lineage develop behavioral deficiency. We also show that TET1 is required for remyelination in adulthood. Transcriptomic, genomic occupancy, and 5-hydroxymethylcytosine (5hmC) profiling reveal a critical TET1-regulated epigenetic program for oligodendrocyte differentiation that includes genes associated with myelination, cell division, and calcium transport. Tet1-deficient OPCs exhibit reduced calcium activity, increasing calcium activity rescues the differentiation defects in vitro. Deletion of a TET1-5hmC target gene, Itpr2, impairs the onset of OPC differentiation. Together, our results suggest that stage-specific TET1-mediated epigenetic programming and intracellular signaling are important for proper myelination and remyelination in mice.
Collapse
|
16
|
Ion Channels as New Attractive Targets to Improve Re-Myelination Processes in the Brain. Int J Mol Sci 2021; 22:ijms22147277. [PMID: 34298893 PMCID: PMC8305962 DOI: 10.3390/ijms22147277] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/28/2021] [Accepted: 07/01/2021] [Indexed: 12/20/2022] Open
Abstract
Multiple sclerosis (MS) is the most demyelinating disease of the central nervous system (CNS) characterized by neuroinflammation. Oligodendrocyte progenitor cells (OPCs) are cycling cells in the developing and adult CNS that, under demyelinating conditions, migrate to the site of lesions and differentiate into mature oligodendrocytes to remyelinate damaged axons. However, this process fails during disease chronicization due to impaired OPC differentiation. Moreover, OPCs are crucial players in neuro-glial communication as they receive synaptic inputs from neurons and express ion channels and neurotransmitter/neuromodulator receptors that control their maturation. Ion channels are recognized as attractive therapeutic targets, and indeed ligand-gated and voltage-gated channels can both be found among the top five pharmaceutical target groups of FDA-approved agents. Their modulation ameliorates some of the symptoms of MS and improves the outcome of related animal models. However, the exact mechanism of action of ion-channel targeting compounds is often still unclear due to the wide expression of these channels on neurons, glia, and infiltrating immune cells. The present review summarizes recent findings in the field to get further insights into physio-pathophysiological processes and possible therapeutic mechanisms of drug actions.
Collapse
|
17
|
Boscia F, Elkjaer ML, Illes Z, Kukley M. Altered Expression of Ion Channels in White Matter Lesions of Progressive Multiple Sclerosis: What Do We Know About Their Function? Front Cell Neurosci 2021; 15:685703. [PMID: 34276310 PMCID: PMC8282214 DOI: 10.3389/fncel.2021.685703] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 05/23/2021] [Indexed: 12/19/2022] Open
Abstract
Despite significant advances in our understanding of the pathophysiology of multiple sclerosis (MS), knowledge about contribution of individual ion channels to axonal impairment and remyelination failure in progressive MS remains incomplete. Ion channel families play a fundamental role in maintaining white matter (WM) integrity and in regulating WM activities in axons, interstitial neurons, glia, and vascular cells. Recently, transcriptomic studies have considerably increased insight into the gene expression changes that occur in diverse WM lesions and the gene expression fingerprint of specific WM cells associated with secondary progressive MS. Here, we review the ion channel genes encoding K+, Ca2+, Na+, and Cl- channels; ryanodine receptors; TRP channels; and others that are significantly and uniquely dysregulated in active, chronic active, inactive, remyelinating WM lesions, and normal-appearing WM of secondary progressive MS brain, based on recently published bulk and single-nuclei RNA-sequencing datasets. We discuss the current state of knowledge about the corresponding ion channels and their implication in the MS brain or in experimental models of MS. This comprehensive review suggests that the intense upregulation of voltage-gated Na+ channel genes in WM lesions with ongoing tissue damage may reflect the imbalance of Na+ homeostasis that is observed in progressive MS brain, while the upregulation of a large number of voltage-gated K+ channel genes may be linked to a protective response to limit neuronal excitability. In addition, the altered chloride homeostasis, revealed by the significant downregulation of voltage-gated Cl- channels in MS lesions, may contribute to an altered inhibitory neurotransmission and increased excitability.
Collapse
Affiliation(s)
- Francesca Boscia
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, University of Naples "Federico II", Naples, Italy
| | - Maria Louise Elkjaer
- Neurology Research Unit, Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Zsolt Illes
- Neurology Research Unit, Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark.,Department of Neurology, Odense University Hospital, Odense, Denmark
| | - Maria Kukley
- Achucarro Basque Center for Neuroscience, Leioa, Spain.,Ikerbasque Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
18
|
Piller M, Werkman IL, Brown RI, Latimer AJ, Kucenas S. Glutamate Signaling via the AMPAR Subunit GluR4 Regulates Oligodendrocyte Progenitor Cell Migration in the Developing Spinal Cord. J Neurosci 2021; 41:5353-5371. [PMID: 33975920 PMCID: PMC8221590 DOI: 10.1523/jneurosci.2562-20.2021] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 04/28/2021] [Accepted: 05/03/2021] [Indexed: 11/21/2022] Open
Abstract
Oligodendrocyte progenitor cells (OPCs) are specified from discrete precursor populations during gliogenesis and migrate extensively from their origins, ultimately distributing throughout the brain and spinal cord during early development. Subsequently, a subset of OPCs differentiates into mature oligodendrocytes, which myelinate axons. This process is necessary for efficient neuronal signaling and organism survival. Previous studies have identified several factors that influence OPC development, including excitatory glutamatergic synapses that form between neurons and OPCs during myelination. However, little is known about how glutamate signaling affects OPC migration before myelination. In this study, we use in vivo, time-lapse imaging in zebrafish in conjunction with genetic and pharmacological perturbation to investigate OPC migration and myelination when the GluR4A ionotropic glutamate receptor subunit is disrupted. In our studies, we observed that gria4a mutant embryos and larvae displayed abnormal OPC migration and altered dorsoventral distribution in the spinal cord. Genetic mosaic analysis confirmed that these effects were cell-autonomous, and we identified that voltage-gated calcium channels were downstream of glutamate receptor signaling in OPCs and could rescue the migration and myelination defects we observed when glutamate signaling was perturbed. These results offer new insights into the complex system of neuron-OPC interactions and reveal a cell-autonomous role for glutamatergic signaling in OPCs during neural development.SIGNIFICANCE STATEMENT The migration of oligodendrocyte progenitor cells (OPCs) is an essential process during development that leads to uniform oligodendrocyte distribution and sufficient myelination for central nervous system function. Here, we demonstrate that the AMPA receptor (AMPAR) subunit GluR4A is an important driver of OPC migration and myelination in vivo and that activated voltage-gated calcium channels are downstream of glutamate receptor signaling in mediating this migration.
Collapse
Affiliation(s)
- Melanie Piller
- Department of Biology, University of Virginia, Charlottesville, Virginia 22904
| | - Inge L Werkman
- Department of Biology, University of Virginia, Charlottesville, Virginia 22904
| | - Robin Isadora Brown
- Department of Biology, University of Virginia, Charlottesville, Virginia 22904
| | - Andrew J Latimer
- Department of Biology, University of Virginia, Charlottesville, Virginia 22904
| | - Sarah Kucenas
- Department of Biology, University of Virginia, Charlottesville, Virginia 22904
| |
Collapse
|
19
|
Windrem MS, Schanz SJ, Zou L, Chandler-Militello D, Kuypers NJ, Nedergaard M, Lu Y, Mariani JN, Goldman SA. Human Glial Progenitor Cells Effectively Remyelinate the Demyelinated Adult Brain. Cell Rep 2021; 31:107658. [PMID: 32433967 DOI: 10.1016/j.celrep.2020.107658] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 02/14/2020] [Accepted: 04/18/2020] [Indexed: 12/12/2022] Open
Abstract
Neonatally transplanted human glial progenitor cells (hGPCs) can myelinate the brains of myelin-deficient shiverer mice, rescuing their phenotype and survival. Yet, it has been unclear whether implanted hGPCs are similarly able to remyelinate the diffusely demyelinated adult CNS. We, therefore, ask if hGPCs could remyelinate both congenitally hypomyelinated adult shiverers and normal adult mice after cuprizone demyelination. In adult shiverers, hGPCs broadly disperse and differentiate as myelinating oligodendrocytes after subcortical injection, improving both host callosal conduction and ambulation. Implanted hGPCs similarly remyelinate denuded axons after cuprizone demyelination, whether delivered before or after demyelination. RNA sequencing (RNA-seq) of hGPCs back from cuprizone-demyelinated brains reveals their transcriptional activation of oligodendrocyte differentiation programs, while distinguishing them from hGPCs not previously exposed to demyelination. These data indicate the ability of transplanted hGPCs to disperse throughout the adult CNS, to broadly myelinate regions of dysmyelination, and also to be recruited as myelinogenic oligodendrocytes later in life, upon demyelination-associated demand.
Collapse
Affiliation(s)
- Martha S Windrem
- Center for Translational Neuromedicine and the Department of Neurology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Steven J Schanz
- Center for Translational Neuromedicine and the Department of Neurology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Lisa Zou
- Center for Translational Neuromedicine and the Department of Neurology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Devin Chandler-Militello
- Center for Translational Neuromedicine and the Department of Neurology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Nicholas J Kuypers
- Center for Translational Neuromedicine and the Department of Neurology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Maiken Nedergaard
- Center for Translational Neuromedicine and the Department of Neurology, University of Rochester Medical Center, Rochester, NY 14642, USA; Center for Translational Neuromedicine, University of Copenhagen, Copenhagen, Denmark
| | - Yuan Lu
- Center for Translational Neuromedicine and the Department of Neurology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - John N Mariani
- Center for Translational Neuromedicine and the Department of Neurology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Steven A Goldman
- Center for Translational Neuromedicine and the Department of Neurology, University of Rochester Medical Center, Rochester, NY 14642, USA; Center for Translational Neuromedicine, University of Copenhagen, Copenhagen, Denmark; Neuroscience Center, Rigshospitalet, Copenhagen, Denmark.
| |
Collapse
|
20
|
Galichet C, Clayton RW, Lovell-Badge R. Novel Tools and Investigative Approaches for the Study of Oligodendrocyte Precursor Cells (NG2-Glia) in CNS Development and Disease. Front Cell Neurosci 2021; 15:673132. [PMID: 33994951 PMCID: PMC8116629 DOI: 10.3389/fncel.2021.673132] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 04/07/2021] [Indexed: 12/11/2022] Open
Abstract
Oligodendrocyte progenitor cells (OPCs), also referred to as NG2-glia, are the most proliferative cell type in the adult central nervous system. While the primary role of OPCs is to serve as progenitors for oligodendrocytes, in recent years, it has become increasingly clear that OPCs fulfil a number of other functions. Indeed, independent of their role as stem cells, it is evident that OPCs can regulate the metabolic environment, directly interact with and modulate neuronal function, maintain the blood brain barrier (BBB) and regulate inflammation. In this review article, we discuss the state-of-the-art tools and investigative approaches being used to characterize the biology and function of OPCs. From functional genetic investigation to single cell sequencing and from lineage tracing to functional imaging, we discuss the important discoveries uncovered by these techniques, such as functional and spatial OPC heterogeneity, novel OPC marker genes, the interaction of OPCs with other cells types, and how OPCs integrate and respond to signals from neighboring cells. Finally, we review the use of in vitro assay to assess OPC functions. These methodologies promise to lead to ever greater understanding of this enigmatic cell type, which in turn will shed light on the pathogenesis and potential treatment strategies for a number of diseases, such as multiple sclerosis (MS) and gliomas.
Collapse
Affiliation(s)
- Christophe Galichet
- Laboratory of Stem Cell Biology and Developmental Genetics, The Francis Crick Institute, London, United Kingdom
| | | | | |
Collapse
|
21
|
Manousi A, Göttle P, Reiche L, Cui QL, Healy LM, Akkermann R, Gruchot J, Schira-Heinen J, Antel JP, Hartung HP, Küry P. Identification of novel myelin repair drugs by modulation of oligodendroglial differentiation competence. EBioMedicine 2021; 65:103276. [PMID: 33714029 PMCID: PMC7970057 DOI: 10.1016/j.ebiom.2021.103276] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 02/22/2021] [Accepted: 02/23/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND In multiple sclerosis loss of myelin and oligodendrocytes impairs saltatory signal transduction and leads to neuronal loss and functional deficits. Limited capacity of oligodendroglial precursor cells to differentiate into mature cells is the main reason for inefficient myelin repair in the central nervous system. Drug repurposing constitutes a powerful approach for identification of pharmacological compounds promoting this process. METHODS A phenotypic compound screening using the subcellular distribution of a potent inhibitor of oligodendroglial cell differentiation, namely p57kip2, as differentiation competence marker was conducted. Hit compounds were validated in terms of their impact on developmental cell differentiation and myelination using both rat and human primary cell cultures and organotypic cerebellar slice cultures, respectively. Their effect on spontaneous remyelination was then investigated following cuprizone-mediated demyelination of the corpus callosum. FINDINGS A number of novel small molecules able to promote oligodendroglial cell differentiation were identified and a subset was found to foster human oligodendrogenesis as well as myelination ex vivo. Among them the steroid danazol and the anthelminthic parbendazole were found to increase myelin repair. INTERPRETATION We provide evidence that early cellular processes involved in differentiation decisions are applicable for the identification of regeneration promoting drugs and we suggest danazol and parbendazole as potent therapeutic candidates for demyelinating diseases. FUNDING This work was supported by the Jürgen Manchot Foundation, Düsseldorf; Research Commission of the Medical Faculty of Heinrich-Heine-University Düsseldorf; Christiane and Claudia Hempel Foundation; Stifterverband/Novartisstiftung; James and Elisabeth Cloppenburg, Peek and Cloppenburg Düsseldorf Stiftung and International Progressive MS Alliance (BRAVEinMS).
Collapse
Affiliation(s)
- Anastasia Manousi
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, 40225, Germany
| | - Peter Göttle
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, 40225, Germany
| | - Laura Reiche
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, 40225, Germany
| | - Qiao-Ling Cui
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC H4A 3K9, Canada
| | - Luke M Healy
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC H4A 3K9, Canada
| | - Rainer Akkermann
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, 40225, Germany
| | - Joel Gruchot
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, 40225, Germany
| | - Jessica Schira-Heinen
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, 40225, Germany
| | - Jack P Antel
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC H4A 3K9, Canada
| | - Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, 40225, Germany; Brain and Mind Centre, University of Sydney, Camperdown NSW 2050, Australia
| | - Patrick Küry
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, 40225, Germany.
| |
Collapse
|
22
|
Akay LA, Effenberger AH, Tsai LH. Cell of all trades: oligodendrocyte precursor cells in synaptic, vascular, and immune function. Genes Dev 2021; 35:180-198. [PMID: 33526585 PMCID: PMC7849363 DOI: 10.1101/gad.344218.120] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Oligodendrocyte precursor cells (OPCs) are not merely a transitory progenitor cell type, but rather a distinct and heterogeneous population of glia with various functions in the developing and adult central nervous system. In this review, we discuss the fate and function of OPCs in the brain beyond their contribution to myelination. OPCs are electrically sensitive, form synapses with neurons, support blood-brain barrier integrity, and mediate neuroinflammation. We explore how sex and age may influence OPC activity, and we review how OPC dysfunction may play a primary role in numerous neurological and neuropsychiatric diseases. Finally, we highlight areas of future research.
Collapse
Affiliation(s)
- Leyla Anne Akay
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Audrey H Effenberger
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Li-Huei Tsai
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| |
Collapse
|
23
|
Ordaz RP, Garay E, Limon A, Pérez-Samartín A, Sánchez-Gómez MV, Robles-Martínez L, Cisneros-Mejorado A, Matute C, Arellano RO. GABA A Receptors Expressed in Oligodendrocytes Cultured from the Neonatal Rat Contain α3 and γ1 Subunits and Present Differential Functional and Pharmacological Properties. Mol Pharmacol 2020; 99:133-146. [PMID: 33288547 DOI: 10.1124/molpharm.120.000091] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 11/05/2020] [Indexed: 12/25/2022] Open
Abstract
Oligodendrocytes (OLs) express functional GABAA receptors (GABAARs) that are activated by GABA released at synaptic contacts with axons or by ambient GABA in extrasynaptic domains. In both instances, the receptors' molecular identity has not been fully defined. Furthermore, data on their structural diversity in different brain regions and information on age-dependent changes in their molecular composition are scant. This lack of knowledge has delayed access to a better understanding of the role of GABAergic signaling between neurons and OLs. Here, we used functional, and pharmacological analyses, as well as gene and protein expression of GABAAR subunits, to explore the subunit combination that could explain the receptor functional profile expressed in OLs from the neonate rat. We found that GABAAR composed of α3β2γ1 subunits mimicked the characteristics of the endogenous receptor when expressed heterologously in Xenopus laevis oocytes. Either α3 or γ1 subunit silencing by small interfering RNA transfection changed the GABA-response characteristics in oligodendrocyte precursor cells, indicating their participation in the endogenous receptor conformation. Thus, α3 subunit silencing shifted the mean EC50 for GABA from 75.1 to 46.6 µM, whereas γ1 silencing reduced the current amplitude response by 55%. We also observed that β-carbolines differentially enhance GABA responses in oligodendroglia as compared with those in neurons. These results contribute to defining the molecular and pharmacological properties of GABAARs in OLs. Additionally, the identification of β-carbolines as selective enhancers of GABAARs in OLs may help to study the role of GABAergic signaling during myelination. SIGNIFICANCE STATEMENT: GABAergic signaling through GABAA receptors (GABAARs) expressed in the oligodendroglial lineage contributes to the myelination control. Determining the molecular identity and the pharmacology of these receptors is essential to define their specific roles in myelination. Using GABAAR subunit expression and silencing, we identified that the GABAAR subunit combination α3β2γ1 conforms the bulk of GABAARs in oligodendrocytes from rat neonates. Furthermore, we found that these receptors have differential pharmacological properties that allow specific positive modulation by β-carbolines.
Collapse
Affiliation(s)
- Rainald Pablo Ordaz
- Instituto de Neurobiología, Laboratorio de Neurofisiología Celular, Universidad Nacional Autónoma de México, Juriquilla, Querétaro, México (R.P.O., E.G., L.R.-M., A.C.-M., R.O.A.); Mitchell Center for Neurodegenerative Diseases, Department of Neurology, School of Medicine, University of Texas Medical Branch at Galveston, Galveston, Texas (A.L.); and Achucarro Basque Center for Neuroscience, CIBERNED and Departamento de Neurociencias, Universidad del País Vasco (UPV/EHU), Leioa, Spain (A.P.-S., M.V.S.-G., C.M.)
| | - Edith Garay
- Instituto de Neurobiología, Laboratorio de Neurofisiología Celular, Universidad Nacional Autónoma de México, Juriquilla, Querétaro, México (R.P.O., E.G., L.R.-M., A.C.-M., R.O.A.); Mitchell Center for Neurodegenerative Diseases, Department of Neurology, School of Medicine, University of Texas Medical Branch at Galveston, Galveston, Texas (A.L.); and Achucarro Basque Center for Neuroscience, CIBERNED and Departamento de Neurociencias, Universidad del País Vasco (UPV/EHU), Leioa, Spain (A.P.-S., M.V.S.-G., C.M.)
| | - Agenor Limon
- Instituto de Neurobiología, Laboratorio de Neurofisiología Celular, Universidad Nacional Autónoma de México, Juriquilla, Querétaro, México (R.P.O., E.G., L.R.-M., A.C.-M., R.O.A.); Mitchell Center for Neurodegenerative Diseases, Department of Neurology, School of Medicine, University of Texas Medical Branch at Galveston, Galveston, Texas (A.L.); and Achucarro Basque Center for Neuroscience, CIBERNED and Departamento de Neurociencias, Universidad del País Vasco (UPV/EHU), Leioa, Spain (A.P.-S., M.V.S.-G., C.M.)
| | - Alberto Pérez-Samartín
- Instituto de Neurobiología, Laboratorio de Neurofisiología Celular, Universidad Nacional Autónoma de México, Juriquilla, Querétaro, México (R.P.O., E.G., L.R.-M., A.C.-M., R.O.A.); Mitchell Center for Neurodegenerative Diseases, Department of Neurology, School of Medicine, University of Texas Medical Branch at Galveston, Galveston, Texas (A.L.); and Achucarro Basque Center for Neuroscience, CIBERNED and Departamento de Neurociencias, Universidad del País Vasco (UPV/EHU), Leioa, Spain (A.P.-S., M.V.S.-G., C.M.)
| | - María Victoria Sánchez-Gómez
- Instituto de Neurobiología, Laboratorio de Neurofisiología Celular, Universidad Nacional Autónoma de México, Juriquilla, Querétaro, México (R.P.O., E.G., L.R.-M., A.C.-M., R.O.A.); Mitchell Center for Neurodegenerative Diseases, Department of Neurology, School of Medicine, University of Texas Medical Branch at Galveston, Galveston, Texas (A.L.); and Achucarro Basque Center for Neuroscience, CIBERNED and Departamento de Neurociencias, Universidad del País Vasco (UPV/EHU), Leioa, Spain (A.P.-S., M.V.S.-G., C.M.)
| | - Leticia Robles-Martínez
- Instituto de Neurobiología, Laboratorio de Neurofisiología Celular, Universidad Nacional Autónoma de México, Juriquilla, Querétaro, México (R.P.O., E.G., L.R.-M., A.C.-M., R.O.A.); Mitchell Center for Neurodegenerative Diseases, Department of Neurology, School of Medicine, University of Texas Medical Branch at Galveston, Galveston, Texas (A.L.); and Achucarro Basque Center for Neuroscience, CIBERNED and Departamento de Neurociencias, Universidad del País Vasco (UPV/EHU), Leioa, Spain (A.P.-S., M.V.S.-G., C.M.)
| | - Abraham Cisneros-Mejorado
- Instituto de Neurobiología, Laboratorio de Neurofisiología Celular, Universidad Nacional Autónoma de México, Juriquilla, Querétaro, México (R.P.O., E.G., L.R.-M., A.C.-M., R.O.A.); Mitchell Center for Neurodegenerative Diseases, Department of Neurology, School of Medicine, University of Texas Medical Branch at Galveston, Galveston, Texas (A.L.); and Achucarro Basque Center for Neuroscience, CIBERNED and Departamento de Neurociencias, Universidad del País Vasco (UPV/EHU), Leioa, Spain (A.P.-S., M.V.S.-G., C.M.)
| | - Carlos Matute
- Instituto de Neurobiología, Laboratorio de Neurofisiología Celular, Universidad Nacional Autónoma de México, Juriquilla, Querétaro, México (R.P.O., E.G., L.R.-M., A.C.-M., R.O.A.); Mitchell Center for Neurodegenerative Diseases, Department of Neurology, School of Medicine, University of Texas Medical Branch at Galveston, Galveston, Texas (A.L.); and Achucarro Basque Center for Neuroscience, CIBERNED and Departamento de Neurociencias, Universidad del País Vasco (UPV/EHU), Leioa, Spain (A.P.-S., M.V.S.-G., C.M.)
| | - Rogelio O Arellano
- Instituto de Neurobiología, Laboratorio de Neurofisiología Celular, Universidad Nacional Autónoma de México, Juriquilla, Querétaro, México (R.P.O., E.G., L.R.-M., A.C.-M., R.O.A.); Mitchell Center for Neurodegenerative Diseases, Department of Neurology, School of Medicine, University of Texas Medical Branch at Galveston, Galveston, Texas (A.L.); and Achucarro Basque Center for Neuroscience, CIBERNED and Departamento de Neurociencias, Universidad del País Vasco (UPV/EHU), Leioa, Spain (A.P.-S., M.V.S.-G., C.M.)
| |
Collapse
|
24
|
Neuronal activity-dependent myelin repair promotes motor function recovery after contusion spinal cord injury. Brain Res Bull 2020; 166:73-81. [PMID: 33197536 DOI: 10.1016/j.brainresbull.2020.11.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 10/09/2020] [Accepted: 11/11/2020] [Indexed: 12/12/2022]
Abstract
An increasing number of studies connect neuronal activity with developmental myelination but how neuronal activity regulates remyelination has not been clarified. In this study, we induced the demyelination of the dorsal corticospinal tract (dCST) by a mild contusion spinal cord injury (SCI) on the T10 segment, and manipulated the neuronal activity of the primary motor cortex (M1) using chemogenetic viruses to induce activity and to suppress it. We found that oligodendrocyte precursor cell (OPC) proliferation and oligodendrocyte maturity following remyelination was strengthened after 4-week of neuronal activity stimulation. Furthermore, hindlimb motor function was also found to be improved. Vice versa, suppression of neuronal activity attenuated these effects. These results indicate that bidirectional regulation of neuronal activity can effectively modulate the development of oligodendrocyte lineage cells and the remyelination process. Neuronal activity supports the proliferation of OPCs, improves oligodendrocyte maturation and amplifies the axonal remyelination process, even though leads to better motor function recovery. Manipulation of neuronal activity in a non-invasive manner is therefore a promising avenue for exploration towards the treatment of central nervous system (CNS) demyelination diseases.
Collapse
|
25
|
Auderset L, Pitman KA, Cullen CL, Pepper RE, Taylor BV, Foa L, Young KM. Low-Density Lipoprotein Receptor-Related Protein 1 (LRP1) Is a Negative Regulator of Oligodendrocyte Progenitor Cell Differentiation in the Adult Mouse Brain. Front Cell Dev Biol 2020; 8:564351. [PMID: 33282858 PMCID: PMC7691426 DOI: 10.3389/fcell.2020.564351] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 09/14/2020] [Indexed: 12/17/2022] Open
Abstract
Low-density lipoprotein receptor-related protein 1 (LRP1) is a large, endocytic cell surface receptor that is highly expressed by oligodendrocyte progenitor cells (OPCs) and LRP1 expression is rapidly downregulated as OPCs differentiate into oligodendrocytes (OLs). We report that the conditional deletion of Lrp1 from adult mouse OPCs (Pdgfrα-CreER :: Lrp1fl/fl) increases the number of newborn, mature myelinating OLs added to the corpus callosum and motor cortex. As these additional OLs extend a normal number of internodes that are of a normal length, Lrp1-deletion increases adult myelination. OPC proliferation is also elevated following Lrp1 deletion in vivo, however, this may be a secondary, homeostatic response to increased OPC differentiation, as our in vitro experiments show that LRP1 is a direct negative regulator of OPC differentiation, not proliferation. Deleting Lrp1 from adult OPCs also increases the number of newborn mature OLs added to the corpus callosum in response to cuprizone-induced demyelination. These data suggest that the selective blockade of LRP1 function on adult OPCs may enhance myelin repair in demyelinating diseases such as multiple sclerosis.
Collapse
Affiliation(s)
- Loic Auderset
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Kimberley A Pitman
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Carlie L Cullen
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Renee E Pepper
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Bruce V Taylor
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Lisa Foa
- School of Medicine, University of Tasmania, Hobart, TAS, Australia
| | - Kaylene M Young
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| |
Collapse
|
26
|
Lima-Filho CM, Nogaroli L, Hedin-Pereira C, Azevedo SMFO, Soares RM. Effects of saxitoxins exposure on oligodendrocyte development in mouse neonates. Toxicon 2020; 188:89-94. [PMID: 33069750 DOI: 10.1016/j.toxicon.2020.10.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 09/09/2020] [Accepted: 10/14/2020] [Indexed: 11/28/2022]
Abstract
Saxitoxins (STXs) are neurotoxins produced by cyanobacteria and dinoflagellates, and they are primarily known to block voltage-gated sodium channels in neurons. The present study aimed to obtain further information regarding the effects of these toxins on neurodevelopment by investigating the responses of murine subventricular zone (SVZ) neural progenitors to STXs. An in vitro neonatal mouse SVZ explant model was exposed to different concentrations of toxic cyanobacterial extracts to evaluate the migration and differentiation of SVZ-derived progenitor cells. To test the ability of STX to cross the placental barrier, pregnant mice received a single intraperitoneal injection of STXs (7.5 μg/kg body weight) on gestational day fifteen. Immunocytochemistry was performed to detect proliferating and differentiating progenitors, including oligodendrocyte progenitor cells (OPCs). It was found that specific proliferation of OPCs was significantly increased, but there was no corresponding increase in the number of differentiated oligodendrocytes, which may indicate a negative effect on the maturation process of these cells. Additionally, the data showed that STXs crossed the placental barrier. Thus, STXs can be considered a potential risk to fetal neurodevelopment.
Collapse
Affiliation(s)
- Cesar Macedo Lima-Filho
- Laboratory of Ecophysiology and Toxicology of Cyanobacteria, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho, 373, CCS-Bloco G, Ilha do Fundão, CEP: 21941-902, Rio de Janeiro, Brazil
| | - Luciana Nogaroli
- Laboratory of Cellular Neuroanatomy, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho, 373, CCS-Bloco G, Ilha do Fundão, CEP: 21941-902, Rio de Janeiro, Brazil
| | - Cecilia Hedin-Pereira
- Laboratory of Cellular Neuroanatomy, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho, 373, CCS-Bloco G, Ilha do Fundão, CEP: 21941-902, Rio de Janeiro, Brazil
| | - Sandra M F O Azevedo
- Laboratory of Ecophysiology and Toxicology of Cyanobacteria, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho, 373, CCS-Bloco G, Ilha do Fundão, CEP: 21941-902, Rio de Janeiro, Brazil
| | - Raquel M Soares
- Multidisciplinary Center of Research in Biology, NUMPEX-BIO, Campus Duque de Caxias, Universidade Federal do Rio de Janeiro, Rodovia Washington Luiz, km 105, CEP: 25240-005, Duque de Caxias, RJ, Brazil.
| |
Collapse
|
27
|
Impaired Postnatal Myelination in a Conditional Knockout Mouse for the Ferritin Heavy Chain in Oligodendroglial Cells. J Neurosci 2020; 40:7609-7624. [PMID: 32868463 DOI: 10.1523/jneurosci.1281-20.2020] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 08/14/2020] [Accepted: 08/19/2020] [Indexed: 01/22/2023] Open
Abstract
To define the importance of iron storage in oligodendrocyte development and function, the ferritin heavy subunit (Fth) was specifically deleted in oligodendroglial cells. Blocking Fth synthesis in Sox10 or NG2-positive oligodendrocytes during the first or the third postnatal week significantly reduces oligodendrocyte iron storage and maturation. The brain of Fth KO animals presented an important decrease in the expression of myelin proteins and a substantial reduction in the percentage of myelinated axons. This hypomyelination was accompanied by a decline in the number of myelinating oligodendrocytes and with a reduction in proliferating oligodendrocyte progenitor cells (OPCs). Importantly, deleting Fth in Sox10-positive oligodendroglial cells after postnatal day 60 has no effect on myelin production and/or oligodendrocyte quantities. We also tested the capacity of Fth-deficient OPCs to remyelinate the adult brain in the cuprizone model of myelin injury and repair. Fth deletion in NG2-positive OPCs significantly reduces the number of mature oligodendrocytes and myelin production throughout the remyelination process. Furthermore, the corpus callosum of Fth KO animals presented a significant decrease in the percentage of remyelinated axons and a substantial reduction in the average myelin thickness. These results indicate that Fth synthesis during the first three postnatal weeks is important for an appropriate oligodendrocyte development, and suggest that Fth iron storage in adult OPCs is also essential for an effective remyelination of the mouse brain.SIGNIFICANCE STATEMENT To define the importance of iron storage in oligodendrocyte function, we have deleted the ferritin heavy chain (Fth) specifically in the oligodendrocyte lineage. Fth ablation in oligodendroglial cells throughout early postnatal development significantly reduces oligodendrocyte maturation and myelination. In contrast, deletion of Fth in oligodendroglial cells after postnatal day 60 has no effect on myelin production and/or oligodendrocyte numbers. We have also tested the consequences of disrupting Fth iron storage in oligodendrocyte progenitor cells (OPCs) after demyelination. We have found that Fth deletion in NG2-positive OPCs significantly delays the remyelination process in the adult brain. Therefore, Fth iron storage is essential for early oligodendrocyte development as well as for OPC maturation in the demyelinated adult brain.
Collapse
|
28
|
Spontaneous Local Calcium Transients Regulate Oligodendrocyte Development in Culture through Store-Operated Ca 2+ Entry and Release. eNeuro 2020; 7:ENEURO.0347-19.2020. [PMID: 32409508 PMCID: PMC7438061 DOI: 10.1523/eneuro.0347-19.2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 04/30/2020] [Accepted: 05/06/2020] [Indexed: 12/14/2022] Open
Abstract
Oligodendrocytes (OLs) insulate axonal fibers for fast conduction of nerve impulses by wrapping axons of the CNS with compact myelin membranes. Differentiating OLs undergo drastic chances in cell morphology. Bipolar oligodendroglial precursor cells (OPCs) transform into highly ramified multipolar OLs, which then expand myelin membranes that enwrap axons. While significant progress has been made in understanding the molecular and genetic mechanisms underlying CNS myelination and its disruption in diseases, the cellular mechanisms that regulate OL differentiation are not fully understood. Here, we report that developing rat OLs in culture exhibit spontaneous Ca2+ local transients (sCaLTs) in their process arbors in the absence of neurons. Importantly, we find that the frequency of sCaLTs markedly increases as OLs undergo extensive process outgrowth and branching. We further show that sCaLTs are primarily generated through a combination of Ca2+ influx through store-operated Ca2+ entry (SOCE) and Ca2+ release from internal Ca2+ stores. Inhibition of sCaLTs impairs the elaboration and branching of OL processes, as well as substantially reduces the formation of large myelin sheets in culture. Together, our findings identify an important role for spontaneous local Ca2+ signaling in OL development.
Collapse
|
29
|
Deletion of Voltage-Gated Calcium Channels in Astrocytes during Demyelination Reduces Brain Inflammation and Promotes Myelin Regeneration in Mice. J Neurosci 2020; 40:3332-3347. [PMID: 32169969 DOI: 10.1523/jneurosci.1644-19.2020] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 02/28/2020] [Accepted: 03/03/2020] [Indexed: 12/28/2022] Open
Abstract
To determine whether Cav1.2 voltage-gated Ca2+ channels contribute to astrocyte activation, we generated an inducible conditional knock-out mouse in which the Cav1.2 α subunit was deleted in GFAP-positive astrocytes. This astrocytic Cav1.2 knock-out mouse was tested in the cuprizone model of myelin injury and repair which causes astrocyte and microglia activation in the absence of a lymphocytic response. Deletion of Cav1.2 channels in GFAP-positive astrocytes during cuprizone-induced demyelination leads to a significant reduction in the degree of astrocyte and microglia activation and proliferation in mice of either sex. Concomitantly, the production of proinflammatory factors such as TNFα, IL1β and TGFβ1 was significantly decreased in the corpus callosum and cortex of Cav1.2 knock-out mice through demyelination. Furthermore, this mild inflammatory environment promotes oligodendrocyte progenitor cells maturation and myelin regeneration across the remyelination phase of the cuprizone model. Similar results were found in animals treated with nimodipine, a Cav1.2 Ca2+ channel inhibitor with high affinity to the CNS. Mice of either sex injected with nimodipine during the demyelination stage of the cuprizone treatment displayed a reduced number of reactive astrocytes and showed a faster and more efficient brain remyelination. Together, these results indicate that Cav1.2 Ca2+ channels play a crucial role in the induction and proliferation of reactive astrocytes during demyelination; and that attenuation of astrocytic voltage-gated Ca2+ influx may be an effective therapy to reduce brain inflammation and promote myelin recovery in demyelinating diseases.SIGNIFICANCE STATEMENT Reducing voltage-gated Ca2+ influx in astrocytes during brain demyelination significantly attenuates brain inflammation and astrocyte reactivity. Furthermore, these changes promote myelin restoration and oligodendrocyte maturation throughout remyelination.
Collapse
|
30
|
Strategies for Neuroprotection in Multiple Sclerosis and the Role of Calcium. Int J Mol Sci 2020; 21:ijms21051663. [PMID: 32121306 PMCID: PMC7084497 DOI: 10.3390/ijms21051663] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 02/16/2020] [Accepted: 02/26/2020] [Indexed: 12/12/2022] Open
Abstract
Calcium ions are vital for maintaining the physiological and biochemical processes inside cells. The central nervous system (CNS) is particularly dependent on calcium homeostasis and its dysregulation has been associated with several neurodegenerative disorders including Parkinson’s disease (PD), Alzheimer’s disease (AD) and Huntington’s disease (HD), as well as with multiple sclerosis (MS). Hence, the modulation of calcium influx into the cells and the targeting of calcium-mediated signaling pathways may present a promising therapeutic approach for these diseases. This review provides an overview on calcium channels in neurons and glial cells. Special emphasis is put on MS, a chronic autoimmune disease of the CNS. While the initial relapsing-remitting stage of MS can be treated effectively with immune modulatory and immunosuppressive drugs, the subsequent progressive stage has remained largely untreatable. Here we summarize several approaches that have been and are currently being tested for their neuroprotective capacities in MS and we discuss which role calcium could play in this regard.
Collapse
|
31
|
Abstract
Cells of the oligodendrocyte lineage express a wide range of Ca2+ channels and receptors that regulate oligodendrocyte progenitor cell (OPC) and oligodendrocyte formation and function. Here we define those key channels and receptors that regulate Ca2+ signaling and OPC development and myelination. We then discuss how the regulation of intracellular Ca2+ in turn affects OPC and oligodendrocyte biology in the healthy nervous system and under pathological conditions. Activation of Ca2+ channels and receptors in OPCs and oligodendrocytes by neurotransmitters converges on regulating intracellular Ca2+, making Ca2+ signaling a central candidate mediator of activity-driven myelination. Indeed, recent evidence indicates that localized changes in Ca2+ in oligodendrocytes can regulate the formation and remodeling of myelin sheaths and perhaps additional functions of oligodendrocytes and OPCs. Thus, decoding how OPCs and myelinating oligodendrocytes integrate and process Ca2+ signals will be important to fully understand central nervous system formation, health, and function.
Collapse
Affiliation(s)
- Pablo M Paez
- Department of Pharmacology and Toxicology and Hunter James Kelly Research Institute, Jacobs School of Medicine and Biomedical Sciences, The State University of New York, University at Buffalo, Buffalo, New York 14203, USA;
| | - David A Lyons
- Centre for Discovery Brain Sciences, Centre for Multiple Sclerosis Research, and Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh EH16 4SB, United Kingdom;
| |
Collapse
|
32
|
Spencer SA, Suárez-Pozos E, Escalante M, Myo YP, Fuss B. Sodium-Calcium Exchangers of the SLC8 Family in Oligodendrocytes: Functional Properties in Health and Disease. Neurochem Res 2020; 45:1287-1297. [PMID: 31927687 DOI: 10.1007/s11064-019-02949-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 12/20/2019] [Accepted: 12/23/2019] [Indexed: 12/30/2022]
Abstract
The solute carrier 8 (SLC8) family of sodium-calcium exchangers (NCXs) functions as an essential regulatory system that couples opposite fluxes of sodium and calcium ions across plasmalemmal membranes. NCXs, thereby, play key roles in maintaining an ion homeostasis that preserves cellular integrity. Hence, alterations in NCX expression and regulation have been found to lead to ionic imbalances that are often associated with intracellular calcium overload and cell death. On the other hand, intracellular calcium has been identified as a key driver for a multitude of downstream signaling events that are crucial for proper functioning of biological systems, thus highlighting the need for a tightly controlled balance. In the CNS, NCXs have been primarily characterized in the context of synaptic transmission and ischemic brain damage. However, a much broader picture is emerging. NCXs are expressed by virtually all cells of the CNS including oligodendrocytes (OLGs), the cells that generate the myelin sheath. With a growing appreciation of dynamic calcium signals in OLGs, NCXs are becoming increasingly recognized for their crucial roles in shaping OLG function under both physiological and pathophysiological conditions. In order to provide a current update, this review focuses on the importance of NCXs in cells of the OLG lineage. More specifically, it provides a brief introduction into plasmalemmal NCXs and their modes of activity, and it discusses the roles of OLG expressed NCXs in regulating CNS myelination and in contributing to CNS pathologies associated with detrimental effects on OLG lineage cells.
Collapse
Affiliation(s)
- Samantha A Spencer
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Box 980709, Richmond, VA, 23298, USA
| | - Edna Suárez-Pozos
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Box 980709, Richmond, VA, 23298, USA
| | - Miguel Escalante
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Box 980709, Richmond, VA, 23298, USA.,Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Yu Par Myo
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Box 980709, Richmond, VA, 23298, USA
| | - Babette Fuss
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Box 980709, Richmond, VA, 23298, USA.
| |
Collapse
|
33
|
Vitamin B Complex Treatment Attenuates Local Inflammation after Peripheral Nerve Injury. Molecules 2019; 24:molecules24244615. [PMID: 31861069 PMCID: PMC6943485 DOI: 10.3390/molecules24244615] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 12/02/2019] [Accepted: 12/13/2019] [Indexed: 01/20/2023] Open
Abstract
Peripheral nerve injury (PNI) leads to a series of cellular and molecular events necessary for axon regeneration and reinnervation of target tissues, among which inflammation is crucial for the orchestration of all these processes. Macrophage activation underlies the pathogenesis of PNI and is characterized by morphological/phenotype transformation from proinflammatory (M1) to an anti-inflammatory (M2) type with different functions in the inflammatory and reparative process. The aim of this study was to evaluate influence of the vitamin B (B1, B2, B3, B5, B6, and B12) complex on the process of neuroinflammation that is in part regulated by l-type CaV1.2 calcium channels. A controlled transection of the motor branch of the femoral peripheral nerve was used as an experimental model. Animals were sacrificed after 1, 3, 7, and 14 injections of vitamin B complex. Isolated nerves were used for immunofluorescence analysis. Treatment with vitamin B complex decreased expression of proinflammatory and increased expression of anti-inflammatory cytokines, thus contributing to the resolution of neuroinflammation. In parallel, B vitamins decreased the number of M1 macrophages that expressed the CaV1.2 channel, and increased the number of M2 macrophages that expressed this channel, suggesting their role in M1/M2 transition after PNI. In conclusion, B vitamins had the potential for treatment of neuroinflammation and neuroregeneration and thereby might be an effective therapy for PNI in humans.
Collapse
|
34
|
Zhang M, Liu Y, Wu S, Zhao X. Ca 2+ Signaling in Oligodendrocyte Development. Cell Mol Neurobiol 2019; 39:1071-1080. [PMID: 31222426 DOI: 10.1007/s10571-019-00705-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 06/11/2019] [Indexed: 12/31/2022]
Abstract
Calcium signaling has essential roles in the development of the nervous system, from neural induction to the proliferation, migration, and differentiation of both neuronal and glia cells. The temporal and spatial dynamics of Ca2+ signals control the highly diverse yet specific transcriptional programs that establish the complex structures of the nervous system. Ca2+-signaling pathways are shaped by interactions among metabotropic signaling cascades, ion channels, intracellular Ca2+ stores, and a multitude of downstream effector proteins that activate specific genetic programs. Progress in the last decade has led to significant advances in our understanding of the functional architecture of Ca2+ signaling networks involved in oligodendrocyte development. In this review, we summarize the molecular and functional organizations of Ca2+-signaling networks during the differentiation of oligodendrocyte, especially its impact on myelin gene expression, proliferation, migration, and myelination. Importantly, the existence of multiple routes of Ca2+ influx opens the possibility that the activity of calcium channels can be manipulated pharmacologically to encourage oligodendrocyte maturation and remyelination after demyelinating episodes in the brain.
Collapse
Affiliation(s)
- Ming Zhang
- Department of Neurobiology, Collaborative Innovation Center for Brain Science and Shaanxi Key Laboratory of Brain Disorders, Fourth Military Medical University, Xi'an, 710032, China
| | - Yuming Liu
- Department of Neurobiology, Collaborative Innovation Center for Brain Science and Shaanxi Key Laboratory of Brain Disorders, Fourth Military Medical University, Xi'an, 710032, China
| | - Shengxi Wu
- Department of Neurobiology, Collaborative Innovation Center for Brain Science and Shaanxi Key Laboratory of Brain Disorders, Fourth Military Medical University, Xi'an, 710032, China.
| | - Xianghui Zhao
- Department of Neurobiology, Collaborative Innovation Center for Brain Science and Shaanxi Key Laboratory of Brain Disorders, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
35
|
Iron Metabolism in the Peripheral Nervous System: The Role of DMT1, Ferritin, and Transferrin Receptor in Schwann Cell Maturation and Myelination. J Neurosci 2019; 39:9940-9953. [PMID: 31676601 DOI: 10.1523/jneurosci.1409-19.2019] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 10/09/2019] [Accepted: 10/29/2019] [Indexed: 12/24/2022] Open
Abstract
Iron is an essential cofactor for many cellular enzymes involved in myelin synthesis, and iron homeostasis unbalance is a central component of peripheral neuropathies. However, iron absorption and management in the PNS are poorly understood. To study iron metabolism in Schwann cells (SCs), we have created 3 inducible conditional KO mice in which three essential proteins implicated in iron uptake and storage, the divalent metal transporter 1 (DMT1), the ferritin heavy chain (Fth), and the transferrin receptor 1 (Tfr1), were postnatally ablated specifically in SCs. Deleting DMT1, Fth, or Tfr1 in vitro significantly reduce SC proliferation, maturation, and the myelination of DRG axons. This was accompanied by an important reduction in iron incorporation and storage. When these proteins were KO in vivo during the first postnatal week, the sciatic nerve of all 3 conditional KO animals displayed a significant reduction in the synthesis of myelin proteins and in the percentage of myelinated axons. Knocking out Fth produced the most severe phenotype, followed by DMT1 and, last, Tfr1. Importantly, DMT1 as well as Fth KO mice showed substantial motor coordination deficits. In contrast, deleting these proteins in mature myelinating SCs results in milder phenotypes characterized by small reductions in the percentage of myelinated axons and minor changes in the g-ratio of myelinated axons. These results indicate that DMT1, Fth, and Tfr1 are critical proteins for early postnatal iron uptake and storage in SCs and, as a consequence, for the normal myelination of the PNS.SIGNIFICANCE STATEMENT To determine the function of the divalent metal transporter 1, the transferrin receptor 1, and the ferritin heavy chain in Schwann cell (SC) maturation and myelination, we created 3 conditional KO mice in which these proteins were postnatally deleted in Sox10-positive SCs. We have established that these proteins are necessary for normal SC iron incorporation and storage, and, as a consequence, for an effective myelination of the PNS. Since iron is indispensable for SC maturation, understanding iron metabolism in SCs is an essential prerequisite for developing therapies for demyelinating diseases in the PNS.
Collapse
|
36
|
Calcium Signaling in Neurons and Glial Cells: Role of Cav1 channels. Neuroscience 2019; 421:95-111. [DOI: 10.1016/j.neuroscience.2019.09.041] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 09/27/2019] [Accepted: 09/30/2019] [Indexed: 11/18/2022]
|
37
|
Pitman KA, Ricci R, Gasperini R, Beasley S, Pavez M, Charlesworth J, Foa L, Young KM. The voltage-gated calcium channel CaV1.2 promotes adult oligodendrocyte progenitor cell survival in the mouse corpus callosum but not motor cortex. Glia 2019; 68:376-392. [PMID: 31605513 PMCID: PMC6916379 DOI: 10.1002/glia.23723] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 09/02/2019] [Accepted: 09/10/2019] [Indexed: 12/21/2022]
Abstract
Throughout life, oligodendrocyte progenitor cells (OPCs) proliferate and differentiate into myelinating oligodendrocytes. OPCs express cell surface receptors and channels that allow them to detect and respond to neuronal activity, including voltage‐gated calcium channel (VGCC)s. The major L‐type VGCC expressed by developmental OPCs, CaV1.2, regulates their differentiation. However, it is unclear whether CaV1.2 similarly influences OPC behavior in the healthy adult central nervous system (CNS). To examine the role of CaV1.2 in adulthood, we conditionally deleted this channel from OPCs by administering tamoxifen to P60 Cacna1cfl/fl (control) and Pdgfrα‐CreER:: Cacna1cfl/fl (CaV1.2‐deleted) mice. Whole cell patch clamp analysis revealed that CaV1.2 deletion reduced L‐type voltage‐gated calcium entry into adult OPCs by ~60%, confirming that it remains the major L‐type VGCC expressed by OPCs in adulthood. The conditional deletion of CaV1.2 from adult OPCs significantly increased their proliferation but did not affect the number of new oligodendrocytes produced or influence the length or number of internodes they elaborated. Unexpectedly, CaV1.2 deletion resulted in the dramatic loss of OPCs from the corpus callosum, such that 7 days after tamoxifen administration CaV1.2‐deleted mice had an OPC density ~42% that of control mice. OPC density recovered within 2 weeks of CaV1.2 deletion, as the lost OPCs were replaced by surviving CaV1.2‐deleted OPCs. As OPC density was not affected in the motor cortex or spinal cord, we conclude that calcium entry through CaV1.2 is a critical survival signal for a subpopulation of callosal OPCs but not for all OPCs in the mature CNS.
Collapse
Affiliation(s)
- Kimberley A Pitman
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Raphael Ricci
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Robert Gasperini
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia.,School of Medicine, University of Tasmania, Hobart, Australia
| | - Shannon Beasley
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Macarena Pavez
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Jac Charlesworth
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Lisa Foa
- School of Medicine, University of Tasmania, Hobart, Australia
| | - Kaylene M Young
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| |
Collapse
|
38
|
Liu XB, Haney JR, Cantero G, Lambert JR, Otero-Garcia M, Truong B, Gropman A, Cobos I, Cederbaum SD, Lipshutz GS. Hepatic arginase deficiency fosters dysmyelination during postnatal CNS development. JCI Insight 2019; 4:130260. [PMID: 31484827 DOI: 10.1172/jci.insight.130260] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 08/01/2019] [Indexed: 12/26/2022] Open
Abstract
Deficiency of arginase is associated with hyperargininemia, and prominent features include spastic diplegia/tetraplegia, clonus, and hyperreflexia; loss of ambulation, intellectual disability and progressive neurological decline are other signs. To gain greater insight into the unique neuromotor features, we performed gene expression profiling of the motor cortex of a murine model of the disorder. Coexpression network analysis suggested an abnormality with myelination, which was supported by limited existing human data. Utilizing electron microscopy, marked dysmyelination was detected in 2-week-old homozygous Arg1-KO mice. The corticospinal tract was found to be adversely affected, supporting dysmyelination as the cause of the unique neuromotor features and implicating oligodendrocyte impairment in a deficiency of hepatic Arg1. Following neonatal hepatic gene therapy to express Arg1, the subcortical white matter, pyramidal tract, and corticospinal tract all showed a remarkable recovery in terms of myelinated axon density and ultrastructural integrity with active wrapping of axons by nearby oligodendrocyte processes. These findings support the following conclusions: arginase deficiency is a leukodystrophy affecting the brain and spinal cord while sparing the peripheral nervous system, and neonatal AAV hepatic gene therapy can rescue the defects associated with myelinated axons, strongly implicating the functional recovery of oligodendrocytes after restoration of hepatic arginase activity.
Collapse
Affiliation(s)
| | - Jillian R Haney
- Department of Psychiatry.,Intellectual and Developmental Disabilities Research Center, and.,Semel Institute for Neuroscience, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Gloria Cantero
- Neuromuscular Disorders Unit, Department of Neurology, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | | | | | - Brian Truong
- Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Andrea Gropman
- Neurogenetics and Neurodevelopmental Pediatrics and Genetics, Children's National Health System, Washington, DC, USA
| | - Inma Cobos
- Department of Pathology and Laboratory Medicine and
| | - Stephen D Cederbaum
- Department of Psychiatry.,Intellectual and Developmental Disabilities Research Center, and.,Semel Institute for Neuroscience, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Gerald S Lipshutz
- Department of Surgery.,Department of Psychiatry.,Intellectual and Developmental Disabilities Research Center, and.,Semel Institute for Neuroscience, David Geffen School of Medicine at UCLA, Los Angeles, California.,Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA.,Molecular Biology Institute, UCLA, Los Angeles, California, USA
| |
Collapse
|
39
|
Barron T, Kim JH. Neuronal input triggers Ca 2+ influx through AMPA receptors and voltage-gated Ca 2+ channels in oligodendrocytes. Glia 2019; 67:1922-1932. [PMID: 31313856 PMCID: PMC6771819 DOI: 10.1002/glia.23670] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 06/13/2019] [Accepted: 06/18/2019] [Indexed: 12/25/2022]
Abstract
Communication between neurons and developing oligodendrocytes (OLs) leading to OL Ca2+ rise is critical for axon myelination and OL development. Here, we investigate signaling factors and sources of Ca2+ rise in OLs in the mouse brainstem. Glutamate puff or axon fiber stimulation induces a Ca2+ rise in pre‐myelinating OLs, which is primarily mediated by Ca2+‐permeable AMPA receptors. During glutamate application, inward currents via AMPA receptors and elevated extracellular K+ caused by increased neuronal activity collectively lead to OL depolarization, triggering Ca2+ influx via P/Q‐ and L‐type voltage‐gated Ca2+ (Cav) channels. Thus, glutamate is a key signaling factor in dynamic communication between neurons and OLs that triggers Ca2+ transients via AMPARs and Cav channels in developing OLs. The results provide a mechanism for OL Ca2+ dynamics in response to neuronal input, which has implications for OL development and myelination.
Collapse
Affiliation(s)
- Tara Barron
- Department of Cellular and Integrative Physiology, UT Health San Antonio, San Antonio, Texas
| | - Jun Hee Kim
- Department of Cellular and Integrative Physiology, UT Health San Antonio, San Antonio, Texas
| |
Collapse
|
40
|
Crazy Little Thing Called Sox-New Insights in Oligodendroglial Sox Protein Function. Int J Mol Sci 2019; 20:ijms20112713. [PMID: 31159496 PMCID: PMC6600536 DOI: 10.3390/ijms20112713] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 05/29/2019] [Accepted: 05/30/2019] [Indexed: 02/06/2023] Open
Abstract
In the central nervous system, oligodendrocytes wrap axons with myelin sheaths, which is essential for rapid transfer of electric signals and their trophic support. In oligodendroglia, transcription factors of the Sox protein family are pivotal regulators of a variety of developmental processes. These include specification, proliferation, and migration of oligodendrocyte precursor cells as well as terminal differentiation to mature myelinating oligodendrocytes. Sox proteins are further affected in demyelinating diseases and are involved in remyelination following damage of the central nervous system. Here we summarize and discuss latest findings on transcriptional regulation of Sox proteins, their function, target genes, and interaction with other transcription factors and chromatin remodelers in oligodendroglia with physiological and pathophysiological relevance.
Collapse
|
41
|
Albrecht S, Korr S, Nowack L, Narayanan V, Starost L, Stortz F, Araúzo‐Bravo MJ, Meuth SG, Kuhlmann T, Hundehege P. The K
2P
‐channel TASK1 affects Oligodendroglial differentiation but not myelin restoration. Glia 2019; 67:870-883. [DOI: 10.1002/glia.23577] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 11/23/2018] [Accepted: 11/26/2018] [Indexed: 12/14/2022]
Affiliation(s)
- Stefanie Albrecht
- Institute of NeuropathologyUniversity Hospital Münster Münster Germany
| | - Sabrina Korr
- Institute of NeuropathologyUniversity Hospital Münster Münster Germany
- Department of Neurology with Institute of Translational NeurologyUniversity Hospital Münster Münster Germany
- Cells in Motion, Cluster of Excellence Münster Germany
| | - Luise Nowack
- Institute of NeuropathologyUniversity Hospital Münster Münster Germany
- Department of Neurology with Institute of Translational NeurologyUniversity Hospital Münster Münster Germany
| | - Venu Narayanan
- Department of Neurology with Institute of Translational NeurologyUniversity Hospital Münster Münster Germany
| | - Laura Starost
- Institute of NeuropathologyUniversity Hospital Münster Münster Germany
| | - Franziska Stortz
- Institute of NeuropathologyUniversity Hospital Münster Münster Germany
| | - Marcos J. Araúzo‐Bravo
- Group of Computational Biology and Systems Biomedicine, Biodonostia Health Research Institute San Sebastian Spain
- IKERBASQUE, Basque Foundation for Science Bilbao Spain
| | - Sven G. Meuth
- Department of Neurology with Institute of Translational NeurologyUniversity Hospital Münster Münster Germany
- Cells in Motion, Cluster of Excellence Münster Germany
| | - Tanja Kuhlmann
- Institute of NeuropathologyUniversity Hospital Münster Münster Germany
| | - Petra Hundehege
- Department of Neurology with Institute of Translational NeurologyUniversity Hospital Münster Münster Germany
- Cells in Motion, Cluster of Excellence Münster Germany
| |
Collapse
|
42
|
Cytoskeletal Regulation of Oligodendrocyte Differentiation and Myelination. J Neurosci 2018; 37:7797-7799. [PMID: 28821599 DOI: 10.1523/jneurosci.1398-17.2017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 07/09/2017] [Accepted: 07/11/2017] [Indexed: 11/21/2022] Open
|
43
|
Cheli VT, Santiago González DA, Zamora NN, Lama TN, Spreuer V, Rasmusson RL, Bett GC, Panagiotakos G, Paez PM. Enhanced oligodendrocyte maturation and myelination in a mouse model of Timothy syndrome. Glia 2018; 66:2324-2339. [PMID: 30151840 PMCID: PMC6697123 DOI: 10.1002/glia.23468] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 12/13/2017] [Accepted: 05/16/2018] [Indexed: 01/09/2023]
Abstract
To study the role of L-type voltage-gated Ca++ channels in oligodendrocyte development, we used a mouse model of Timothy syndrome (TS) in which a gain-of-function mutation in the α1 subunit of the L-type Ca++ channel Cav1.2 gives rise to an autism spectrum disorder (ASD). Oligodendrocyte progenitor cells (OPCs) isolated from the cortex of TS mice showed greater L-type Ca++ influx and displayed characteristics suggestive of advanced maturation compared to control OPCs, including a more complex morphology and higher levels of myelin protein expression. Consistent with this, expression of Cav1.2 channels bearing the TS mutation in wild-type OPCs triggered process formation and promoted oligodendrocyte-neuron interaction via the activation of Ca++ /calmodulin-dependent protein kinase II. To ascertain whether accelerated OPC maturation correlated with functional enhancements, we examined myelination in the TS brain at different postnatal time points. The expression of myelin proteins was significantly higher in the corpus callosum, cortex and striatum of TS animals, and immunohistochemical analysis for oligodendrocyte stage-specific markers revealed an increase in the density of myelinating oligodendrocytes in several areas of the TS brain. Along the same line, electron microscopy studies in the corpus callosum of TS animals showed significant increases both in the percentage of myelinated axons and in the thickness of myelin sheaths. In summary, these data indicate that OPC development and oligodendrocyte myelination is enhanced in the brain of TS mice, and suggest that this mouse model of a syndromic ASD is a useful tool to explore the role of L-type Ca++ channels in myelination.
Collapse
Affiliation(s)
- Veronica T. Cheli
- Hunter James Kelly Research Institute, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, The State University of New York, University at Buffalo, Buffalo, New York
| | - Diara A. Santiago González
- Hunter James Kelly Research Institute, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, The State University of New York, University at Buffalo, Buffalo, New York
| | - Norma N. Zamora
- Hunter James Kelly Research Institute, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, The State University of New York, University at Buffalo, Buffalo, New York
| | - Tenzing N. Lama
- Hunter James Kelly Research Institute, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, The State University of New York, University at Buffalo, Buffalo, New York
| | - Vilma Spreuer
- Hunter James Kelly Research Institute, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, The State University of New York, University at Buffalo, Buffalo, New York
| | - Randall L. Rasmusson
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, The State University of New York, University at Buffalo, Buffalo, New York
| | - Glenna C. Bett
- Department of Obstetrics and Gynecology, Jacobs School of Medicine and Biomedical Sciences, The State University of New York, University at Buffalo, Buffalo, New York
| | - Georgia Panagiotakos
- Department of Biochemistry and Biophysics and Kavli Institute for Fundamental Neuroscience, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, School of Medicine, University of California at San Francisco, San Francisco, California
| | - Pablo M. Paez
- Hunter James Kelly Research Institute, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, The State University of New York, University at Buffalo, Buffalo, New York
| |
Collapse
|
44
|
Cornillot M, Giacco V, Hamilton NB. The role of TRP channels in white matter function and ischaemia. Neurosci Lett 2018; 690:202-209. [PMID: 30366011 DOI: 10.1016/j.neulet.2018.10.042] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 10/09/2018] [Accepted: 10/18/2018] [Indexed: 01/15/2023]
Abstract
Transient receptor potential (TRP) proteins are a large family of tetrameric non-selective cation channels that are widely expressed in the grey and white matter of the CNS, and are increasingly considered as potential therapeutic targets in brain disorders. Here we briefly review the evidence for TRP channel expression in glial cells and their possible role in both glial cell physiology and stroke. Despite their contribution to important functions, our understanding of the roles of TRP channels in glia is still in its infancy. The evidence reviewed here indicates that further investigation is needed to determine whether TRP channel inhibition can decrease damage or increase repair in stroke and other diseases affecting the white matter.
Collapse
Affiliation(s)
- Marion Cornillot
- Wolfson Centre for Age Related Disease, King's College London, Guy's Campus, London, SE1 1UL, United Kingdom
| | - Vincenzo Giacco
- Wolfson Centre for Age Related Disease, King's College London, Guy's Campus, London, SE1 1UL, United Kingdom
| | - Nicola B Hamilton
- Wolfson Centre for Age Related Disease, King's College London, Guy's Campus, London, SE1 1UL, United Kingdom.
| |
Collapse
|
45
|
Schirmer L, Möbius W, Zhao C, Cruz-Herranz A, Ben Haim L, Cordano C, Shiow LR, Kelley KW, Sadowski B, Timmons G, Pröbstel AK, Wright JN, Sin JH, Devereux M, Morrison DE, Chang SM, Sabeur K, Green AJ, Nave KA, Franklin RJ, Rowitch DH. Oligodendrocyte-encoded Kir4.1 function is required for axonal integrity. eLife 2018; 7:36428. [PMID: 30204081 PMCID: PMC6167053 DOI: 10.7554/elife.36428] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 09/09/2018] [Indexed: 12/17/2022] Open
Abstract
Glial support is critical for normal axon function and can become dysregulated in white matter (WM) disease. In humans, loss-of-function mutations of KCNJ10, which encodes the inward-rectifying potassium channel KIR4.1, causes seizures and progressive neurological decline. We investigated Kir4.1 functions in oligodendrocytes (OLs) during development, adulthood and after WM injury. We observed that Kir4.1 channels localized to perinodal areas and the inner myelin tongue, suggesting roles in juxta-axonal K+ removal. Conditional knockout (cKO) of OL-Kcnj10 resulted in late onset mitochondrial damage and axonal degeneration. This was accompanied by neuronal loss and neuro-axonal dysfunction in adult OL-Kcnj10 cKO mice as shown by delayed visual evoked potentials, inner retinal thinning and progressive motor deficits. Axon pathologies in OL-Kcnj10 cKO were exacerbated after WM injury in the spinal cord. Our findings point towards a critical role of OL-Kir4.1 for long-term maintenance of axonal function and integrity during adulthood and after WM injury.
Collapse
Affiliation(s)
- Lucas Schirmer
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, California, United States.,Department of Pediatrics, University of California, San Francisco, San Francisco, United States.,Department of Paediatrics, University of Cambridge, Cambridge, United Kingdom.,Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Wiebke Möbius
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
| | - Chao Zhao
- Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom.,Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Andrés Cruz-Herranz
- Department of Neurology, University of California, San Francisco, San Francisco, United States.,Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, United States
| | - Lucile Ben Haim
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, California, United States.,Department of Pediatrics, University of California, San Francisco, San Francisco, United States
| | - Christian Cordano
- Department of Neurology, University of California, San Francisco, San Francisco, United States.,Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, United States
| | - Lawrence R Shiow
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, California, United States.,Department of Pediatrics, University of California, San Francisco, San Francisco, United States
| | - Kevin W Kelley
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, California, United States.,Department of Pediatrics, University of California, San Francisco, San Francisco, United States
| | - Boguslawa Sadowski
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
| | - Garrett Timmons
- Department of Neurology, University of California, San Francisco, San Francisco, United States.,Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, United States
| | - Anne-Katrin Pröbstel
- Department of Neurology, University of California, San Francisco, San Francisco, United States.,Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, United States
| | - Jackie N Wright
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, California, United States.,Department of Pediatrics, University of California, San Francisco, San Francisco, United States
| | - Jung Hyung Sin
- Department of Neurology, University of California, San Francisco, San Francisco, United States.,Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, United States
| | - Michael Devereux
- Department of Neurology, University of California, San Francisco, San Francisco, United States.,Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, United States
| | - Daniel E Morrison
- Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom.,Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Sandra M Chang
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, California, United States.,Department of Pediatrics, University of California, San Francisco, San Francisco, United States
| | - Khalida Sabeur
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, California, United States.,Department of Pediatrics, University of California, San Francisco, San Francisco, United States
| | - Ari J Green
- Department of Neurology, University of California, San Francisco, San Francisco, United States.,Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, United States.,Department of Ophthalmology, University of California, San Francisco, San Francisco, United States
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
| | - Robin Jm Franklin
- Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom.,Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - David H Rowitch
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, California, United States.,Department of Pediatrics, University of California, San Francisco, San Francisco, United States.,Department of Paediatrics, University of Cambridge, Cambridge, United Kingdom.,Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom.,Department of Neurosurgery, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
46
|
Muscarinic Receptor M 3R Signaling Prevents Efficient Remyelination by Human and Mouse Oligodendrocyte Progenitor Cells. J Neurosci 2018; 38:6921-6932. [PMID: 29959237 DOI: 10.1523/jneurosci.1862-17.2018] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 05/23/2018] [Accepted: 06/17/2018] [Indexed: 12/13/2022] Open
Abstract
Muscarinic receptor antagonists act as potent inducers of oligodendrocyte differentiation and accelerate remyelination. However, the use of muscarinic antagonists in the clinic is limited by poor understanding of the operant receptor subtype, and questions regarding possible species differences between rodents and humans. Based on high selective expression in human oligodendrocyte progenitor cells (OPCs), we hypothesized that M3R is the functionally relevant receptor. Lentiviral M3R knockdown in human primary CD140a/PDGFαR+ OPCs resulted in enhanced differentiation in vitro and substantially reduced the calcium response following muscarinic agonist treatment. Importantly, following transplantation in hypomyelinating shiverer/rag2 mice, M3R knockdown improved remyelination by human OPCs. Furthermore, conditional M3R ablation in adult NG2-expressing OPCs increased oligodendrocyte differentiation and led to improved spontaneous remyelination in mice. Together, we demonstrate that M3R receptor mediates muscarinic signaling in human OPCs that act to delay differentiation and remyelination, suggesting that M3 receptors are viable targets for human demyelinating disease.SIGNIFICANCE STATEMENT The identification of drug targets aimed at improving remyelination in patients with demyelination disease is a key step in development of effective regenerative therapies to treat diseases, such as multiple sclerosis. Muscarinic receptor antagonists have been identified as effective potentiators of remyelination, but the receptor subtypes that mediate these receptors are unclear. In this study, we show that genetic M3R ablation in both mouse and human cells results in improved remyelination and is mediated by acceleration of oligodendrocyte commitment from oligodendrocyte progenitor cells. Therefore, M3R represents an attractive target for induced remyelination in human disease.
Collapse
|
47
|
Zhang J, Lakowicz JR. A superior bright NIR luminescent nanoparticle preparation and indicating calcium signaling detection in cells and small animals. Cell Biosci 2018; 8:37. [PMID: 29928497 PMCID: PMC5987641 DOI: 10.1186/s13578-018-0235-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 05/21/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Near-field fluorescence (NFF) effects were employed to develop a novel near-infrared (NIR) luminescent nanoparticle (LNP) with superior brightness. The LNP is used as imaging contrast agent for cellular and small animal imaging and furthermore suggested to use for detecting voltage-sensitive calcium in living cells and animals with high sensitivity. RESULTS NIR Indocyanine green (ICG) dye was conjugated with human serum albumin (HSA) followed by covalently binding to gold nanorod (AuNR). The AuNR displayed dual plasmons from transverse and longitudinal axis, and the longitudinal plasmon was localized at the NIR region which could efficiently couple with the excitation and emission of ICG dye leading to a largely enhanced NFF. The enhancement factor was measured to be about 16-fold using both ensemble and single nanoparticle spectral methods. As an imaging contrast agent, the ICG-HSA-Au complex (abbreviate as ICG-Au) was conjugated on HeLa cells and fluorescence cell images were recorded on a time-resolved confocal microscope. The emission signals of ICG-Au complexes were distinctly resolved as the individual spots that were observed over the cellular backgrounds due to their strong brightness as well as shortened lifetime. The LNPs were also tested to have a low cytotoxicity. The ICG-Au complexes were injected below the skin surface of mouse showing emission spots 5-fold brighter than those from the same amount of free ICG-HSA conjugates. CONCLUSIONS Based on the observations in this research, the excitation and emission of NIR ICG dyes were found to be able to sufficiently couple with the longitudinal plasmon of AuNRs leading to a largely enhanced NFF. Using the LNP with super-brightness as a contrast agent, the ICG-Au complex could be resolved from the background in the cell and small animal imaging. The novel NIR LNP has also a great potential for detection of voltage-gated calcium concentration in the cell and living animal with a high sensitivity.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Biochemistry and Molecular Biology, Center for Fluorescence Spectroscopy, University of Maryland School of Medicine, 725 West Lombard Street, Baltimore, MD 21201 USA
- Present Address: Vigene Biosciences Inc., 9430 Key W. Ave Suite 105, Rockville, MD 20850 USA
| | - Joseph. R. Lakowicz
- Department of Biochemistry and Molecular Biology, Center for Fluorescence Spectroscopy, University of Maryland School of Medicine, 725 West Lombard Street, Baltimore, MD 21201 USA
| |
Collapse
|
48
|
Li T, Wang L, Ma T, Wang S, Niu J, Li H, Xiao L. Dynamic Calcium Release From Endoplasmic Reticulum Mediated by Ryanodine Receptor 3 Is Crucial for Oligodendroglial Differentiation. Front Mol Neurosci 2018; 11:162. [PMID: 29867353 PMCID: PMC5968115 DOI: 10.3389/fnmol.2018.00162] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 04/30/2018] [Indexed: 12/15/2022] Open
Abstract
Increased intracellular Ca2+ in oligodendrocyte progenitor cells (OPCs) is important to initiate their differentiation, but the intracellular Ca2+ channel involved in this process remains unclear. As a Ca2+-induced Ca2+ release (CICR) channel that mediates endoplasmic reticulum (ER) Ca2+ release, the role of ryanodine receptors (RyRs) in oligodendroglial development is unexplored. In the present study, we observed that among the three mammalian isoforms, oligodendroglial lineage cells selectively expressed RyR3. Strong RyR3-positive signal was distributed all over the cytoplasm and processes in OPCs and/or immature OLs (imOLs), whereas it gradually decreased and was located mainly around the perinuclear region in mature oligodendrocytes (OLs). In addition, RyR3-mediated intracellular Ca2+ waves following caffeine stimulation were correlated with the expression pattern of RyR3, in which high flat Ca2+ fluctuations and oscillatory Ca2+ waves were more frequently recorded in OPCs and/or imOLs than in OLs. Through further functional exploration, we demonstrated that pretreatment with the RyR antagonist ryanodine could neutralize the increase in intracellular Ca2+ induced by OPC differentiation and reduce the number of mature OLs. Moreover, gene-level knockdown of RyR3 by lentivirus in OPCs resulted in inhibition of OPC differentiation. Taken together, our results provide new insight into the crucial role of RyR3-mediated ER Ca2+ release in the regulation of OPC differentiation and/or myelination.
Collapse
Affiliation(s)
- Tao Li
- Chongqing Key Laboratory of Neurobiology, Department of Histology and Embryology, Third Military Medical University, Chongqing, China
| | - Lingyun Wang
- Chongqing Key Laboratory of Neurobiology, Department of Histology and Embryology, Third Military Medical University, Chongqing, China
| | - Teng Ma
- Chongqing Key Laboratory of Neurobiology, Department of Histology and Embryology, Third Military Medical University, Chongqing, China
| | - Shouyu Wang
- Chongqing Key Laboratory of Neurobiology, Department of Histology and Embryology, Third Military Medical University, Chongqing, China
| | - Jianqin Niu
- Chongqing Key Laboratory of Neurobiology, Department of Histology and Embryology, Third Military Medical University, Chongqing, China
| | - Hongli Li
- Chongqing Key Laboratory of Neurobiology, Department of Histology and Embryology, Third Military Medical University, Chongqing, China
| | - Lan Xiao
- Chongqing Key Laboratory of Neurobiology, Department of Histology and Embryology, Third Military Medical University, Chongqing, China
| |
Collapse
|
49
|
TRPV4 is functionally expressed in oligodendrocyte precursor cells and increases their proliferation. Pflugers Arch 2018; 470:705-716. [PMID: 29569183 DOI: 10.1007/s00424-018-2130-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 02/17/2018] [Accepted: 02/20/2018] [Indexed: 10/17/2022]
Abstract
Oligodendrocytes, which differentiate from oligodendrocyte precursor cells (OPCs), ensheath axons with myelin, play an essential role in rapid conduction of action potentials and metabolically support neurons. Elucidation of the mechanisms underlying the proliferation, migration, differentiation, and survival of OPCs is considered indispensable for determining the causes of central nervous system diseases. However, the relationship between these functions of OPCs and their intracellular Ca2+ signaling has not been fully elucidated. Here, we investigated the function of transient receptor potential vanilloid 4 (TRPV4), a Ca2+-permeable channel that responds to hypo-osmolarity, mild temperature, mechanical stimulation, and endogenous arachidonic acid metabolites, in OPCs. Trpv4 mRNA was detected in OPCs in vivo and in primary cultured rat OPCs. In Ca2+ imaging experiments, treatment with the selective TRPV4 agonist GSK1016790A induced sustained elevation of the intracellular Ca2+ concentration in OPCs in a concentration-dependent manner, which was almost completely suppressed by co-treatment with the selective TRPV4 antagonist HC067047. Stimulation of TRPV4 by GSK1016790A augmented OPC proliferation, which was abolished by co-treatment with HC067047, the intracellular Ca2+ chelator BAPTA-AM, and the protein kinase C inhibitor bisindolylmaleimide II. By contrast, GSK1016790A did not significantly affect the migration or differentiation of OPCs. Taken together, these results suggest that TRPV4 is functionally expressed in OPCs and increases the proliferation of these cells without affecting their ability to differentiate into oligodendrocytes.
Collapse
|
50
|
Weider M, Starost LJ, Groll K, Küspert M, Sock E, Wedel M, Fröb F, Schmitt C, Baroti T, Hartwig AC, Hillgärtner S, Piefke S, Fadler T, Ehrlich M, Ehlert C, Stehling M, Albrecht S, Jabali A, Schöler HR, Winkler J, Kuhlmann T, Wegner M. Nfat/calcineurin signaling promotes oligodendrocyte differentiation and myelination by transcription factor network tuning. Nat Commun 2018; 9:899. [PMID: 29500351 PMCID: PMC5834605 DOI: 10.1038/s41467-018-03336-3] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 02/05/2018] [Indexed: 02/06/2023] Open
Abstract
Oligodendrocytes produce myelin for rapid transmission and saltatory conduction of action potentials in the vertebrate central nervous system. Activation of the myelination program requires several transcription factors including Sox10, Olig2, and Nkx2.2. Functional interactions among them are poorly understood and important components of the regulatory network are still unknown. Here, we identify Nfat proteins as Sox10 targets and regulators of oligodendroglial differentiation in rodents and humans. Overall levels and nuclear fraction increase during differentiation. Inhibition of Nfat activity impedes oligodendrocyte differentiation in vitro and in vivo. On a molecular level, Nfat proteins cooperate with Sox10 to relieve reciprocal repression of Olig2 and Nkx2.2 as precondition for oligodendroglial differentiation and myelination. As Nfat activity depends on calcium-dependent activation of calcineurin signaling, regulatory network and oligodendroglial differentiation become sensitive to calcium signals. NFAT proteins are also detected in human oligodendrocytes, downregulated in active multiple sclerosis lesions and thus likely relevant in demyelinating disease.
Collapse
Affiliation(s)
- Matthias Weider
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054, Erlangen, Germany
| | - Laura Julia Starost
- Institute of Neuropathology, University Hospital Münster, D-48149, Münster, Germany.,Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, D-48149, Münster, Germany
| | - Katharina Groll
- Institute of Neuropathology, University Hospital Münster, D-48149, Münster, Germany
| | - Melanie Küspert
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054, Erlangen, Germany
| | - Elisabeth Sock
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054, Erlangen, Germany
| | - Miriam Wedel
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054, Erlangen, Germany
| | - Franziska Fröb
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054, Erlangen, Germany
| | - Christian Schmitt
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054, Erlangen, Germany
| | - Tina Baroti
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054, Erlangen, Germany
| | - Anna C Hartwig
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054, Erlangen, Germany
| | - Simone Hillgärtner
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054, Erlangen, Germany
| | - Sandra Piefke
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054, Erlangen, Germany
| | - Tanja Fadler
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054, Erlangen, Germany
| | - Marc Ehrlich
- Institute of Neuropathology, University Hospital Münster, D-48149, Münster, Germany.,Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, D-48149, Münster, Germany
| | - Corinna Ehlert
- Institute of Neuropathology, University Hospital Münster, D-48149, Münster, Germany
| | - Martin Stehling
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, D-48149, Münster, Germany
| | - Stefanie Albrecht
- Institute of Neuropathology, University Hospital Münster, D-48149, Münster, Germany
| | - Ammar Jabali
- Institute of Neuropathology, University Hospital Münster, D-48149, Münster, Germany
| | - Hans R Schöler
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, D-48149, Münster, Germany
| | - Jürgen Winkler
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054, Erlangen, Germany
| | - Tanja Kuhlmann
- Institute of Neuropathology, University Hospital Münster, D-48149, Münster, Germany.
| | - Michael Wegner
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054, Erlangen, Germany.
| |
Collapse
|