1
|
Duan Y, Ye C, Liao J, Xie X. LY2940094, an NOPR antagonist, promotes oligodendrocyte generation and myelin recovery in an NOPR independent manner. Neurotherapeutics 2024; 21:e00424. [PMID: 39004556 PMCID: PMC11581876 DOI: 10.1016/j.neurot.2024.e00424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 07/05/2024] [Accepted: 07/05/2024] [Indexed: 07/16/2024] Open
Abstract
The myelin sheath plays crucial roles in brain development and neuronal functions. In the central nervous system, myelin is generated by oligodendrocytes, that differentiate from oligodendrocyte progenitor cells (OPC). In demyelinating diseases, the differentiation capacity of OPC is impaired and remyelination is dampened. Boosting remyelination by promoting OPC differentiation is a novel strategy for the treatment of demyelinating diseases. The opioid system, which consists of four receptors and their ligands, has been implicated in OPC differentiation and myelin formation. However, the exact roles of each opioid receptor and the relevant pharmacological molecules in OPC differentiation and myelin formation remain elusive. In the present study, specific agonists and antagonists of each opioid receptor were used to explore the function of opioid receptors in OPC differentiation. Nociceptin/orphanin FQ receptor (NOPR) specific antagonist LY2940094 was found to stimulate OPC differentiation and myelination in both in vitro and in vivo models. Unexpectedly, other NOPR ligands did not affect OPC differentiation, and NOPR knockdown did not mimic or impede the effect of LY2940094. LY2940094 was found to modulate the expression of the oligodendrocytes differentiation-associated transcription factors ID4 and Myrf, although the exact mechanism remains unclear. Since LY2940094 has been tested clinically to treat depression and alcohol dependency and has displayed an acceptable safety profile, it may provide an alternative approach to treat demyelinating diseases.
Collapse
Affiliation(s)
- Yanhui Duan
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Chenyuan Ye
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China; State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jingyi Liao
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China; 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
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China; State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, China.
| |
Collapse
|
2
|
Emery B, Wood TL. Regulators of Oligodendrocyte Differentiation. Cold Spring Harb Perspect Biol 2024; 16:a041358. [PMID: 38503504 PMCID: PMC11146316 DOI: 10.1101/cshperspect.a041358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2024]
Abstract
Myelination has evolved as a mechanism to ensure fast and efficient propagation of nerve impulses along axons. Within the central nervous system (CNS), myelination is carried out by highly specialized glial cells, oligodendrocytes. The formation of myelin is a prolonged aspect of CNS development that occurs well into adulthood in humans, continuing throughout life in response to injury or as a component of neuroplasticity. The timing of myelination is tightly tied to the generation of oligodendrocytes through the differentiation of their committed progenitors, oligodendrocyte precursor cells (OPCs), which reside throughout the developing and adult CNS. In this article, we summarize our current understanding of some of the signals and pathways that regulate the differentiation of OPCs, and thus the myelination of CNS axons.
Collapse
Affiliation(s)
- Ben Emery
- Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health & Science University, Portland, Oregon 97239, USA
| | - Teresa L Wood
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey 07103, USA
| |
Collapse
|
3
|
Hu B, Moiseev D, Schena I, Faezov B, Dunbrack R, Chernoff J, Li J. PAK2 is necessary for myelination in the peripheral nervous system. Brain 2024; 147:1809-1821. [PMID: 38079473 PMCID: PMC11068108 DOI: 10.1093/brain/awad413] [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: 07/19/2023] [Revised: 10/03/2023] [Accepted: 11/12/2023] [Indexed: 02/12/2024] Open
Abstract
Myelination enables electrical impulses to propagate on axons at the highest speed, encoding essential life functions. The Rho family GTPases, RAC1 and CDC42, have been shown to critically regulate Schwann cell myelination. P21-activated kinase 2 (PAK2) is an effector of RAC1/CDC42, but its specific role in myelination remains undetermined. We produced a Schwann cell-specific knockout mouse of Pak2 (scPak2-/-) to evaluate PAK2's role in myelination. Deletion of Pak2, specifically in mouse Schwann cells, resulted in severe hypomyelination, slowed nerve conduction velocity and behaviour dysfunctions in the scPak2-/- peripheral nerve. Many Schwann cells in scPak2-/- sciatic nerves were arrested at the stage of axonal sorting. These abnormalities were rescued by reintroducing Pak2, but not the kinase-dead mutation of Pak2, via lentivirus delivery to scPak2-/- Schwann cells in vivo. Moreover, ablation of Pak2 in Schwann cells blocked the promyelinating effect driven by neuregulin-1, prion protein and inactivated RAC1/CDC42. Conversely, the ablation of Pak2 in neurons exhibited no phenotype. Such PAK2 activity can also be either enhanced or inhibited by different myelin lipids. We have identified a novel promyelinating factor, PAK2, that acts as a critical convergence point for multiple promyelinating signalling pathways. The promyelination by PAK2 is Schwann cell-autonomous. Myelin lipids, identified as inhibitors or activators of PAK2, may be utilized to develop therapies for repairing abnormal myelin in peripheral neuropathies.
Collapse
Affiliation(s)
- Bo Hu
- Department of Neurology, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Daniel Moiseev
- Department of Neurology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Isabella Schena
- Department of Neurology, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Bulat Faezov
- Cancer Biology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
- Fundamental Medicine and Biology, Kazan Federal University, Kazan 420008, Russia
| | - Roland Dunbrack
- Cancer Biology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Jonathan Chernoff
- Cancer Biology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Jun Li
- Department of Neurology, Houston Methodist Research Institute, Houston, TX 77030, USA
| |
Collapse
|
4
|
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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 10/13/2023] [Indexed: 11/09/2023]
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
|
5
|
Xu J, Peng Q, Cai J, Shangguan J, Su W, Chen G, Sun H, Zhu C, Gu Y. The Schwann cell-specific G-protein Gαo (Gnao1) is a cell-intrinsic controller contributing to the regulation of myelination in peripheral nerve system. Acta Neuropathol Commun 2024; 12:24. [PMID: 38331815 PMCID: PMC10854112 DOI: 10.1186/s40478-024-01720-3] [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: 08/02/2023] [Accepted: 12/27/2023] [Indexed: 02/10/2024] Open
Abstract
Myelin sheath abnormality is the cause of various neurodegenerative diseases (NDDs). G-proteins and their coupled receptors (GPCRs) play the important roles in myelination. Gnao1, encoding the major Gα protein (Gαo) in mammalian nerve system, is required for normal motor function. Here, we show that Gnao1 restricted to Schwann cell (SCs) lineage, but not neurons, negatively regulate SC differentiation, myelination, as well as re-myelination in peripheral nervous system (PNS). Mice lacking Gnao1 expression in SCs exhibit faster re-myelination and motor function recovery after nerve injury. Conversely, mice with Gnao1 overexpression in SCs display the insufficient myelinating capacity and delayed re-myelination. In vitro, Gnao1 deletion in SCs promotes SC differentiation. We found that Gnao1 knockdown in SCs resulting in the elevation of cAMP content and the activation of PI3K/AKT pathway, both associated with SC differentiation. The analysis of RNA sequencing data further evidenced that Gnao1 deletion cause the increased expression of myelin-related molecules and activation of regulatory pathways. Taken together, our data indicate that Gnao1 negatively regulated SC differentiation by reducing cAMP level and inhibiting PI3K-AKT cascade activation, identifying a novel drug target for the treatment of demyelinating diseases.
Collapse
Affiliation(s)
- Jinghui Xu
- Jiangsu Key Laboratory of Neuroregeneration, Co-Innovation Center of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, JS, 226001, People's Republic of China
| | - Qianqian Peng
- Jiangsu Key Laboratory of Neuroregeneration, Co-Innovation Center of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, JS, 226001, People's Republic of China
| | - Jieyi Cai
- Jiangsu Key Laboratory of Neuroregeneration, Co-Innovation Center of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, JS, 226001, People's Republic of China
| | - Jianghong Shangguan
- Jiangsu Key Laboratory of Neuroregeneration, Co-Innovation Center of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, JS, 226001, People's Republic of China
| | - Wenfeng Su
- Jiangsu Key Laboratory of Neuroregeneration, Co-Innovation Center of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, JS, 226001, People's Republic of China
| | - Gang Chen
- Jiangsu Key Laboratory of Neuroregeneration, Co-Innovation Center of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, JS, 226001, People's Republic of China
| | - Hualin Sun
- Jiangsu Key Laboratory of Neuroregeneration, Co-Innovation Center of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, JS, 226001, People's Republic of China
| | - Changlai Zhu
- Jiangsu Key Laboratory of Neuroregeneration, Co-Innovation Center of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, JS, 226001, People's Republic of China.
| | - Yun Gu
- Jiangsu Key Laboratory of Neuroregeneration, Co-Innovation Center of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, JS, 226001, People's Republic of China.
| |
Collapse
|
6
|
Kinjo T, Ebisawa S, Nokubo T, Hashimoto M, Yamada T, Oshio M, Nakamura R, Uno K, Kuramoto N. Post-translational modifications of the apelin receptor regulate its functional expression. AIMS Neurosci 2023; 10:282-299. [PMID: 38188005 PMCID: PMC10767067 DOI: 10.3934/neuroscience.2023022] [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: 07/14/2023] [Revised: 10/24/2023] [Accepted: 10/26/2023] [Indexed: 01/09/2024] Open
Abstract
Post-translational modifications (PTMs) are protein modifications that occur after protein biosynthesis, playing a crucial role in regulating protein function. They are involved in the functional expression of G-protein-coupled receptors (GPCRs), as well as intracellular and secretory protein signaling. Here, we aimed to investigate the PTMs of the apelin receptor (APLNR), a GPCR and their potential influence on the receptor's function. In an in vitro experiment using HEK cells, we only observed glycosylation as a PTM of the APLNR and ineffective receptor signaling by the agonist, (Pyr1)-apelin-13. In contrast, when analyzing mouse spinal cord, we detected glycosylation and other PTMs, excluding isopeptidation. This suggests that additional PTMs are involved in the functional expression of the APLNR in vitro. In summary, these findings suggest that the APLNR in vivo requires multiple PTMs for functional expression. To comprehensively understand the pharmacological effects of the APLNR, it is essential to establish an in vitro system that adequately replicates the receptor's PTM profile. Nonetheless, it is crucial to overcome the challenge of heat-sensitive proteolysis in APLNR studies. By elucidating the regulation of PTMs, further research has the potential to advance the analysis and pharmacological studies of both the apelin/APLNR system and GPCR signal modulation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Nobuyuki Kuramoto
- Laboratory of Molecular Pharmacology, Faculty of Pharmaceutical Sciences, Setsunan University, Hirakata, Osaka 573-0101, Japan
| |
Collapse
|
7
|
Silva ME, Hernández-Andrade M, Abasolo N, Espinoza-Cruells C, Mansilla JB, Reyes CR, Aranda S, Esteban Y, Rodriguez-Calvo R, Martorell L, Muntané G, Rivera FJ, Vilella E. DDR1 and Its Ligand, Collagen IV, Are Involved in In Vitro Oligodendrocyte Maturation. Int J Mol Sci 2023; 24:ijms24021742. [PMID: 36675255 PMCID: PMC9866737 DOI: 10.3390/ijms24021742] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 01/09/2023] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
Discoidin domain receptor 1 (DDR1) is a tyrosine kinase receptor expressed in epithelial cells from different tissues in which collagen binding activates pleiotropic functions. In the brain, DDR1 is mainly expressed in oligodendrocytes (OLs), the function of which is unclear. Whether collagen can activate DDR1 in OLs has not been studied. Here, we assessed the expression of DDR1 during in vitro OL differentiation, including collagen IV incubation, and the capability of collagen IV to induce DDR1 phosphorylation. Experiments were performed using two in vitro models of OL differentiation: OLs derived from adult rat neural stem cells (NSCs) and the HOG16 human oligodendroglial cell line. Immunocytofluorescence, western blotting, and ELISA were performed to analyze these questions. The differentiation of OLs from NSCs was addressed using oligodendrocyte transcription factor 2 (Olig2) and myelin basic protein (MBP). In HOG16 OLs, collagen IV induced DDR1 phosphorylation through slow and sustained kinetics. In NSC-derived OLs, DDR1 was found in a high proportion of differentiating cells (MBP+/Olig2+), but its protein expression was decreased in later stages. The addition of collagen IV did not change the number of DDR1+/MBP+ cells but did accelerate OL branching. Here, we provide the first demonstration that collagen IV mediates the phosphorylation of DDR1 in HOG16 cells and that the in vitro co-expression of DDR1 and MBP is associated with accelerated branching during the differentiation of primary OLs.
Collapse
Affiliation(s)
- Maria Elena Silva
- Laboratory of Stem Cells and Neuroregeneration, Institute of Anatomy, Histology and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia 5090000, Chile
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia 5090000, Chile
- Institute of Pharmacy, Faculty of Sciences, Universidad Austral de Chile, Valdivia 5090000, Chile
| | - Matías Hernández-Andrade
- Laboratory of Stem Cells and Neuroregeneration, Institute of Anatomy, Histology and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia 5090000, Chile
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia 5090000, Chile
| | - Nerea Abasolo
- Hospital Universitari Institut Pere Mata, Institut d’Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, 43206 Reus, Spain
| | - Cristóbal Espinoza-Cruells
- Laboratory of Stem Cells and Neuroregeneration, Institute of Anatomy, Histology and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia 5090000, Chile
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia 5090000, Chile
| | - Josselyne B. Mansilla
- Laboratory of Stem Cells and Neuroregeneration, Institute of Anatomy, Histology and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia 5090000, Chile
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia 5090000, Chile
| | - Carolina R. Reyes
- Laboratory of Stem Cells and Neuroregeneration, Institute of Anatomy, Histology and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia 5090000, Chile
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia 5090000, Chile
| | - Selena Aranda
- Hospital Universitari Institut Pere Mata, Institut d’Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, 43206 Reus, Spain
- Centro de Investigación Biomédica en Red en Salud Mental, CIBERSAM-Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Yaiza Esteban
- Vascular Medicine and Metabolism Unit, Research Unit on Lipids and Atherosclerosis, “Sant Joan” University Hospital, Institut d’Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, 43204 Reus, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas (CIBERDEM), 28029 Madrid, Spain
| | - Ricardo Rodriguez-Calvo
- Vascular Medicine and Metabolism Unit, Research Unit on Lipids and Atherosclerosis, “Sant Joan” University Hospital, Institut d’Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, 43204 Reus, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas (CIBERDEM), 28029 Madrid, Spain
| | - Lourdes Martorell
- Hospital Universitari Institut Pere Mata, Institut d’Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, 43206 Reus, Spain
- Centro de Investigación Biomédica en Red en Salud Mental, CIBERSAM-Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Gerard Muntané
- Hospital Universitari Institut Pere Mata, Institut d’Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, 43206 Reus, Spain
- Centro de Investigación Biomédica en Red en Salud Mental, CIBERSAM-Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Francisco J. Rivera
- Laboratory of Stem Cells and Neuroregeneration, Institute of Anatomy, Histology and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia 5090000, Chile
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia 5090000, Chile
- Translational Regenerative Neurobiology Group, Molecular and Integrative Biosciences Research Program (MIBS), Faculty of Biological and Environmental Sciences, University of Helsinki, 00014 Helsinki, Finland
- Correspondence: or (F.J.R.); (E.V.); Tel.: +358-50-598-8142 or +56-63-229-3011 (F.J.R.); +34-658-513-138 (E.V.)
| | - Elisabet Vilella
- Hospital Universitari Institut Pere Mata, Institut d’Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, 43206 Reus, Spain
- Centro de Investigación Biomédica en Red en Salud Mental, CIBERSAM-Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: or (F.J.R.); (E.V.); Tel.: +358-50-598-8142 or +56-63-229-3011 (F.J.R.); +34-658-513-138 (E.V.)
| |
Collapse
|
8
|
Humanized zebrafish as a tractable tool for in vivo evaluation of pro-myelinating drugs. Cell Chem Biol 2022; 29:1541-1555.e7. [PMID: 36126653 DOI: 10.1016/j.chembiol.2022.08.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 05/25/2022] [Accepted: 08/24/2022] [Indexed: 12/14/2022]
Abstract
Therapies that promote neuroprotection and axonal survival by enhancing myelin regeneration are an unmet need to prevent disability progression in multiple sclerosis. Numerous potentially beneficial compounds have originated from phenotypic screenings but failed in clinical trials. It is apparent that current cell- and animal-based disease models are poor predictors of positive treatment options, arguing for novel experimental approaches. Here we explore the experimental power of humanized zebrafish to foster the identification of pro-remyelination compounds via specific inhibition of GPR17. Using biochemical and imaging techniques, we visualize the expression of zebrafish (zf)-gpr17 during the distinct stages of oligodendrocyte development, thereby demonstrating species-conserved expression between zebrafish and mammals. We also demonstrate species-conserved function of zf-Gpr17 using genetic loss-of-function and rescue techniques. Finally, using GPR17-humanized zebrafish, we provide proof of principle for in vivo analysis of compounds acting via targeted inhibition of human GPR17. We anticipate that GPR17-humanized zebrafish will markedly improve the search for effective pro-myelinating pharmacotherapies.
Collapse
|
9
|
Suo N, He B, Cui S, Yang Y, Wang M, Yuan Q, Xie X. The orphan G protein-coupled receptor GPR149 is a negative regulator of myelination and remyelination. Glia 2022; 70:1992-2008. [PMID: 35758525 DOI: 10.1002/glia.24233] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 06/09/2022] [Accepted: 06/16/2022] [Indexed: 12/14/2022]
Abstract
Myelin sheath, formed by oligodendrocytes (OLs) in the central nervous system (CNS) and Schwann cells in periphery, plays a critical role in supporting neuronal functions. OLs, differentiated from oligodendrocyte precursor cells (OPCs), are important for myelination during development and myelin repair in CNS demyelinating disease. To identify mechanisms of myelin development and remyelination after myelin damage is of great clinical interest. Here we show that the orphan G protein-coupled receptor GPR149, enriched in OPCs, negatively regulate OPC to OL differentiation, myelination, as well as remyelination. The expression of GPR149 is downregulated during OPCs differentiation into OLs. GPR149 deficiency does not affect the number of OPCs, but promotes OPC to OL differentiation which results in earlier development of myelin. In cuprizone-induced demyelination model, GPR149 deficiency significantly enhances myelin regeneration. Further study indicates that GPR149 may regulate OL differentiation and myelin formation via MAPK/ERK pathway. Our study suggests that deleting or blocking GPR149 might be an intriguing way to promote myelin repair in demyelinating diseases.
Collapse
Affiliation(s)
- Na Suo
- CAS Key Laboratory of Receptor Research, the National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Bingqing He
- CAS Key Laboratory of Receptor Research, the National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,School of Pharmacy, University of Chinese Academy of Sciences, Beijing, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Shihao Cui
- CAS Key Laboratory of Receptor Research, the National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,School of Pharmacy, University of Chinese Academy of Sciences, Beijing, China
| | - Ying Yang
- CAS Key Laboratory of Receptor Research, the National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,School of Pharmacy, University of Chinese Academy of Sciences, Beijing, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Min Wang
- CAS Key Laboratory of Receptor Research, the National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Qianting Yuan
- CAS Key Laboratory of Receptor Research, the National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Xin Xie
- CAS Key Laboratory of Receptor Research, the National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,School of Pharmacy, University of Chinese Academy of Sciences, Beijing, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, China.,State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| |
Collapse
|
10
|
Oscillatory calcium release and sustained store-operated oscillatory calcium signaling prevents differentiation of human oligodendrocyte progenitor cells. Sci Rep 2022; 12:6160. [PMID: 35418597 PMCID: PMC9007940 DOI: 10.1038/s41598-022-10095-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 03/31/2022] [Indexed: 11/08/2022] Open
Abstract
Endogenous remyelination in demyelinating diseases such as multiple sclerosis is contingent upon the successful differentiation of oligodendrocyte progenitor cells (OPCs). Signaling via the Gαq-coupled muscarinic receptor (M1/3R) inhibits human OPC differentiation and impairs endogenous remyelination in experimental models. We hypothesized that calcium release following Gαq-coupled receptor (GqR) activation directly regulates human OPC (hOPC) cell fate. In this study, we show that specific GqR agonists activating muscarinic and metabotropic glutamate receptors induce characteristic oscillatory calcium release in hOPCs and that these agonists similarly block hOPC maturation in vitro. Both agonists induce calcium release from endoplasmic reticulum (ER) stores and store operated calcium entry (SOCE) likely via STIM/ORAI-based channels. siRNA mediated knockdown (KD) of obligate calcium sensors STIM1 and STIM2 decreased the magnitude of muscarinic agonist induced oscillatory calcium release and attenuated SOCE in hOPCs. In addition, STIM2 expression was necessary to maintain the frequency of calcium oscillations and STIM2 KD reduced spontaneous OPC differentiation. Furthermore, STIM2 siRNA prevented the effects of muscarinic agonist treatment on OPC differentiation suggesting that SOCE is necessary for the anti-differentiative action of muscarinic receptor-dependent signaling. Finally, using a gain-of-function approach with an optogenetic STIM lentivirus, we demonstrate that independent activation of SOCE was sufficient to significantly block hOPC differentiation and this occurred in a frequency dependent manner while increasing hOPC proliferation. These findings suggest that intracellular calcium oscillations directly regulate hOPC fate and that modulation of calcium oscillation frequency may overcome inhibitory Gαq-coupled signaling that impairs myelin repair.
Collapse
|
11
|
Ahmadzai MM, McClain JL, Dharshika C, Seguella L, Giancola F, De Giorgio R, Gulbransen BD. LPAR1 regulates enteric nervous system function through glial signaling and contributes to chronic intestinal pseudo-obstruction. J Clin Invest 2022; 132:149464. [PMID: 35166239 PMCID: PMC8843750 DOI: 10.1172/jci149464] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 12/21/2021] [Indexed: 12/30/2022] Open
Abstract
Gastrointestinal motility disorders involve alterations to the structure and/or function of the enteric nervous system (ENS) but the causal mechanisms remain unresolved in most cases. Homeostasis and disease in the ENS are processes that are regulated by enteric glia. Signaling mediated through type I lysophosphatidic acid receptors (LPAR1) has recently emerged as an important mechanism that contributes to disease, in part, through effects on peripheral glial survival and function. Enteric glia express LPAR1 but its role in ENS function and motility disorders is unknown. We used a combination of genetic, immunohistochemical, calcium imaging, and in vivo pharmacological approaches to investigate the role of LPAR1 in enteric glia. LPAR1 was enriched in enteric glia in mice and humans and LPA stimulated intracellular calcium responses in enteric glia, subsequently recruiting activity in a subpopulation of myenteric neurons. Blocking LPAR1 in vivo with AM966 attenuated gastrointestinal motility in mice and produced marked enteric neuro- and gliopathy. Samples from humans with chronic intestinal pseudo-obstruction (CIPO), a severe motility disorder, showed reduced glial LPAR1 expression in the colon and ileum. These data suggest that enteric glial LPAR1 signaling regulates gastrointestinal motility through enteric glia and could contribute to severe motility disorders in humans such as CIPO.
Collapse
Affiliation(s)
- Mohammad M Ahmadzai
- Department of Physiology, Neuroscience Program.,College of Osteopathic Medicine, and
| | | | - Christine Dharshika
- Department of Physiology, Neuroscience Program.,College of Human Medicine, Michigan State University, East Lansing, Michigan, USA
| | - Luisa Seguella
- Department of Physiology, Neuroscience Program.,Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome, Italy
| | - Fiorella Giancola
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy.,St. Orsola-Malpighi Hospital, Bologna, Italy
| | - Roberto De Giorgio
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | | |
Collapse
|
12
|
The oligodendrocyte-enriched orphan G protein-coupled receptor Gpr62 is dispensable for central nervous system myelination. Neural Dev 2021; 16:6. [PMID: 34844642 PMCID: PMC8630896 DOI: 10.1186/s13064-021-00156-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 11/15/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Myelination is a highly regulated process in the vertebrate central nervous system (CNS) whereby oligodendrocytes wrap axons with multiple layers of insulating myelin in order to allow rapid electrical conduction. Establishing the proper pattern of myelin in neural circuits requires communicative axo-glial interactions, however, the molecular interactions that occur between oligodendrocytes and axons during developmental myelination and myelin maintenance remain to be fully elucidated. Our previous work identified G protein-coupled receptor 62 (Gpr62), an uncharacterized orphan g-protein coupled receptor, as being selectively expressed by mature oligodendrocytes within the CNS, suggesting a potential role in myelination or axoglial interactions. However, no studies to date have assessed the functional requirement for Gpr62 in oligodendrocyte development or CNS myelination. METHODS To address this, we generated a knockout mouse strain lacking the Gpr62 gene. We assessed CNS myelination during both postnatal development and adulthood using immunohistochemistry, electron microscopy and western blot. In addition, we utilized AAV-mediated expression of a tagged Gpr62 in oligodendrocytes to determine the subcellular localization of the protein in vivo. RESULTS We find that virally expressed Gpr62 protein is selectively expressed on the adaxonal myelin layer, suggestive of a potential role for Gpr62 in axo-myelinic signaling. Nevertheless, Gpr62 knockout mice display normal oligodendrocyte numbers and apparently normal myelination within the CNS during both postnatal development and adulthood. CONCLUSIONS We conclude that in spite of being well-placed to mediate neuronal-oligodendrocyte communications, Gpr62 is overall dispensable for CNS myelination.
Collapse
|
13
|
Motavaf M, Piao X. Oligodendrocyte Development and Implication in Perinatal White Matter Injury. Front Cell Neurosci 2021; 15:764486. [PMID: 34803612 PMCID: PMC8599582 DOI: 10.3389/fncel.2021.764486] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 10/11/2021] [Indexed: 12/12/2022] Open
Abstract
Perinatal white matter injury (WMI) is the most common brain injury in premature infants and can lead to life-long neurological deficits such as cerebral palsy. Preterm birth is typically accompanied by inflammation and hypoxic-ischemic events. Such perinatal insults negatively impact maturation of oligodendrocytes (OLs) and cause myelination failure. At present, no treatment options are clinically available to prevent or cure WMI. Given that arrested OL maturation plays a central role in the etiology of perinatal WMI, an increased interest has emerged regarding the functional restoration of these cells as potential therapeutic strategy. Cell transplantation and promoting endogenous oligodendrocyte function are two potential options to address this major unmet need. In this review, we highlight the underlying pathophysiology of WMI with a specific focus on OL biology and their implication for the development of new therapeutic targets.
Collapse
Affiliation(s)
- Mahsa Motavaf
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Xianhua Piao
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, United States.,Newborn Brain Research Institute, University of California, San Francisco, San Francisco, CA, United States.,Weill Institute for Neuroscience, University of California, San Francisco, San Francisco, CA, United States.,Division of Neonatology, Department of Pediatrics, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
14
|
Qian Z, Li H, Yang H, Yang Q, Lu Z, Wang L, Chen Y, Li X. Osteocalcin attenuates oligodendrocyte differentiation and myelination via GPR37 signaling in the mouse brain. SCIENCE ADVANCES 2021; 7:eabi5811. [PMID: 34678058 PMCID: PMC8535816 DOI: 10.1126/sciadv.abi5811] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 09/01/2021] [Indexed: 05/24/2023]
Abstract
The bone-derived hormone osteocalcin (OCN) is crucial for brain development and neural cognitive functions, yet the exact roles of OCN in central nervous system (CNS) remain elusive. Here, we find that genetic deletion of OCN facilitates oligodendrocyte (OL) differentiation and hypermyelination in the CNS. Although dispensable for the proliferation of oligodendrocyte precursor cells (OPCs), OCN is critical for the myelination of OLs, which affects myelin production and remyelination after demyelinating injury. Genome-wide RNA sequencing analyses reveal that OCN regulates a number of G protein–coupled receptors and myelination-associated transcription factors, of which Myrf might be a key downstream effector in OLs. GPR37 is identified as a previously unknown receptor for OCN, thus regulating OL differentiation and CNS myelination. Overall, these findings suggest that OCN orchestrates the transition between OPCs and myelinating OLs via GPR37 signaling, and hence, the OCN/GPR37 pathway regulates myelin homeostasis in the CNS.
Collapse
Affiliation(s)
- Zhengjiang Qian
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Center for Excellence in Brain Science and Intelligence Technology, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Hongchao Li
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Center for Excellence in Brain Science and Intelligence Technology, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Haiyang Yang
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Center for Excellence in Brain Science and Intelligence Technology, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qin Yang
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Center for Excellence in Brain Science and Intelligence Technology, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Zhonghua Lu
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Center for Excellence in Brain Science and Intelligence Technology, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Liping Wang
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Center for Excellence in Brain Science and Intelligence Technology, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Ying Chen
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen 361005, Fujian, China
| | - Xiang Li
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Center for Excellence in Brain Science and Intelligence Technology, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| |
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: 30] [Impact Index Per Article: 7.5] [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
|
Brooks LJ, Clements MP, Burden JJ, Kocher D, Richards L, Devesa SC, Zakka L, Woodberry M, Ellis M, Jaunmuktane Z, Brandner S, Morrison G, Pollard SM, Dirks PB, Marguerat S, Parrinello S. The white matter is a pro-differentiative niche for glioblastoma. Nat Commun 2021; 12:2184. [PMID: 33846316 PMCID: PMC8042097 DOI: 10.1038/s41467-021-22225-w] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 02/23/2021] [Indexed: 02/02/2023] Open
Abstract
Glioblastomas are hierarchically organised tumours driven by glioma stem cells that retain partial differentiation potential. Glioma stem cells are maintained in specialised microenvironments, but whether, or how, they undergo lineage progression outside of these niches remains unclear. Here we identify the white matter as a differentiative niche for glioblastomas with oligodendrocyte lineage competency. Tumour cells in contact with white matter acquire pre-oligodendrocyte fate, resulting in decreased proliferation and invasion. Differentiation is a response to white matter injury, which is caused by tumour infiltration itself in a tumoursuppressive feedback loop. Mechanistically, tumour cell differentiation is driven by selective white matter upregulation of SOX10, a master regulator of normal oligodendrogenesis. SOX10 overexpression or treatment with myelination-promoting agents that upregulate endogenous SOX10, mimic this response, leading to niche-independent pre-oligodendrocyte differentiation and tumour suppression in vivo. Thus, glioblastoma recapitulates an injury response and exploiting this latent programme may offer treatment opportunities for a subset of patients.
Collapse
Affiliation(s)
- Lucy J Brooks
- Samantha Dickson Brain Cancer Unit, UCL Cancer Institute, London, WC1E 6DD, UK
| | - Melanie P Clements
- Samantha Dickson Brain Cancer Unit, UCL Cancer Institute, London, WC1E 6DD, UK
| | - Jemima J Burden
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Daniela Kocher
- Samantha Dickson Brain Cancer Unit, UCL Cancer Institute, London, WC1E 6DD, UK
| | - Luca Richards
- Samantha Dickson Brain Cancer Unit, UCL Cancer Institute, London, WC1E 6DD, UK
| | - Sara Castro Devesa
- Samantha Dickson Brain Cancer Unit, UCL Cancer Institute, London, WC1E 6DD, UK
| | - Leila Zakka
- Samantha Dickson Brain Cancer Unit, UCL Cancer Institute, London, WC1E 6DD, UK
| | - Megan Woodberry
- Samantha Dickson Brain Cancer Unit, UCL Cancer Institute, London, WC1E 6DD, UK
| | - Michael Ellis
- Samantha Dickson Brain Cancer Unit, UCL Cancer Institute, London, WC1E 6DD, UK
| | - Zane Jaunmuktane
- Division of Neuropathology, National Hospital for Neurology and Neurosurgery, University College London NHS Foundation Trust, Queen Square, WC1N 3BG, London, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, WC1N 3BG, London, UK
| | - Sebastian Brandner
- Division of Neuropathology, National Hospital for Neurology and Neurosurgery, University College London NHS Foundation Trust, Queen Square, WC1N 3BG, London, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, WC1N 3BG, London, UK
| | - Gillian Morrison
- MRC Centre for Regenerative Medicine and Edinburgh Cancer Research UK Cancer Centre, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Steven M Pollard
- MRC Centre for Regenerative Medicine and Edinburgh Cancer Research UK Cancer Centre, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Peter B Dirks
- Division of Neurosurgery, Arthur and Sonia Labatt Brain Tumor Research Center, Departments of Surgery and Molecular Genetics, Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
| | - Samuel Marguerat
- MRC London Institute of Medical Sciences, Du Cane Road, London, W12 0NN, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Simona Parrinello
- Samantha Dickson Brain Cancer Unit, UCL Cancer Institute, London, WC1E 6DD, UK.
| |
Collapse
|
17
|
Breton JM, Long KLP, Barraza MK, Perloff OS, Kaufer D. Hormonal Regulation of Oligodendrogenesis II: Implications for Myelin Repair. Biomolecules 2021; 11:290. [PMID: 33669242 PMCID: PMC7919830 DOI: 10.3390/biom11020290] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/10/2021] [Accepted: 02/13/2021] [Indexed: 02/07/2023] Open
Abstract
Alterations in myelin, the protective and insulating sheath surrounding axons, affect brain function, as is evident in demyelinating diseases where the loss of myelin leads to cognitive and motor dysfunction. Recent evidence suggests that changes in myelination, including both hyper- and hypo-myelination, may also play a role in numerous neurological and psychiatric diseases. Protecting myelin and promoting remyelination is thus crucial for a wide range of disorders. Oligodendrocytes (OLs) are the cells that generate myelin, and oligodendrogenesis, the creation of new OLs, continues throughout life and is necessary for myelin plasticity and remyelination. Understanding the regulation of oligodendrogenesis and myelin plasticity within disease contexts is, therefore, critical for the development of novel therapeutic targets. In our companion manuscript, we review literature demonstrating that multiple hormone classes are involved in the regulation of oligodendrogenesis under physiological conditions. The majority of hormones enhance oligodendrogenesis, increasing oligodendrocyte precursor cell differentiation and inducing maturation and myelin production in OLs. Thus, hormonal treatments present a promising route to promote remyelination. Here, we review the literature on hormonal regulation of oligodendrogenesis within the context of disorders. We focus on steroid hormones, including glucocorticoids and sex hormones, peptide hormones such as insulin-like growth factor 1, and thyroid hormones. For each hormone, we describe whether they aid in OL survival, differentiation, or remyelination, and we discuss their mechanisms of action, if known. Several of these hormones have yielded promising results in both animal models and in human conditions; however, a better understanding of hormonal effects, interactions, and their mechanisms will ultimately lead to more targeted therapeutics for myelin repair.
Collapse
Affiliation(s)
- Jocelyn M Breton
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA
| | - Kimberly L P Long
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA
| | - Matthew K Barraza
- Molecular and Cellular Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - Olga S Perloff
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Daniela Kaufer
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA
- Integrative Biology, University of California Berkeley, Berkeley, CA 94720, USA
- Canadian Institute for Advanced Research, Toronto, ON M5G1M1, Canada
| |
Collapse
|
18
|
Diaz J, Gérard X, Emerit MB, Areias J, Geny D, Dégardin J, Simonutti M, Guerquin MJ, Collin T, Viollet C, Billard JM, Métin C, Hubert L, Larti F, Kahrizi K, Jobling R, Agolini E, Shaheen R, Zigler A, Rouiller-Fabre V, Rozet JM, Picaud S, Novelli A, Alameer S, Najmabadi H, Cohn R, Munnich A, Barth M, Lugli L, Alkuraya FS, Blaser S, Gashlan M, Besmond C, Darmon M, Masson J. YIF1B mutations cause a post-natal neurodevelopmental syndrome associated with Golgi and primary cilium alterations. Brain 2021; 143:2911-2928. [PMID: 33103737 DOI: 10.1093/brain/awaa235] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 05/29/2020] [Accepted: 06/13/2020] [Indexed: 12/30/2022] Open
Abstract
Human post-natal neurodevelopmental delay is often associated with cerebral alterations that can lead, by themselves or associated with peripheral deficits, to premature death. Here, we report the clinical features of 10 patients from six independent families with mutations in the autosomal YIF1B gene encoding a ubiquitous protein involved in anterograde traffic from the endoplasmic reticulum to the cell membrane, and in Golgi apparatus morphology. The patients displayed global developmental delay, motor delay, visual deficits with brain MRI evidence of ventricle enlargement, myelination alterations and cerebellar atrophy. A similar profile was observed in the Yif1b knockout (KO) mouse model developed to identify the cellular alterations involved in the clinical defects. In the CNS, mice lacking Yif1b displayed neuronal reduction, altered myelination of the motor cortex, cerebellar atrophy, enlargement of the ventricles, and subcellular alterations of endoplasmic reticulum and Golgi apparatus compartments. Remarkably, although YIF1B was not detected in primary cilia, biallelic YIF1B mutations caused primary cilia abnormalities in skin fibroblasts from both patients and Yif1b-KO mice, and in ciliary architectural components in the Yif1b-KO brain. Consequently, our findings identify YIF1B as an essential gene in early post-natal development in human, and provide a new genetic target that should be tested in patients developing a neurodevelopmental delay during the first year of life. Thus, our work is the first description of a functional deficit linking Golgipathies and ciliopathies, diseases so far associated exclusively to mutations in genes coding for proteins expressed within the primary cilium or related ultrastructures. We therefore propose that these pathologies should be considered as belonging to a larger class of neurodevelopmental diseases depending on proteins involved in the trafficking of proteins towards specific cell membrane compartments.
Collapse
Affiliation(s)
- Jorge Diaz
- INSERM UMR894, Center for Psychiatry and Neuroscience, Paris F-75014, Université Paris Descartes, Sorbonne Paris Cité - Paris 5, France
| | - Xavier Gérard
- INSERM UMR-S1163 Imagine Institute for Genetic Diseases, Paris Descartes-Sorbonne Paris Cité University, France
| | - Michel-Boris Emerit
- INSERM UMR894, Center for Psychiatry and Neuroscience, Paris F-75014, Université Paris Descartes, Sorbonne Paris Cité - Paris 5, France
| | - Julie Areias
- INSERM UMR894, Center for Psychiatry and Neuroscience, Paris F-75014, Université Paris Descartes, Sorbonne Paris Cité - Paris 5, France
| | - David Geny
- INSERM UMR894, Center for Psychiatry and Neuroscience, Paris F-75014, Université Paris Descartes, Sorbonne Paris Cité - Paris 5, France
| | - Julie Dégardin
- INSERM UMR-S968, Institut de la vision, Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, Paris F-75012, Université Pierre et Marie Curie, France
| | - Manuel Simonutti
- INSERM UMR-S968, Institut de la vision, Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, Paris F-75012, Université Pierre et Marie Curie, France
| | | | - Thibault Collin
- Saint Pères Paris Institute for the Neurosciences CNRS - UMR 8003 Université de Paris, Paris 75006, France
| | - Cécile Viollet
- INSERM UMR894, Center for Psychiatry and Neuroscience, Paris F-75014, Université Paris Descartes, Sorbonne Paris Cité - Paris 5, France
| | - Jean-Marie Billard
- INSERM UMR894, Center for Psychiatry and Neuroscience, Paris F-75014, Université Paris Descartes, Sorbonne Paris Cité - Paris 5, France
| | - Christine Métin
- INSERM, UMR-S1270, Institut du Fer à Moulin, Sorbonne Université, Paris F-75005, France
| | - Laurence Hubert
- INSERM UMR-S1163 Imagine Institute for Genetic Diseases, Paris Descartes-Sorbonne Paris Cité University, France
| | - Farzaneh Larti
- University of Social Welfare and Rehabilitation Sciences, Genetics Research Center, Tehran 19834, Iran
| | - Kimia Kahrizi
- University of Social Welfare and Rehabilitation Sciences, Genetics Research Center, Tehran 19834, Iran
| | - Rebekah Jobling
- The Hospital for Sick Children, Molecular Genetics, Toronto, Canada
| | - Emanuele Agolini
- Laboratory of Medical Genetics, Bambino Gesù Children's Hospital, 00146 Rome, Italy
| | - Ranad Shaheen
- King Faisal Specialist Hospital and Research Center, Developmental Genetics Unit, Riyadh 11211, Saudi Arabia
| | | | | | - Jean-Michel Rozet
- INSERM UMR-S1163 Imagine Institute for Genetic Diseases, Paris Descartes-Sorbonne Paris Cité University, France
| | - Serge Picaud
- INSERM UMR-S968, Institut de la vision, Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, Paris F-75012, Université Pierre et Marie Curie, France
| | - Antonio Novelli
- Laboratory of Medical Genetics, Bambino Gesù Children's Hospital, 00146 Rome, Italy
| | - Seham Alameer
- Department of Pediatrics, King Khaled National Guard Hospital, King Abdulaziz Medical City, Jeddah, Saudi Arabia
| | - Hossein Najmabadi
- University of Social Welfare and Rehabilitation Sciences, Genetics Research Center, Tehran 19834, Iran
| | - Ronald Cohn
- The Hospital for Sick Children, Molecular Genetics, Toronto, Canada
| | - Arnold Munnich
- INSERM UMR-S1163 Imagine Institute for Genetic Diseases, Paris Descartes-Sorbonne Paris Cité University, France
| | | | - Licia Lugli
- Division of Neonatal Intensive Care Unit, Department of Pediatrics, University Hospital, 41125 Modena, Italy
| | - Fowzan S Alkuraya
- King Faisal Specialist Hospital and Research Center, Developmental Genetics Unit, Riyadh 11211, Saudi Arabia
| | - Susan Blaser
- Division of Neuroradiology, The Hospital for Sick Children, University of Toronto, Toronto, Canada
| | - Maha Gashlan
- King Faisal Specialist Hospital and Research Center, Developmental Genetics Unit, Riyadh 11211, Saudi Arabia
| | - Claude Besmond
- INSERM UMR-S1163 Imagine Institute for Genetic Diseases, Paris Descartes-Sorbonne Paris Cité University, France
| | - Michèle Darmon
- INSERM UMR894, Center for Psychiatry and Neuroscience, Paris F-75014, Université Paris Descartes, Sorbonne Paris Cité - Paris 5, France.,INSERM, UMR-S1270, Institut du Fer à Moulin, Sorbonne Université, Paris F-75005, France
| | - Justine Masson
- INSERM UMR894, Center for Psychiatry and Neuroscience, Paris F-75014, Université Paris Descartes, Sorbonne Paris Cité - Paris 5, France.,INSERM, UMR-S1270, Institut du Fer à Moulin, Sorbonne Université, Paris F-75005, France
| |
Collapse
|
19
|
Long KLP, Breton JM, Barraza MK, Perloff OS, Kaufer D. Hormonal Regulation of Oligodendrogenesis I: Effects across the Lifespan. Biomolecules 2021; 11:biom11020283. [PMID: 33672939 PMCID: PMC7918364 DOI: 10.3390/biom11020283] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/04/2021] [Accepted: 02/06/2021] [Indexed: 02/07/2023] Open
Abstract
The brain’s capacity to respond to changing environments via hormonal signaling is critical to fine-tuned function. An emerging body of literature highlights a role for myelin plasticity as a prominent type of experience-dependent plasticity in the adult brain. Myelin plasticity is driven by oligodendrocytes (OLs) and their precursor cells (OPCs). OPC differentiation regulates the trajectory of myelin production throughout development, and importantly, OPCs maintain the ability to proliferate and generate new OLs throughout adulthood. The process of oligodendrogenesis, the creation of new OLs, can be dramatically influenced during early development and in adulthood by internal and environmental conditions such as hormones. Here, we review the current literature describing hormonal regulation of oligodendrogenesis within physiological conditions, focusing on several classes of hormones: steroid, peptide, and thyroid hormones. We discuss hormonal regulation at each stage of oligodendrogenesis and describe mechanisms of action, where known. Overall, the majority of hormones enhance oligodendrogenesis, increasing OPC differentiation and inducing maturation and myelin production in OLs. The mechanisms underlying these processes vary for each hormone but may ultimately converge upon common signaling pathways, mediated by specific receptors expressed across the OL lineage. However, not all of the mechanisms have been fully elucidated, and here, we note the remaining gaps in the literature, including the complex interactions between hormonal systems and with the immune system. In the companion manuscript in this issue, we discuss the implications of hormonal regulation of oligodendrogenesis for neurological and psychiatric disorders characterized by white matter loss. Ultimately, a better understanding of the fundamental mechanisms of hormonal regulation of oligodendrogenesis across the entire lifespan, especially in vivo, will progress both basic and translational research.
Collapse
Affiliation(s)
- Kimberly L. P. Long
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA 94720, USA; (J.M.B.); (D.K.)
- Correspondence:
| | - Jocelyn M. Breton
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA 94720, USA; (J.M.B.); (D.K.)
| | - Matthew K. Barraza
- Department of Molecular and Cellular Biology, University of California, Berkeley, CA 94720, USA;
| | - Olga S. Perloff
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA 94143, USA;
| | - Daniela Kaufer
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA 94720, USA; (J.M.B.); (D.K.)
- Department of Integrative Biology, University of California, Berkeley, CA 94720, USA
- Canadian Institute for Advanced Research, Toronto, ON M5G 1M1, Canada
| |
Collapse
|
20
|
Ganesh RA, Venkataraman K, Sirdeshmukh R. GPR56: An adhesion GPCR involved in brain development, neurological disorders and cancer. Brain Res 2020; 1747:147055. [PMID: 32798453 DOI: 10.1016/j.brainres.2020.147055] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 07/04/2020] [Accepted: 08/11/2020] [Indexed: 12/15/2022]
Abstract
GPR56/ADGRG1 is a member of the adhesion G-protein coupled receptor (aGPCR) family and one of the important players in the normal development of the brain. It plays a pivotal role in the diverse neurobiological processes, including cortical formation, oligodendrocyte development, and myelination. Mutations in GPR56 are known to cause brain malformation, myelination defects and are also implied in many cancers, including brain tumors. Since its identification almost two decades ago, GPR56 has emerged from an orphaned and uncharacterized GPCR to an increasingly well studied receptor. Yet, much needs to be understood about GPR56, both in terms of its molecular interactions and biological functions that may be relevant in normal health and disease. The review is focussed on the recent available knowledge of GPR56, which would give useful insights into its known and potential roles in the human brain, neurological disorders, and brain tumors like glioblastoma.
Collapse
Affiliation(s)
- Raksha A Ganesh
- Mazumdar Shaw Center for Translational Research, Narayana Health, Bangalore 560099, India; Center for Bio-Separation Technology, Vellore Institute of Technology, Vellore 632104, India
| | - Krishnan Venkataraman
- Center for Bio-Separation Technology, Vellore Institute of Technology, Vellore 632104, India
| | - Ravi Sirdeshmukh
- Mazumdar Shaw Center for Translational Research, Narayana Health, Bangalore 560099, India; Institute of Bioinformatics, International Tech Park, Bangalore 560066, India; Manipal Academy of Higher Education, Manipal 576104, India.
| |
Collapse
|
21
|
Agolini E, Cherchi C, Bellacchio E, Martinelli D, Cocciadiferro D, Cutrera R, Chiarini Testa MB, Barone C, Bianca S, Novelli A. Expanding the clinical and molecular spectrum of lethal congenital contracture syndrome 8 associated with biallelic variants of ADCY6. Clin Genet 2020; 97:649-654. [PMID: 31846058 DOI: 10.1111/cge.13691] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 12/06/2019] [Accepted: 12/11/2019] [Indexed: 11/26/2022]
Abstract
Arthrogryposis multiplex congenita (AMC) is defined as congenital, non-progressive contractures in more than two joints and in multiple body areas, resulting from reduced fetal mobility. So far, more than 400 causative genes for AMC have been identified. Some isolated AMC phenotypes arise as a result of mutations in genes encoding components required for motor neuron structure, function, and myelination, as in the case of ADCY6 encoding the enzyme adenylyl cyclase type 6. ADCY6 inactivation, due to biallelic variants, have been previously associated with the lethal congenital contracture syndrome 8 (LCCS8). So far, only four LCCS8 patients, from two families, have been reported. Here, we describe a new patient affected by a severe form of AMC, harboring two novel compound heterozygous variants in ADCY6. Our findings expand the clinical and mutational spectrum of LCCS8, showing a possible correlation between the impact of the ADCY6 missense variants reported to date, predicted by molecular modeling, and the severity of the phenotype.
Collapse
Affiliation(s)
- Emanuele Agolini
- Laboratory of Medical Genetics Unit, Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Claudio Cherchi
- Respiratory Unit, Academic Department of Pediatrics, Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Emanuele Bellacchio
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Diego Martinelli
- Division of Metabolism, Department of Pediatric Specialties, Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Dario Cocciadiferro
- Laboratory of Medical Genetics Unit, Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Renato Cutrera
- Respiratory Unit, Academic Department of Pediatrics, Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Maria B Chiarini Testa
- Respiratory Unit, Academic Department of Pediatrics, Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Chiara Barone
- Medical Genetics, Referral Centre for Rare Genetic Diseases, ARNAS Garibaldi, Catania, Italy
| | - Sebastiano Bianca
- Medical Genetics, Referral Centre for Rare Genetic Diseases, ARNAS Garibaldi, Catania, Italy
| | - Antonio Novelli
- Laboratory of Medical Genetics Unit, Ospedale Pediatrico Bambino Gesù, Rome, Italy
| |
Collapse
|
22
|
Theoretical and Experimental Approaches Aimed at Drug Design Targeting Neurodegenerative Diseases. Processes (Basel) 2019. [DOI: 10.3390/pr7120940] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
In recent years, green chemistry has been strengthening, showing how basic and applied sciences advance globally, protecting the environment and human health. A clear example of this evolution is the synergy that now exists between theoretical and computational methods to design new drugs in the most efficient possible way, using the minimum of reagents and obtaining the maximum yield. The development of compounds with potential therapeutic activity against multiple targets associated with neurodegenerative diseases/disorders (NDD) such as Alzheimer’s disease is a hot topic in medical chemistry, where different scientists from various disciplines collaborate to find safe, active, and effective drugs. NDD are a public health problem, affecting mainly the population over 60 years old. To generate significant progress in the pharmacological treatment of NDD, it is necessary to employ different experimental strategies of green chemistry, medical chemistry, and molecular biology, coupled with computational and theoretical approaches such as molecular simulations and chemoinformatics, all framed in the rational drug design targeting NDD. Here, we review how green chemistry and computational approaches have been used to develop new compounds with the potential application against NDD, as well as the challenges and new directions of the drug development multidisciplinary process.
Collapse
|
23
|
Bradley EC, Cunningham RL, Wilde C, Morgan RK, Klug EA, Letcher SM, Schöneberg T, Monk KR, Liebscher I, Petersen SC. In vivo identification of small molecules mediating Gpr126/Adgrg6 signaling during Schwann cell development. Ann N Y Acad Sci 2019; 1456:44-63. [PMID: 31529518 PMCID: PMC7189964 DOI: 10.1111/nyas.14233] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 07/30/2019] [Accepted: 08/26/2019] [Indexed: 12/13/2022]
Abstract
Gpr126/Adgrg6, an adhesion family G protein-coupled receptor (aGPCR), is required for the development of myelinating Schwann cells in the peripheral nervous system. Myelin supports and insulates vertebrate axons to permit rapid signal propagation throughout the nervous system. In mammals and zebrafish, mutations in Gpr126 arrest Schwann cells at early developmental stages. We exploited the optical and pharmacological tractability of larval zebrafish to uncover drugs that mediate myelination by activating Gpr126 or functioning in parallel. Using a fluorescent marker of mature myelinating glia (Tg[mbp:EGFP-CAAX]), we screened hypomorphic gpr126 mutant larvae for restoration of myelin basic protein (mbp) expression along peripheral nerves following small molecule treatment. Our screens identified five compounds sufficient to promote mbp expression in gpr126 hypomorphs. Using an allelic series of gpr126 mutants, we parsed the ability of small molecules to restore mbp, suggesting differences in drug efficacy dependent on Schwann cell developmental state. Finally, we identify apomorphine hydrochloride as a direct small molecule activator of Gpr126 using combined in vivo/in vitro assays and show that aporphine class compounds promote Schwann cell development in vivo. Our results demonstrate the utility of in vivo screening for aGPCR modulators and identify small molecules that interact with the gpr126-mediated myelination program.
Collapse
Affiliation(s)
| | - Rebecca L. Cunningham
- Department of Developmental Biology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Caroline Wilde
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Rory K. Morgan
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA
| | - Emma A. Klug
- Department of Neuroscience, Kenyon College, Gambier, OH, USA
| | | | - Torsten Schöneberg
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Kelly R. Monk
- Department of Developmental Biology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA
| | - Ines Liebscher
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Sarah C. Petersen
- Department of Neuroscience, Kenyon College, Gambier, OH, USA
- Department of Developmental Biology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| |
Collapse
|
24
|
Jessen KR, Arthur-Farraj P. Repair Schwann cell update: Adaptive reprogramming, EMT, and stemness in regenerating nerves. Glia 2019; 67:421-437. [PMID: 30632639 DOI: 10.1002/glia.23532] [Citation(s) in RCA: 245] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 08/20/2018] [Accepted: 09/05/2018] [Indexed: 12/16/2022]
Abstract
Schwann cells respond to nerve injury by cellular reprogramming that generates cells specialized for promoting regeneration and repair. These repair cells clear redundant myelin, attract macrophages, support survival of damaged neurons, encourage axonal growth, and guide axons back to their targets. There are interesting parallels between this response and that found in other tissues. At the cellular level, many other tissues also react to injury by cellular reprogramming, generating cells specialized to promote tissue homeostasis and repair. And at the molecular level, a common feature possessed by Schwann cells and many other cells is the injury-induced activation of genes associated with epithelial-mesenchymal transitions and stemness, differentiation states that are linked to cellular plasticity and that help injury-induced tissue remodeling. The number of signaling systems regulating Schwann cell plasticity is rapidly increasing. Importantly, this includes mechanisms that are crucial for the generation of functional repair Schwann cells and nerve regeneration, although they have no or a minor role elsewhere in the Schwann cell lineage. This encourages the view that selective tools can be developed to control these particular cells, amplify their repair supportive functions and prevent their deterioration. In this review, we discuss the emerging similarities between the injury response seen in nerves and in other tissues and survey the transcription factors, epigenetic mechanisms, and signaling cascades that control repair Schwann cells, with emphasis on systems that selectively regulate the Schwann cell injury response.
Collapse
Affiliation(s)
- Kristjan R Jessen
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Peter Arthur-Farraj
- John Van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
25
|
InanlooRahatloo K, Peymani F, Kahrizi K, Najmabadi H. Whole-Transcriptome Analysis Reveals Dysregulation of Actin-Cytoskeleton Pathway in Intellectual Disability Patients. Neuroscience 2019; 404:423-444. [PMID: 30742961 DOI: 10.1016/j.neuroscience.2019.01.029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Revised: 01/07/2019] [Accepted: 01/17/2019] [Indexed: 12/14/2022]
Abstract
A significant level of genetic heterogeneity has been demonstrated in intellectual disability (ID). More than 700 genes have been identified in ID patients. To identify molecular pathways underlying this heterogeneity, we applied whole-transcriptome analysis using RNA-Seq in consanguineous families with ID. Significant changes in expression of genes related to neuronal and actin cytoskeletal functions were observed in all the ID families. Remarkably, we found a significant down-regulation of SHTN1 gene and up-regulation of FGFR2 gene in all ID patients. FGFR2, but not SHTN1, was previously reported as an ID causing gene. Detailed gene ontology analyses identified pathways linked to tyrosine protein kinase, actin cytoskeleton, and axonogenesis to be affected in ID patients. The findings reported here provide new insights into the candidate genes and molecular pathways underling ID and highlight the key role of actin cytoskeleton in etiology of ID.
Collapse
Affiliation(s)
- Kolsoum InanlooRahatloo
- School of Biology, College of Science, University of Tehran, Tehran, Iran; Genetic Research Center, University of social welfare and Rehabilitation Sciences, Tehran, Iran.
| | - Fatemeh Peymani
- Genetic Research Center, University of social welfare and Rehabilitation Sciences, Tehran, Iran
| | - Kimia Kahrizi
- Genetic Research Center, University of social welfare and Rehabilitation Sciences, Tehran, Iran
| | - Hossein Najmabadi
- Genetic Research Center, University of social welfare and Rehabilitation Sciences, Tehran, Iran.
| |
Collapse
|
26
|
Peng WT, Sun WY, Li XR, Sun JC, Du JJ, Wei W. Emerging Roles of G Protein-Coupled Receptors in Hepatocellular Carcinoma. Int J Mol Sci 2018; 19:ijms19051366. [PMID: 29734668 PMCID: PMC5983678 DOI: 10.3390/ijms19051366] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Revised: 04/24/2018] [Accepted: 04/26/2018] [Indexed: 12/13/2022] Open
Abstract
Among a great variety of cell surface receptors, the largest superfamily is G protein-coupled receptors (GPCRs), also known as seven-transmembrane domain receptors. GPCRs can modulate diverse signal-transduction pathways through G protein-dependent or independent pathways which involve β-arrestins, G protein receptor kinases (GRKs), ion channels, or Src kinases under physiological and pathological conditions. Recent studies have revealed the crucial role of GPCRs in the tumorigenesis and the development of cancer metastasis. We will sum up the functions of GPCRs—particularly those coupled to chemokines, prostaglandin, lysophosphatidic acid, endothelin, catecholamine, and angiotensin—in the proliferation, invasion, metastasis, and angiogenesis of hepatoma cells and the development of hepatocellular carcinoma (HCC) in this review. We also highlight the potential avenues of GPCR-based therapeutics for HCC.
Collapse
Affiliation(s)
- Wen-Ting Peng
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei 230032, China.
- Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
- Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei 230032, China.
| | - Wu-Yi Sun
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei 230032, China.
- Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
- Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei 230032, China.
| | - Xin-Ran Li
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei 230032, China.
- Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
- Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei 230032, China.
| | - Jia-Chang Sun
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei 230032, China.
- Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
- Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei 230032, China.
| | - Jia-Jia Du
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei 230032, China.
- Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
- Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei 230032, China.
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei 230032, China.
- Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
- Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei 230032, China.
| |
Collapse
|
27
|
Merten N, Fischer J, Simon K, Zhang L, Schröder R, Peters L, Letombe AG, Hennen S, Schrage R, Bödefeld T, Vermeiren C, Gillard M, Mohr K, Lu QR, Brüstle O, Gomeza J, Kostenis E. Repurposing HAMI3379 to Block GPR17 and Promote Rodent and Human Oligodendrocyte Differentiation. Cell Chem Biol 2018; 25:775-786.e5. [PMID: 29706593 DOI: 10.1016/j.chembiol.2018.03.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 02/11/2018] [Accepted: 03/23/2018] [Indexed: 12/22/2022]
Abstract
Identification of additional uses for existing drugs is a hot topic in drug discovery and a viable alternative to de novo drug development. HAMI3379 is known as an antagonist of the cysteinyl-leukotriene CysLT2 receptor, and was initially developed to treat cardiovascular and inflammatory disorders. In our study we identified HAMI3379 as an antagonist of the orphan G protein-coupled receptor GPR17. HAMI3379 inhibits signaling of recombinant human, rat, and mouse GPR17 across various cellular backgrounds, and of endogenous GPR17 in primary rodent oligodendrocytes. GPR17 blockade by HAMI3379 enhanced maturation of primary rat and mouse oligodendrocytes, but was without effect in oligodendrocytes from GPR17 knockout mice. In human oligodendrocytes prepared from inducible pluripotent stem cells, GPR17 is expressed and its activation impaired oligodendrocyte differentiation. HAMI3379, conversely, efficiently favored human oligodendrocyte differentiation. We propose that HAMI3379 holds promise for pharmacological exploitation of orphan GPR17 to enhance regenerative strategies for the promotion of remyelination in patients.
Collapse
Affiliation(s)
- Nicole Merten
- Molecular, Cellular and Pharmacobiology Section, Institute of Pharmaceutical Biology, University of Bonn, 53115 Bonn, Germany
| | - Julia Fischer
- Institute of Reconstructive Neurobiology, LIFE&BRAIN Center, University of Bonn, 53105 Bonn, Germany
| | - Katharina Simon
- Molecular, Cellular and Pharmacobiology Section, Institute of Pharmaceutical Biology, University of Bonn, 53115 Bonn, Germany
| | - Liguo Zhang
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Ralf Schröder
- Molecular, Cellular and Pharmacobiology Section, Institute of Pharmaceutical Biology, University of Bonn, 53115 Bonn, Germany
| | - Lucas Peters
- Molecular, Cellular and Pharmacobiology Section, Institute of Pharmaceutical Biology, University of Bonn, 53115 Bonn, Germany
| | | | - Stephanie Hennen
- Molecular, Cellular and Pharmacobiology Section, Institute of Pharmaceutical Biology, University of Bonn, 53115 Bonn, Germany
| | - Ramona Schrage
- UCB Biopharma, CNS Research, 1420 Braine-l'Alleud, Belgium
| | - Theresa Bödefeld
- Pharmacology and Toxicology Section, Institute of Pharmacy, University of Bonn, 53115 Bonn, Germany
| | | | - Michel Gillard
- UCB Biopharma, CNS Research, 1420 Braine-l'Alleud, Belgium
| | - Klaus Mohr
- Pharmacology and Toxicology Section, Institute of Pharmacy, University of Bonn, 53115 Bonn, Germany
| | - Qing Richard Lu
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Oliver Brüstle
- Institute of Reconstructive Neurobiology, LIFE&BRAIN Center, University of Bonn, 53105 Bonn, Germany
| | - Jesus Gomeza
- Molecular, Cellular and Pharmacobiology Section, Institute of Pharmaceutical Biology, University of Bonn, 53115 Bonn, Germany.
| | - Evi Kostenis
- Molecular, Cellular and Pharmacobiology Section, Institute of Pharmaceutical Biology, University of Bonn, 53115 Bonn, Germany.
| |
Collapse
|
28
|
The G protein-coupled receptors deorphanization landscape. Biochem Pharmacol 2018; 153:62-74. [PMID: 29454621 DOI: 10.1016/j.bcp.2018.02.016] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 02/13/2018] [Indexed: 12/14/2022]
Abstract
G protein-coupled receptors (GPCRs) are usually highlighted as being both the largest family of membrane proteins and the most productive source of drug targets. However, most of the GPCRs are understudied and hence cannot be used immediately for innovative therapeutic strategies. Besides, there are still around 100 orphan receptors, with no described endogenous ligand and no clearly defined function. The race to discover new ligands for these elusive receptors seems to be less intense than before. Here, we present an update of the various strategies employed to assign a function to these receptors and to discover new ligands. We focus on the recent advances in the identification of endogenous ligands with a detailed description of newly deorphanized receptors. Replication being a key parameter in these endeavors, we also discuss the latest controversies about problematic ligand-receptor pairings. In this context, we propose several recommendations in order to strengthen the reporting of new ligand-receptor pairs.
Collapse
|
29
|
García-León JA, Kumar M, Boon R, Chau D, One J, Wolfs E, Eggermont K, Berckmans P, Gunhanlar N, de Vrij F, Lendemeijer B, Pavie B, Corthout N, Kushner SA, Dávila JC, Lambrichts I, Hu WS, Verfaillie CM. SOX10 Single Transcription Factor-Based Fast and Efficient Generation of Oligodendrocytes from Human Pluripotent Stem Cells. Stem Cell Reports 2018; 10:655-672. [PMID: 29337119 PMCID: PMC5830935 DOI: 10.1016/j.stemcr.2017.12.014] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 12/15/2017] [Accepted: 12/15/2017] [Indexed: 02/06/2023] Open
Abstract
Scarce access to primary samples and lack of efficient protocols to generate oligodendrocytes (OLs) from human pluripotent stem cells (hPSCs) are hampering our understanding of OL biology and the development of novel therapies. Here, we demonstrate that overexpression of the transcription factor SOX10 is sufficient to generate surface antigen O4-positive (O4+) and myelin basic protein-positive OLs from hPSCs in only 22 days, including from patients with multiple sclerosis or amyotrophic lateral sclerosis. The SOX10-induced O4+ population resembles primary human OLs at the transcriptome level and can myelinate neurons in vivo. Using in vitro OL-neuron co-cultures, myelination of neurons by OLs can also be demonstrated, which can be adapted to a high-throughput screening format to test the response of pro-myelinating drugs. In conclusion, we provide an approach to generate OLs in a very rapid and efficient manner, which can be used for disease modeling, drug discovery efforts, and potentially for therapeutic OL transplantation. SOX10 is sufficient to generate myelinating human OLs from hPSCs in only 22 days SOX10-induced OLs resemble primary human OLs at the transcriptome level The methodology allows efficient generation of OLs from MS and ALS patients OL-neuron co-cultures respond to myelinating drugs in a high-throughput setting
Collapse
Affiliation(s)
- Juan Antonio García-León
- Department of Development and Regeneration, Stem Cell Biology and Embryology, KU Leuven Stem Cell Institute, Herestraat 49, Onderwijs en Navorsing 4, Box 804, 3000 Leuven, Belgium.
| | - Manoj Kumar
- Department of Development and Regeneration, Stem Cell Biology and Embryology, KU Leuven Stem Cell Institute, Herestraat 49, Onderwijs en Navorsing 4, Box 804, 3000 Leuven, Belgium
| | - Ruben Boon
- Department of Development and Regeneration, Stem Cell Biology and Embryology, KU Leuven Stem Cell Institute, Herestraat 49, Onderwijs en Navorsing 4, Box 804, 3000 Leuven, Belgium
| | - David Chau
- Department of Chemical Engineering and Materials Science, University of Minnesota, 421 Washington Avenue Southeast, Minneapolis, MN 55455, USA
| | - Jennifer One
- Department of Chemical Engineering and Materials Science, University of Minnesota, 421 Washington Avenue Southeast, Minneapolis, MN 55455, USA
| | - Esther Wolfs
- Laboratory of Morphology, Biomedical Research Institute (BIOMED), Hasselt University, Agoralaan, Building C, 3590 Diepenbeek, Belgium
| | - Kristel Eggermont
- Department of Development and Regeneration, Stem Cell Biology and Embryology, KU Leuven Stem Cell Institute, Herestraat 49, Onderwijs en Navorsing 4, Box 804, 3000 Leuven, Belgium
| | - Pieter Berckmans
- Department of Development and Regeneration, Stem Cell Biology and Embryology, KU Leuven Stem Cell Institute, Herestraat 49, Onderwijs en Navorsing 4, Box 804, 3000 Leuven, Belgium
| | - Nilhan Gunhanlar
- Department of Psychiatry, Erasmus MC, 3015 CE Rotterdam, the Netherlands
| | - Femke de Vrij
- Department of Psychiatry, Erasmus MC, 3015 CE Rotterdam, the Netherlands
| | - Bas Lendemeijer
- Department of Psychiatry, Erasmus MC, 3015 CE Rotterdam, the Netherlands
| | - Benjamin Pavie
- VIB Center for Brain and Disease Research, VIB-Leuven, 3000 Leuven, Belgium; VIB Bio Imaging Core, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Nikky Corthout
- VIB Center for Brain and Disease Research, VIB-Leuven, 3000 Leuven, Belgium; VIB Bio Imaging Core, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Steven A Kushner
- Department of Psychiatry, Erasmus MC, 3015 CE Rotterdam, the Netherlands
| | - José Carlos Dávila
- Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, Research Biomedical Institute of Málaga (IBIMA), University of Málaga, Campus de Teatinos s/n, 29010 Málaga, Spain; Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Ivo Lambrichts
- Laboratory of Morphology, Biomedical Research Institute (BIOMED), Hasselt University, Agoralaan, Building C, 3590 Diepenbeek, Belgium
| | - Wei-Shou Hu
- Department of Chemical Engineering and Materials Science, University of Minnesota, 421 Washington Avenue Southeast, Minneapolis, MN 55455, USA
| | - Catherine M Verfaillie
- Department of Development and Regeneration, Stem Cell Biology and Embryology, KU Leuven Stem Cell Institute, Herestraat 49, Onderwijs en Navorsing 4, Box 804, 3000 Leuven, Belgium.
| |
Collapse
|
30
|
Kiepura AJ, Kochański A. Charcot-Marie-Tooth type 1A drug therapies: role of adenylyl cyclase activity and G-protein coupled receptors in disease pathomechanism. Acta Neurobiol Exp (Wars) 2018. [DOI: 10.21307/ane-2018-018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
31
|
Cole KLH, Early JJ, Lyons DA. Drug discovery for remyelination and treatment of MS. Glia 2017; 65:1565-1589. [PMID: 28618073 DOI: 10.1002/glia.23166] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 04/20/2017] [Accepted: 04/24/2017] [Indexed: 12/19/2022]
Abstract
Glia constitute the majority of the cells in our nervous system, yet there are currently no drugs that target glia for the treatment of disease. Given ongoing discoveries of the many roles of glia in numerous diseases of the nervous system, this is likely to change in years to come. Here we focus on the possibility that targeting the oligodendrocyte lineage to promote regeneration of myelin (remyelination) represents a therapeutic strategy for the treatment of the demyelinating disease multiple sclerosis, MS. We discuss how hypothesis driven studies have identified multiple targets and pathways that can be manipulated to promote remyelination in vivo, and how this work has led to the first ever remyelination clinical trials. We also highlight how recent chemical discovery screens have identified a host of small molecule compounds that promote oligodendrocyte differentiation in vitro. Some of these compounds have also been shown to promote myelin regeneration in vivo, with one already being trialled in humans. Promoting oligodendrocyte differentiation and remyelination represents just one potential strategy for the treatment of MS. The pathology of MS is complex, and its complete amelioration may require targeting multiple biological processes in parallel. Therefore, we present an overview of new technologies and models for phenotypic analyses and screening that can be exploited to study complex cell-cell interactions in in vitro and in vivo systems. Such technological platforms will provide insight into fundamental mechanisms and increase capacities for drug-discovery of relevance to glia and currently intractable disorders of the CNS.
Collapse
Affiliation(s)
- Katy L H Cole
- Centre for Neuroregeneration, MS Society Centre for Translational Research, Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, EH16 4SB, United Kingdom
| | - Jason J Early
- Centre for Neuroregeneration, MS Society Centre for Translational Research, Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, EH16 4SB, United Kingdom
| | - David A Lyons
- Centre for Neuroregeneration, MS Society Centre for Translational Research, Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, EH16 4SB, United Kingdom
| |
Collapse
|
32
|
Klingseisen A, Lyons DA. Axonal Regulation of Central Nervous System Myelination: Structure and Function. Neuroscientist 2017; 24:7-21. [PMID: 28397586 DOI: 10.1177/1073858417703030] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Approximately half of the human brain consists of myelinated axons. Central nervous system (CNS) myelin is made by oligodendrocytes and is essential for nervous system formation, health, and function. Once thought simply as a static insulator that facilitated rapid impulse conduction, myelin is now known to be made and remodeled in to adult life. Oligodendrocytes have a remarkable capacity to differentiate by default, but many aspects of their development can be influenced by axons. However, how axons and oligodendrocytes interact and cooperate to regulate myelination in the CNS remains unclear. Here, we review recent advances in our understanding of how such interactions generate the complexity of myelination known to exist in vivo. We highlight intriguing results that indicate that the cross-sectional size of an axon alone may regulate myelination to a surprising degree. We also review new studies, which have highlighted diversity in the myelination of axons of different neuronal subtypes and circuits, and structure-function relationships, which suggest that myelinated axons can be exquisitely fine-tuned to mediate precise conduction needs. We also discuss recent advances in our understanding of how neuronal activity regulates CNS myelination, and aim to provide an integrated overview of how axon-oligodendrocyte interactions sculpt neuronal circuit structure and function.
Collapse
Affiliation(s)
- Anna Klingseisen
- 1 Centre for Neuroregeneration, University of Edinburgh, Edinburgh, UK
| | - David A Lyons
- 1 Centre for Neuroregeneration, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
33
|
Boerboom A, Dion V, Chariot A, Franzen R. Molecular Mechanisms Involved in Schwann Cell Plasticity. Front Mol Neurosci 2017; 10:38. [PMID: 28261057 PMCID: PMC5314106 DOI: 10.3389/fnmol.2017.00038] [Citation(s) in RCA: 139] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 01/31/2017] [Indexed: 01/09/2023] Open
Abstract
Schwann cell incredible plasticity is a hallmark of the utmost importance following nerve damage or in demyelinating neuropathies. After injury, Schwann cells undergo dedifferentiation before redifferentiating to promote nerve regeneration and complete functional recovery. This review updates and discusses the molecular mechanisms involved in the negative regulation of myelination as well as in the reprogramming of Schwann cells taking place early following nerve lesion to support repair. Significant advance has been made on signaling pathways and molecular components that regulate SC regenerative properties. These include for instance transcriptional regulators such as c-Jun or Notch, the MAPK and the Nrg1/ErbB2/3 pathways. This comprehensive overview ends with some therapeutical applications targeting factors that control Schwann cell plasticity and highlights the need to carefully modulate and balance this capacity to drive nerve repair.
Collapse
Affiliation(s)
| | - Valérie Dion
- GIGA-Neurosciences, University of Liège Liège, Belgium
| | - Alain Chariot
- GIGA-Molecular Biology of Diseases, University of LiègeLiège, Belgium; Walloon Excellence in Lifesciences and Biotechnology (WELBIO)Wavre, Belgium
| | | |
Collapse
|