1
|
Salzer J, Feltri ML, Jacob C. Schwann Cell Development and Myelination. Cold Spring Harb Perspect Biol 2024; 16:a041360. [PMID: 38503507 PMCID: PMC11368196 DOI: 10.1101/cshperspect.a041360] [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
Glial cells in the peripheral nervous system (PNS), which arise from the neural crest, include axon-associated Schwann cells (SCs) in nerves, synapse-associated SCs at the neuromuscular junction, enteric glia, perikaryon-associated satellite cells in ganglia, and boundary cap cells at the border between the central nervous system (CNS) and the PNS. Here, we focus on axon-associated SCs. These SCs progress through a series of formative stages, which culminate in the generation of myelinating SCs that wrap large-caliber axons and of nonmyelinating (Remak) SCs that enclose multiple, small-caliber axons. In this work, we describe SC development, extrinsic signals from the axon and extracellular matrix (ECM) and the intracellular signaling pathways they activate that regulate SC development, and the morphogenesis and organization of myelinating SCs and the myelin sheath. We review the impact of SCs on the biology and integrity of axons and their emerging role in regulating peripheral nerve architecture. Finally, we explain how transcription and epigenetic factors control and fine-tune SC development and myelination.
Collapse
Affiliation(s)
- James Salzer
- Neuroscience Institute, New York University Grossman School of Medicine, New York, New York 10016, USA
| | - M Laura Feltri
- Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York 14203, USA
- IRCCS Neurological Institute Carlo Besta, Milano 20133, Italy
- Department of Biotechnology and Translational Sciences, Universita' Degli Studi di Milano, Milano 20133, Italy
| | - Claire Jacob
- Faculty of Biology, Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University Mainz, Mainz 55128, Germany
| |
Collapse
|
2
|
Zhang Y, Wang Y, Dou H, Wang S, Qu D, Peng X, Zou N, Yang L. Caffeine improves mitochondrial dysfunction in the white matter of neonatal rats with hypoxia-ischemia through deacetylation: a proteomic analysis of lysine acetylation. Front Mol Neurosci 2024; 17:1394886. [PMID: 38745725 PMCID: PMC11091324 DOI: 10.3389/fnmol.2024.1394886] [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: 03/02/2024] [Accepted: 04/10/2024] [Indexed: 05/16/2024] Open
Abstract
Aims White matter damage (WMD) is linked to both cerebral palsy and cognitive deficits in infants born prematurely. The focus of this study was to examine how caffeine influences the acetylation of proteins within the neonatal white matter and to evaluate its effectiveness in treating white matter damage caused by hypoxia-ischemia. Main methods We employed a method combining affinity enrichment with advanced liquid chromatography and mass spectrometry to profile acetylation in proteins from the white matter of neonatal rats grouped into control (Sham), hypoxic-ischemic (HI), and caffeine-treated (Caffeine) groups. Key findings Our findings included 1,999 sites of lysine acetylation across 1,123 proteins, with quantifiable changes noted in 1,342 sites within 689 proteins. Analysis of these patterns identified recurring sequences adjacent to the acetylation sites, notably YKacN, FkacN, and G * * * GkacS. Investigation into the biological roles of these proteins through Gene Ontology analysis indicated their involvement in a variety of cellular processes, predominantly within mitochondrial locations. Further analysis indicated that the acetylation of tau (Mapt), a protein associated with microtubules, was elevated in the HI condition; however, caffeine treatment appeared to mitigate this over-modification, thus potentially aiding in reducing oxidative stress, inflammation in the nervous system, and improving mitochondrial health. Caffeine inhibited acetylated Mapt through sirtuin 2 (SITR2), promoted Mapt nuclear translocation, and improved mitochondrial dysfunction, which was subsequently weakened by the SIRT2 inhibitor, AK-7. Significance Caffeine-induced changes in lysine acetylation may play a key role in improving mitochondrial dysfunction and inhibiting oxidative stress and neuroinflammation.
Collapse
Affiliation(s)
- Yajun Zhang
- Department of Anesthesiology, Dalian Women and Children's Medical Group, Dalian, Liaoning, China
| | - Yuqian Wang
- Department of Pediatrics, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Haiping Dou
- Department of Pediatrics, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Shanshan Wang
- Department of Pediatrics, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Danyang Qu
- Department of Pediatrics, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Xin Peng
- Department of Pediatrics, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Ning Zou
- Department of Pediatrics, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Liu Yang
- Department of Pediatrics, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
3
|
Shin KO, Crumrine DA, Kim S, Lee Y, Kim B, Abuabara K, Park C, Uchida Y, Wakefield JS, Meyer JM, Jeong S, Park BD, Park K, Elias PM. Phenotypic overlap between atopic dermatitis and autism. BMC Neurosci 2021; 22:43. [PMID: 34157971 PMCID: PMC8218496 DOI: 10.1186/s12868-021-00645-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 06/01/2021] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Autism, a childhood behavioral disorder, belongs to a large suite of diseases, collectively referred to as autism spectrum disorders (ASD). Though multifactorial in etiology, approximately 10% of ASD are associated with atopic dermatitis (AD). Moreover, ASD prevalence increases further as AD severity worsens, though these disorders share no common causative mutations. We assessed here the link between these two disorders in the standard, valproic acid mouse model of ASD. In prior studies, there was no evidence of skin involvement, but we hypothesized that cutaneous involvement could be detected in experiments conducted in BALB/c mice. BALB/c is an albino, laboratory-bred strain of the house mouse and is among the most widely used inbred strains used in animal experimentation. METHODS We performed our studies in valproic acid (VPA)-treated BALB/c hairless mice, a standard mouse model of ASD. Mid-trimester pregnant mice received a single intraperitoneal injection of either valproic acid sodium salt dissolved in saline or saline alone on embryonic day 12.5 and were housed individually until postnatal day 21. Only the brain and epidermis appeared to be affected, while other tissues remain unchanged. At various postnatal time points, brain, skin and blood samples were obtained for histology and for quantitation of tissue sphingolipid content and cytokine levels. RESULTS AD-like changes in ceramide content occurred by day one postpartum in both VPA-treated mouse skin and brain. The temporal co-emergence of AD and ASD, and the AD phenotype-dependent increase in ASD prevalence correlated with early appearance of cytokine markers (i.e., interleukin [IL]-4, 5, and 13), as well as mast cells in skin and brain. The high levels of interferon (IFN)γ not only in skin, but also in brain likely account for a significant decline in esterified very-long-chain N-acyl fatty acids in brain ceramides, again mimicking known IFNγ-induced changes in AD. CONCLUSION Baseline involvement of both AD and ASD could reflect concurrent neuro- and epidermal toxicity, possibly because both epidermis and neural tissues originate from the embryonic neuroectoderm. These studies illuminate the shared susceptibility of the brain and epidermis to a known neurotoxin, suggesting that the atopic diathesis could be extended to include ASD.
Collapse
Affiliation(s)
- Kyong-Oh Shin
- Department of Food Science/Nutrition, & Convergence Program of Material Science for Medicine/Pharmaceutics, and the Korean Institute of Nutrition, Hallym University, Chuncheon, South Korea
| | - Debra A Crumrine
- Dept. of Dermatology, University of California, NCIRE, and Veterans Affairs Medical Center, 4150 Clement Street, MS 190, San Francisco, CA, 94121, USA
| | - Sungeun Kim
- Department of Food Science/Nutrition, & Convergence Program of Material Science for Medicine/Pharmaceutics, and the Korean Institute of Nutrition, Hallym University, Chuncheon, South Korea
| | - Yerin Lee
- Department of Food Science/Nutrition, & Convergence Program of Material Science for Medicine/Pharmaceutics, and the Korean Institute of Nutrition, Hallym University, Chuncheon, South Korea
| | - Bogyeong Kim
- Department of Food Science/Nutrition, & Convergence Program of Material Science for Medicine/Pharmaceutics, and the Korean Institute of Nutrition, Hallym University, Chuncheon, South Korea
| | - Katrina Abuabara
- Department of Dermatology, University of San Francisco, San Francisco, CA, USA
| | - Chaehyeong Park
- Dept. of Dermatology, University of California, NCIRE, and Veterans Affairs Medical Center, 4150 Clement Street, MS 190, San Francisco, CA, 94121, USA
| | - Yoshikazu Uchida
- Dept. of Dermatology, University of California, NCIRE, and Veterans Affairs Medical Center, 4150 Clement Street, MS 190, San Francisco, CA, 94121, USA
| | - Joan S Wakefield
- Dept. of Dermatology, University of California, NCIRE, and Veterans Affairs Medical Center, 4150 Clement Street, MS 190, San Francisco, CA, 94121, USA
| | - Jason M Meyer
- Dept. of Dermatology, University of California, NCIRE, and Veterans Affairs Medical Center, 4150 Clement Street, MS 190, San Francisco, CA, 94121, USA
| | - Sekyoo Jeong
- Dept of Cosmetic Science, Seowon University, Cheongju, South Korea
| | - Byeong Deog Park
- Sphingobrain Inc., San Francisco, CA, USA
- Dr. Raymond Laboratories, Inc, Englewood Cliffs, NJ, USA
| | - Kyungho Park
- Department of Food Science/Nutrition, & Convergence Program of Material Science for Medicine/Pharmaceutics, and the Korean Institute of Nutrition, Hallym University, Chuncheon, South Korea.
| | - Peter M Elias
- Dept. of Dermatology, University of California, NCIRE, and Veterans Affairs Medical Center, 4150 Clement Street, MS 190, San Francisco, CA, 94121, USA.
| |
Collapse
|
4
|
Jokkel Z, Piroska M, Szalontai L, Hernyes A, Tarnoki DL, Tarnoki AD. Twin and family studies on epigenetics of autoimmune diseases. TWIN AND FAMILY STUDIES OF EPIGENETICS 2021:169-191. [DOI: 10.1016/b978-0-12-820951-6.00009-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
5
|
Tondelli M, Vaudano AE, Sisodiya SM, Meletti S. Valproate Use Is Associated With Posterior Cortical Thinning and Ventricular Enlargement in Epilepsy Patients. Front Neurol 2020; 11:622. [PMID: 32714274 PMCID: PMC7351506 DOI: 10.3389/fneur.2020.00622] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 05/27/2020] [Indexed: 01/06/2023] Open
Abstract
Valproate is a drug widely used to treat epilepsy, bipolar disorder, and occasionally to prevent migraine headache. Despite its clinical efficacy, prenatal exposure to valproate is associated with neurodevelopmental impairments and its use in children and adults was associated with rare cases of reversible brain atrophy and ventricular enlargement. To determine whether valproate use is related with structural brain changes we examined through a cross-sectional study cortical and subcortical structures in a group of 152 people with epilepsy and a normal clinical brain MRI. Patients were grouped into those currently using valproate (n = 54), those taking drugs other than valproate (n = 47), and drug-naïve patients (n = 51) at the time of MRI, irrespectively of their epilepsy syndrome. Cortical thickness and subcortical volumes were analyzed using Freesurfer, version 5.0. Subjects exposed to valproate (either in mono- or polytherapy) showed reduced cortical thickness in the occipital lobe, more precisely in the cuneus bilaterally, in the left lingual gyrus, and in left and right pericalcarine gyri when compared to patients who used other antiepileptic drugs, to drug-naïve epilepsy patients, and to healthy controls. Considering the subgroup of patients using valproate monotherapy (n = 25), both comparisons with healthy controls and drug-naïve groups confirmed occipital lobe cortical thickness reduction. Moreover, patients using valproate showed increased left and right lateral ventricle volume compared to all other groups. Notably, subjects who were non-valproate users at the time of MRI, but who had valproate exposure in the past (n = 27) did not show these cortical or subcortical brain changes. Cortical changes in the posterior cortex, particularly in the visual cortex, and ventricular enlargement, are present in people with epilepsy using valproate, independently from clinical and demographical variables. These findings are relevant both for the efficacy and adverse events profile of valproate use in people with epilepsy.
Collapse
Affiliation(s)
| | | | - Sanjay M Sisodiya
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, United Kingdom.,Chalfont Centre for Epilepsy, Chalfont, United Kingdom
| | - Stefano Meletti
- Neurology Unit, OCSAE Hospital, AOU Modena, Modena, Italy.,Department of Biomedical, Metabolic and Neural Science, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
6
|
Duman M, Martinez-Moreno M, Jacob C, Tapinos N. Functions of histone modifications and histone modifiers in Schwann cells. Glia 2020; 68:1584-1595. [PMID: 32034929 DOI: 10.1002/glia.23795] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 01/24/2020] [Accepted: 01/28/2020] [Indexed: 01/25/2023]
Abstract
Schwann cells (SCs) are the main glial cells present in the peripheral nervous system (PNS). Their primary functions are to insulate peripheral axons to protect them from the environment and to enable fast conduction of electric signals along big caliber axons by enwrapping them in a thick myelin sheath rich in lipids. In addition, SCs have the peculiar ability to foster axonal regrowth after a lesion by demyelinating and converting into repair cells that secrete neurotrophic factors and guide axons back to their former target to finally remyelinate regenerated axons. The different steps of SC development and their role in the maintenance of PNS integrity and regeneration after lesion are controlled by various factors among which transcription factors and chromatin-remodeling enzymes hold major functions. In this review, we discussed how histone modifications and histone-modifying enzymes control SC development, maintenance of PNS integrity and response to injury. The functions of histone modifiers as part of chromatin-remodeling complexes are discussed in another review published in the same issue of Glia.
Collapse
Affiliation(s)
- Mert Duman
- Faculty of Biology, Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Margot Martinez-Moreno
- Department of Neurosurgery, Molecular Neuroscience & Neuro-Oncology Laboratory, Brown University, Providence, Rhode Island
| | - Claire Jacob
- Faculty of Biology, Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Nikos Tapinos
- Department of Neurosurgery, Molecular Neuroscience & Neuro-Oncology Laboratory, Brown University, Providence, Rhode Island
| |
Collapse
|
7
|
Ornelas IM, Khandker L, Wahl SE, Hashimoto H, Macklin WB, Wood TL. The mechanistic target of rapamycin pathway downregulates bone morphogenetic protein signaling to promote oligodendrocyte differentiation. Glia 2020; 68:1274-1290. [PMID: 31904150 DOI: 10.1002/glia.23776] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 11/26/2019] [Accepted: 11/29/2019] [Indexed: 12/13/2022]
Abstract
Oligodendrocyte precursor cells (OPCs) differentiate and mature into oligodendrocytes, which produce myelin in the central nervous system. Prior studies have shown that the mechanistic target of rapamycin (mTOR) is necessary for proper myelination of the mouse spinal cord and that bone morphogenetic protein (BMP) signaling inhibits oligodendrocyte differentiation, in part by promoting expression of inhibitor of DNA binding 2 (Id2). Here we provide evidence that mTOR functions specifically in the transition from early stage OPC to immature oligodendrocyte by downregulating BMP signaling during postnatal spinal cord development. When mTOR is deleted from the oligodendrocyte lineage, expression of the FK506 binding protein 1A (FKBP12), a suppressor of BMP receptor activity, is reduced, downstream Smad activity is increased and Id2 expression is elevated. Additionally, mTOR inhibition with rapamycin in differentiating OPCs alters the transcriptional complex present at the Id2 promoter. Deletion of mTOR in oligodendroglia in vivo resulted in fewer late stage OPCs and fewer newly formed oligodendrocytes in the spinal cord with no effect on OPC proliferation or cell cycle exit. Finally, we demonstrate that inhibiting BMP signaling rescues the rapamycin-induced deficit in myelin protein expression. We conclude that mTOR promotes early oligodendrocyte differentiation by suppressing BMP signaling in OPCs.
Collapse
Affiliation(s)
- Isis M Ornelas
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey
| | - Luipa Khandker
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey
| | - Stacey E Wahl
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey
| | - Hirokazu Hashimoto
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, Colorado
| | - Wendy B Macklin
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, Colorado
| | - Teresa L Wood
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey
| |
Collapse
|
8
|
Modrak M, Talukder MAH, Gurgenashvili K, Noble M, Elfar JC. Peripheral nerve injury and myelination: Potential therapeutic strategies. J Neurosci Res 2019; 98:780-795. [PMID: 31608497 DOI: 10.1002/jnr.24538] [Citation(s) in RCA: 127] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 08/30/2019] [Accepted: 09/16/2019] [Indexed: 12/11/2022]
Abstract
Traumatic peripheral nerve injury represents a major clinical and public health problem that often leads to significant functional impairment and permanent disability. Despite modern diagnostic procedures and advanced microsurgical techniques, functional recovery after peripheral nerve repair is often unsatisfactory. Therefore, there is an unmet need for new therapeutic or adjunctive strategies to promote the functional recovery in nerve injury patients. In contrast to the central nervous system, Schwann cells in the peripheral nervous system play a pivotal role in several aspects of nerve repair such as degeneration, remyelination, and axonal growth. Several non-surgical approaches, including pharmacological, electrical, cell-based, and laser therapies, have been employed to promote myelination and enhance functional recovery after peripheral nerve injury. This review will succinctly discuss the potential therapeutic strategies in the context of myelination following peripheral neurotrauma.
Collapse
Affiliation(s)
- Max Modrak
- School of Medicine & Dentistry, The University of Rochester Medical Center, Rochester, New York, USA
| | - M A Hassan Talukder
- Department of Orthopaedics & Rehabilitation, Penn State Hershey College of Medicine, Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Khatuna Gurgenashvili
- Department of Neurology, Penn State Hershey College of Medicine, Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Mark Noble
- Department of Biomedical Genetics, The University of Rochester Medical Center, Rochester, New York, USA
| | - John C Elfar
- Department of Orthopaedics & Rehabilitation, Penn State Hershey College of Medicine, Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| |
Collapse
|
9
|
The Impact of Protein Acetylation/Deacetylation on Systemic Lupus Erythematosus. Int J Mol Sci 2018; 19:ijms19124007. [PMID: 30545086 PMCID: PMC6321219 DOI: 10.3390/ijms19124007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 11/30/2018] [Accepted: 12/10/2018] [Indexed: 02/08/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic inflammatory autoimmune disease in which the body’s immune system mistakenly attacks healthy cells. Although the exact cause of SLE has not been identified, it is clear that both genetics and environmental factors trigger the disease. Identical twins have a 24% chance of getting lupus disease if the other one is affected. Internal factors such as female gender and sex hormones, the major histocompatibility complex (MHC) locus and other genetic polymorphisms have been shown to affect SLE, as well as external, environmental influences such as sunlight exposure, smoking, vitamin D deficiency, and certain infections. Several studies have reported and proposed multiple associations between the alteration of the epigenome and the pathogenesis of autoimmune disease. Epigenetic factors contributing to SLE include microRNAs, DNA methylation status, and the acetylation/deacetylation of histone proteins. Additionally, the acetylation of non-histone proteins can also influence cellular function. A better understanding of non-genomic factors that regulate SLE will provide insight into the mechanisms that initiate and facilitate disease and also contribute to the development of novel therapeutics that can specifically target pathogenic molecular pathways.
Collapse
|
10
|
Gomis-Coloma C, Velasco-Aviles S, Gomez-Sanchez JA, Casillas-Bajo A, Backs J, Cabedo H. Class IIa histone deacetylases link cAMP signaling to the myelin transcriptional program of Schwann cells. J Cell Biol 2018; 217:1249-1268. [PMID: 29472387 PMCID: PMC5881490 DOI: 10.1083/jcb.201611150] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 10/06/2017] [Accepted: 01/17/2018] [Indexed: 12/11/2022] Open
Abstract
Schwann cells respond to cyclic adenosine monophosphate (cAMP) halting proliferation and expressing myelin proteins. Here we show that cAMP signaling induces the nuclear shuttling of the class IIa histone deacetylase (HDAC)-4 in these cells, where it binds to the promoter and blocks the expression of c-Jun, a negative regulator of myelination. To do it, HDAC4 does not interfere with the transcriptional activity of MEF2. Instead, by interacting with NCoR1, it recruits HDAC3 and deacetylates histone 3 in the promoter of c-Jun, blocking gene expression. Importantly, this is enough to up-regulate Krox20 and start Schwann cell differentiation program-inducing myelin gene expression. Using conditional knockout mice, we also show that HDAC4 together with HDAC5 redundantly contribute to activate the myelin transcriptional program and the development of myelin sheath in vivo. We propose a model in which cAMP signaling shuttles class IIa HDACs into the nucleus of Schwann cells to regulate the initial steps of myelination in the peripheral nervous system.
Collapse
Affiliation(s)
- Clara Gomis-Coloma
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández and Consejo Superior de Investigaciones Científicas, Sant Joan, Alicante, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL) and Fundación para el Fomento de la Investigación Saniatria y Biomédica de la Comunidad Valenciana (FISABIO), Alicante, Spain
| | - Sergio Velasco-Aviles
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández and Consejo Superior de Investigaciones Científicas, Sant Joan, Alicante, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL) and Fundación para el Fomento de la Investigación Saniatria y Biomédica de la Comunidad Valenciana (FISABIO), Alicante, Spain
| | - Jose A Gomez-Sanchez
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández and Consejo Superior de Investigaciones Científicas, Sant Joan, Alicante, Spain
- Department of Cell and Developmental Biology, University College London, London, England, UK
| | - Angeles Casillas-Bajo
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández and Consejo Superior de Investigaciones Científicas, Sant Joan, Alicante, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL) and Fundación para el Fomento de la Investigación Saniatria y Biomédica de la Comunidad Valenciana (FISABIO), Alicante, Spain
| | - Johannes Backs
- Department of Molecular Cardiology and Epigenetics, University of Heidelberg, Heidelberg, Germany
- German Center for Cardiovascular Research, Partner Site Heidelberg/Mannheim, Germany
| | - Hugo Cabedo
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández and Consejo Superior de Investigaciones Científicas, Sant Joan, Alicante, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL) and Fundación para el Fomento de la Investigación Saniatria y Biomédica de la Comunidad Valenciana (FISABIO), Alicante, Spain
| |
Collapse
|
11
|
García-Mateo N, Pascua-Maestro R, Pérez-Castellanos A, Lillo C, Sanchez D, Ganfornina MD. Myelin extracellular leaflet compaction requires apolipoprotein D membrane management to optimize lysosomal-dependent recycling and glycocalyx removal. Glia 2017; 66:670-687. [PMID: 29222871 DOI: 10.1002/glia.23274] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 11/09/2017] [Accepted: 11/17/2017] [Indexed: 11/07/2022]
Abstract
To compact the extracellular sides of myelin, an important transition must take place: from membrane sliding, while building the wraps, to membrane adhesion and water exclusion. Removal of the negatively charged glycocalyx becomes the limiting factor in such transition. What is required to initiate this membrane-zipping process? Knocking-out the Lipocalin Apolipoprotein D (ApoD), essential for lysosomal functional integrity in glial cells, results in a specific defect in myelin extracellular leaflet compaction in peripheral and central nervous system, which results in reduced conduction velocity and suboptimal behavioral outputs: motor learning is compromised. Myelination initiation, growth, intracellular leaflet compaction, myelin thickness or internodal length remain unaltered. Lack of ApoD specifically modifies Plp and P0 protein expression, but not Mbp or Mag. Late in myelin maturation period, ApoD affects lipogenic and growth-related, but not stress-responsive, signaling pathways. Without ApoD, the sialylated glycocalyx is maintained and ganglioside content remains high. In peripheral nervous system, Neu3 membrane sialidase and lysosomal Neu1 are coordinately expressed with ApoD in subsets of Schwann cells. ApoD-KO myelin becomes depleted of Neu3 and enriched in Fyn, a kinase with pivotal roles in transducing axon-derived signals into myelin properties. In the absence of ApoD, partial permeabilization of lysosomes alters Neu1 location as well. Exogenous ApoD rescues ApoD-KO hypersialylated glycocalyx in astrocytes, demonstrating that ApoD is necessary and sufficient to control glycocalyx composition in glial cells. By ensuring lysosomal functional integrity and adequate subcellular location of effector and regulatory proteins, ApoD guarantees the glycolipid recycling and glycocalyx removal required to complete myelin compaction.
Collapse
Affiliation(s)
- Nadia García-Mateo
- Instituto de Biología y Genética Molecular-Departamento de Bioquímica y Biología Molecular y Fisiología, Universidad de Valladolid-CSIC, Valladolid, Spain
| | - Raquel Pascua-Maestro
- Instituto de Biología y Genética Molecular-Departamento de Bioquímica y Biología Molecular y Fisiología, Universidad de Valladolid-CSIC, Valladolid, Spain
| | - Alberto Pérez-Castellanos
- Instituto de Biología y Genética Molecular-Departamento de Bioquímica y Biología Molecular y Fisiología, Universidad de Valladolid-CSIC, Valladolid, Spain
| | - Concepción Lillo
- Instituto de Neurociencias de Castilla y León, IBSAL, Universidad de Salamanca, Salamanca, Spain
| | - Diego Sanchez
- Instituto de Biología y Genética Molecular-Departamento de Bioquímica y Biología Molecular y Fisiología, Universidad de Valladolid-CSIC, Valladolid, Spain
| | - Maria D Ganfornina
- Instituto de Biología y Genética Molecular-Departamento de Bioquímica y Biología Molecular y Fisiología, Universidad de Valladolid-CSIC, Valladolid, Spain
| |
Collapse
|
12
|
Jacob C. Chromatin-remodeling enzymes in control of Schwann cell development, maintenance and plasticity. Curr Opin Neurobiol 2017; 47:24-30. [PMID: 28850819 DOI: 10.1016/j.conb.2017.08.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 06/28/2017] [Accepted: 08/10/2017] [Indexed: 01/06/2023]
Abstract
Gene regulation is essential for cellular differentiation and plasticity. Schwann cells (SCs), the myelinating glia of the peripheral nervous system (PNS), develop from neural crest cells to mature myelinating SCs and can at early developmental stage differentiate into various cell types. After a PNS lesion, SCs can also convert into repair cells that guide and stimulate axonal regrowth, and remyelinate regenerated axons. What controls their development and versatile nature? Several recent studies highlight the key roles of chromatin modifiers in these processes, allowing SCs to regulate their gene expression profile and thereby acquire or change their identity and quickly react to their environment.
Collapse
Affiliation(s)
- Claire Jacob
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland.
| |
Collapse
|
13
|
van Tilborg E, Heijnen CJ, Benders MJ, van Bel F, Fleiss B, Gressens P, Nijboer CH. Impaired oligodendrocyte maturation in preterm infants: Potential therapeutic targets. Prog Neurobiol 2015; 136:28-49. [PMID: 26655283 DOI: 10.1016/j.pneurobio.2015.11.002] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 11/02/2015] [Accepted: 11/18/2015] [Indexed: 12/20/2022]
Abstract
Preterm birth is an evolving challenge in neonatal health care. Despite declining mortality rates among extremely premature neonates, morbidity rates remain very high. Currently, perinatal diffuse white matter injury (WMI) is the most commonly observed type of brain injury in preterm infants and has become an important research area. Diffuse WMI is associated with impaired cognitive, sensory and psychological functioning and is increasingly being recognized as a risk factor for autism-spectrum disorders, ADHD, and other psychological disturbances. No treatment options are currently available for diffuse WMI and the underlying pathophysiological mechanisms are far from being completely understood. Preterm birth is associated with maternal inflammation, perinatal infections and disrupted oxygen supply which can affect the cerebral microenvironment by causing activation of microglia, astrogliosis, excitotoxicity, and oxidative stress. This intricate interplay of events negatively influences oligodendrocyte development, causing arrested oligodendrocyte maturation or oligodendrocyte cell death, which ultimately results in myelination failure in the developing white matter. This review discusses the current state in perinatal WMI research, ranging from a clinical perspective to basic molecular pathophysiology. The complex regulation of oligodendrocyte development in healthy and pathological conditions is described, with a specific focus on signaling cascades that may play a role in WMI. Furthermore, emerging concepts in the field of WMI and issues regarding currently available animal models are put forward. Novel insights into the molecular mechanisms underlying impeded oligodendrocyte maturation in diffuse WMI may aid the development of novel treatment options which are desperately needed to improve the quality-of-life of preterm neonates.
Collapse
Affiliation(s)
- Erik van Tilborg
- Laboratory of Neuroimmunology and Developmental Origins of Disease, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Cobi J Heijnen
- Laboratory of Neuroimmunology, Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Manon J Benders
- Department of Neonatology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Frank van Bel
- Department of Neonatology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Bobbi Fleiss
- Inserm, Paris U1141, France; Université Paris Diderot, Sorbonne Paris Cité, UMRS, Paris 1141, France; Centre for the Developing Brain, Department of Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' Hospital, London, United Kingdom
| | - Pierre Gressens
- Inserm, Paris U1141, France; Université Paris Diderot, Sorbonne Paris Cité, UMRS, Paris 1141, France; Centre for the Developing Brain, Department of Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' Hospital, London, United Kingdom
| | - Cora H Nijboer
- Laboratory of Neuroimmunology and Developmental Origins of Disease, University Medical Center Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
14
|
The Autotaxin-Lysophosphatidic Acid Axis Modulates Histone Acetylation and Gene Expression during Oligodendrocyte Differentiation. J Neurosci 2015; 35:11399-414. [PMID: 26269646 DOI: 10.1523/jneurosci.0345-15.2015] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
UNLABELLED During development, oligodendrocytes (OLGs), the myelinating cells of the CNS, undergo a stepwise progression during which OLG progenitors, specified from neural stem/progenitor cells, differentiate into fully mature myelinating OLGs. This progression along the OLG lineage is characterized by well synchronized changes in morphology and gene expression patterns. The latter have been found to be particularly critical during the early stages of the lineage, and they have been well described to be regulated by epigenetic mechanisms, especially by the activity of the histone deacetylases HDAC1 and HDAC2. The data presented here identify the extracellular factor autotaxin (ATX) as a novel upstream signal modulating HDAC1/2 activity and gene expression in cells of the OLG lineage. Using the zebrafish as an in vivo model system as well as rodent primary OLG cultures, this functional property of ATX was found to be mediated by its lysophospholipase D (lysoPLD) activity, which has been well characterized to generate the lipid signaling molecule lysophosphatidic acid (LPA). More specifically, the lysoPLD activity of ATX was found to modulate HDAC1/2 regulated gene expression during a time window coinciding with the transition from OLG progenitor to early differentiating OLG. In contrast, HDAC1/2 regulated gene expression during the transition from neural stem/progenitor to OLG progenitor appeared unaffected by ATX and its lysoPLD activity. Thus, together, our data suggest that an ATX-LPA-HDAC1/2 axis regulates OLG differentiation specifically during the transition from OLG progenitor to early differentiating OLG and via a molecular mechanism that is evolutionarily conserved from at least zebrafish to rodent. SIGNIFICANCE STATEMENT The formation of the axon insulating and supporting myelin sheath by differentiating oligodendrocytes (OLGs) in the CNS is considered an essential step during vertebrate development. In addition, loss and/or dysfunction of the myelin sheath has been associated with a variety of neurologic diseases in which repair is limited, despite the presence of progenitor cells with the potential to differentiate into myelinating OLGs. This study characterizes the autotaxin-lysophosphatidic acid signaling axis as a modulator of OLG differentiation in vivo in the developing zebrafish and in vitro in rodent OLGs in culture. These findings provide novel insight into the regulation of developmental myelination, and they are likely to lead to advancing studies related to the stimulation of myelin repair under pathologic conditions.
Collapse
|
15
|
HDAC1/2-Dependent P0 Expression Maintains Paranodal and Nodal Integrity Independently of Myelin Stability through Interactions with Neurofascins. PLoS Biol 2015; 13:e1002258. [PMID: 26406915 PMCID: PMC4583457 DOI: 10.1371/journal.pbio.1002258] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2015] [Accepted: 08/19/2015] [Indexed: 12/16/2022] Open
Abstract
The pathogenesis of peripheral neuropathies in adults is linked to maintenance mechanisms that are not well understood. Here, we elucidate a novel critical maintenance mechanism for Schwann cell (SC)–axon interaction. Using mouse genetics, ablation of the transcriptional regulators histone deacetylases 1 and 2 (HDAC1/2) in adult SCs severely affected paranodal and nodal integrity and led to demyelination/remyelination. Expression levels of the HDAC1/2 target gene myelin protein zero (P0) were reduced by half, accompanied by altered localization and stability of neurofascin (NFasc)155, NFasc186, and loss of Caspr and septate-like junctions. We identify P0 as a novel binding partner of NFasc155 and NFasc186, both in vivo and by in vitro adhesion assay. Furthermore, we demonstrate that HDAC1/2-dependent P0 expression is crucial for the maintenance of paranodal/nodal integrity and axonal function through interaction of P0 with neurofascins. In addition, we show that the latter mechanism is impaired by some P0 mutations that lead to late onset Charcot-Marie-Tooth disease. The well-studied Schwann cell protein P0 is revealed to have an unsuspected function critical for the stability of paranodes and nodes in adult nerves. This function is specifically impaired by P0 mutations that lead to late-onset forms of Charcot-Marie-Tooth disease. Peripheral nerves consist mainly of axons and Schwann cells, which form myelin sheaths around axons. Peripheral neuropathies primarily affect axons, their myelin, or both. Etiologies are multiple: they can be inherited, autoimmune, infectious, metabolic (e.g., diabetes), or be due to tumors or toxic agents. However, the pathogenesis mechanisms of these disorders are not well understood. Here, we elucidate a novel critical mechanism in peripheral nerves for the stability of two adjacent structures of major importance for axonal function, the paranodes and nodes of Ranvier. We find that disruption of these structures causes a form of peripheral neuropathy. Ablation of the transcriptional regulators histone deacetylases (HDAC)1 and 2 in adult Schwann cells results in motor and sensory dysfunction, disruption of paranodal/nodal integrity, and loss of myelin. Expression of the HDAC1/2 target gene myelin protein zero (P0) was reduced by half, leading to altered localization of paranodal and nodal neurofascins, loss of paranodal Caspr, and impairment of axon–Schwann cell interaction in paranodal/nodal regions. We demonstrate that P0, the most abundant protein of peripheral compact myelin, extends to paranodes/nodes to maintain their stability by binding neurofascins. P0–neurofascins binding is affected by P0 mutations responsible for late onset forms of the inherited peripheral neuropathy Charcot-Marie-Tooth disease, identifying a pathogenesis mechanism of these disorders.
Collapse
|
16
|
Jacob C. Transcriptional control of neural crest specification into peripheral glia. Glia 2015; 63:1883-1896. [PMID: 25752517 DOI: 10.1002/glia.22816] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 01/29/2015] [Accepted: 02/20/2015] [Indexed: 12/20/2022]
Abstract
The neural crest is a transient migratory multipotent cell population that originates from the neural plate border and is formed at the end of gastrulation and during neurulation in vertebrate embryos. These cells give rise to many different cell types of the body such as chondrocytes, smooth muscle cells, endocrine cells, melanocytes, and cells of the peripheral nervous system including different subtypes of neurons and peripheral glia. Acquisition of lineage-specific markers occurs before or during migration and/or at final destination. What are the mechanisms that direct specification of neural crest cells into a specific lineage and how do neural crest cells decide on a specific migration route? Those are fascinating and complex questions that have existed for decades and are still in the research focus of developmental biologists. This review discusses transcriptional events and regulations occurring in neural crest cells and derived lineages, which control specification of peripheral glia, namely Schwann cell precursors that interact with peripheral axons and further differentiate into myelinating or nonmyelinating Schwann cells, satellite cells that remain tightly associated with neuronal cell bodies in sensory and autonomous ganglia, and olfactory ensheathing cells that wrap olfactory axons, both at the periphery in the olfactory mucosa and in the central nervous system in the olfactory bulb. Markers of the different peripheral glia lineages including intermediate multipotent cells such as boundary cap cells, as well as the functions of these specific markers, are also reviewed. Enteric ganglia, another type of peripheral glia, will not be discussed in this review. GLIA 2015;63:1883-1896.
Collapse
Affiliation(s)
- Claire Jacob
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
17
|
Liu Y, Liu Y, Nie X, Cao J, Zhu X, Zhang W, Liu Z, Mao X, Yan S, Ni Y, Wang Y. Up-regulation of HDAC4 is associated with Schwann cell proliferation after sciatic nerve crush. Neurochem Res 2014; 39:2105-17. [PMID: 25103231 DOI: 10.1007/s11064-014-1401-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Revised: 06/29/2014] [Accepted: 07/24/2014] [Indexed: 12/12/2022]
Abstract
Histone deacetylase 4 (HDAC4), a member of the class IIa HDACs subfamily, has emerged as a critical regulator of cell growth, differentiation, and migration in various cell types. It was reported that HDAC4 stimulated colon cell proliferation via repression of p21. Also, HDAC4 contributes to platelet-derived growth factor-BB-induced proliferation and migration of vascular smooth muscle cells. Furthermore, HDAC4 may play an important role in the regulation of neuronal differentiation and survival. However, the role of HDAC4 in the process of peripheral nervous system regeneration after injury remains virtually unknown. Herein, we investigated the spatiotemporal expression of HDAC4 in a rat sciatic nerve crush model. We found that sciatic nerve crush induced up-regulated expression of HDAC4 in Schwann cells. Moreover, the expression of the proliferation marker Ki-67 exhibited a similar tendency with that of HDAC4. In cell cultures, we observed increased expression of HDAC4 during the process of TNF-α-induced Schwann cell proliferation, whereas the protein level of p21 was down-regulated. Interference of HDAC4 led to enhanced expression of p21 and impaired proliferation of Schwan cells. Taken together, our findings implicated that HDAC4 was up-regulated in the sciatic nerve after crush, which was associated with proliferation of Schwann cells.
Collapse
Affiliation(s)
- Yonghua Liu
- Department of Pathogen Biology, Medical College, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Wnt signaling in remyelination in multiple sclerosis: friend or foe? Mol Neurobiol 2013; 49:1117-25. [PMID: 24243343 DOI: 10.1007/s12035-013-8584-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 11/01/2013] [Indexed: 12/22/2022]
Abstract
Myelination is critical to normal functioning of the vertebrate nervous system. In demyelinating diseases such as multiple sclerosis, oligodendrocytes, the myelinating cells in the central nervous system, are targeted, resulting in myelin loss, axonal damage, and severe functional impairment. While spontaneous remyelination has been proven a failure in multiple sclerosis, understanding the molecular mechanism underlying oligodendrocyte biology, myelination, and remyelination becomes crucial. To date, a series of signaling pathways in regulating oligodendrocyte development and remyelination have been suggested and, among them, the Wnt/β-catenin/Tcf pathway has been considered a negative factor in the myelinating process. However, this notion has been challenged by recent studies, which showed a pro-myelinating effect of this pathway. This review summarizes the current contradictory concepts concerning the role of the Wnt pathway in the oligodendrocyte development and remyelination process, attempts to address the potential mechanism underlying this controversy, and recommends caution in targeting the Wnt pathway as a potential demyelinating therapy.
Collapse
|
19
|
Dimethyl fumarate regulates histone deacetylase expression in astrocytes. J Neuroimmunol 2013; 263:13-9. [DOI: 10.1016/j.jneuroim.2013.07.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 06/13/2013] [Accepted: 07/09/2013] [Indexed: 12/26/2022]
|
20
|
Zuchero JB, Barres BA. Intrinsic and extrinsic control of oligodendrocyte development. Curr Opin Neurobiol 2013; 23:914-20. [PMID: 23831087 DOI: 10.1016/j.conb.2013.06.005] [Citation(s) in RCA: 126] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Accepted: 06/11/2013] [Indexed: 11/24/2022]
Abstract
Oligodendrocytes (OLs) are the myelinating glia of the central nervous system. Myelin is essential for the rapid propagation of action potentials as well as for metabolic support of axons, and its loss in demyelinating diseases like multiple sclerosis has profound pathological consequences. The many steps in the development of OLs - from the specification of oligodendrocyte precursor cells (OPCs) during embryonic development to their differentiation into OLs that myelinate axons - are under tight regulation. Here we discuss recent advances in understanding how these steps of OL development are controlled intrinsically by transcription factors and chromatin remodeling and extrinsically by signaling molecules and neuronal activity. We also discuss how knowledge of these pathways is now allowing us to take steps toward generating patient-specific OPCs for disease modeling and myelin repair.
Collapse
Affiliation(s)
- J Bradley Zuchero
- Department of Neurobiology, Stanford University, Fairchild Building D205, 299 Campus Drive West, Stanford, CA 94305, United States.
| | | |
Collapse
|
21
|
Histone methylation in the nervous system: functions and dysfunctions. Mol Neurobiol 2012; 47:740-56. [PMID: 23161382 DOI: 10.1007/s12035-012-8376-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Accepted: 11/04/2012] [Indexed: 11/26/2022]
Abstract
Chromatin remodeling is a key epigenetic process controlling the regulation of gene transcription. Local changes of chromatin architecture can be achieved by post-translational modifications of histones such as methylation, acetylation, phosphorylation, ubiquitination, sumoylation, and ADP-ribosylation. These changes are dynamic and allow for rapid repression or de-repression of specific target genes. Chromatin remodeling enzymes are largely involved in the control of cellular differentiation, and loss or gain of function is often correlated with pathological events. For these reasons, research on chromatin remodeling enzymes is currently very active and rapidly expanding, these enzymes representing very promising targets for the design of novel therapeutics in different areas of medicine including oncology and neurology. In this review, we focus on histone methylation in the nervous system. We provide an overview on mammalian histone methyltransferases and demethylases and their mechanisms of action, and we discuss their roles in the development of the nervous system and their involvement in neurodevelopmental, neurodegenerative, and behavioral disorders.
Collapse
|
22
|
de Monasterio-Schrader P, Jahn O, Tenzer S, Wichert SP, Patzig J, Werner HB. Systematic approaches to central nervous system myelin. Cell Mol Life Sci 2012; 69:2879-94. [PMID: 22441408 PMCID: PMC11114939 DOI: 10.1007/s00018-012-0958-9] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2012] [Accepted: 03/05/2012] [Indexed: 12/11/2022]
Abstract
Rapid signal propagation along vertebrate axons is facilitated by their insulation with myelin, a plasma membrane specialization of glial cells. The recent application of 'omics' approaches to the myelinating cells of the central nervous system, oligodendrocytes, revealed their mRNA signatures, enhanced our understanding of how myelination is regulated, and established that the protein composition of myelin is much more complex than previously thought. This review provides a meta-analysis of the > 1,200 proteins thus far identified by mass spectrometry in biochemically purified central nervous system myelin. Contaminating proteins are surprisingly infrequent according to bioinformatic prediction of subcellular localization and comparison with the transcriptional profile of oligodendrocytes. The integration of datasets also allowed the subcategorization of the myelin proteome into functional groups comprising genes that are coregulated during oligodendroglial differentiation. An unexpectedly large number of myelin-related genes cause-when mutated in humans-hereditary diseases affecting the physiology of the white matter. Systematic approaches to oligodendrocytes and myelin thus provide valuable resources for the molecular dissection of developmental myelination, glia-axonal interactions, leukodystrophies, and demyelinating diseases.
Collapse
Affiliation(s)
| | - Olaf Jahn
- Proteomics Group, Max Planck Institute of Experimental Medicine, Göttingen, Germany
- DFG Research Center for Molecular Physiology of the Brain, Göttingen, Germany
| | - Stefan Tenzer
- Institute of Immunology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Sven P. Wichert
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Hermann-Rein-Str. 3, 37075 Göttingen, Germany
| | - Julia Patzig
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Hermann-Rein-Str. 3, 37075 Göttingen, Germany
| | - Hauke B. Werner
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Hermann-Rein-Str. 3, 37075 Göttingen, Germany
| |
Collapse
|
23
|
Rosenzweig I, Vukadinovic Z, Turner AJ, Catani M. Neuroconnectivity and valproic acid: the myelin hypothesis. Neurosci Biobehav Rev 2012; 36:1848-56. [PMID: 22652270 DOI: 10.1016/j.neubiorev.2012.05.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2011] [Revised: 04/09/2012] [Accepted: 05/19/2012] [Indexed: 02/08/2023]
Abstract
Neuropsychiatric medications that directly alter the epigenome, such as valproic acid, can under certain conditions reactivate critical developmental periods and thus impact adult neuroconnectivity. In animal models valproic acid was shown to inhibit the process of postnatal myelination and to replicate age-dependent decline in remyelination efficiency. The human central nervous system's myelination process, unlike that of non-human primates commonly used in the experimental models, is an intricate heterochronous process that continues well into adult life and which probably underlies later life neurocognitive changes and plasticity. Chronic exposure to valproic acid, especially in patients with epilepsy and neuropsychiatric disorders, may profoundly affect this process and its developmental trajectory. Further studies using novel MRI methods that allow in vivo mapping of myelination trajectories across the lifespan are urgently required to address the potential effects of valproic acid on brain development.
Collapse
Affiliation(s)
- Ivana Rosenzweig
- Academic Unit of Sleep and Department of Psychiatry, Royal Brompton Hospital, London, UK.
| | | | | | | |
Collapse
|
24
|
Salerno KM, Jing X, Diges CM, Cornuet PK, Glorioso JC, Albers KM. Sox11 modulates brain-derived neurotrophic factor expression in an exon promoter-specific manner. J Neurosci Res 2012; 90:1011-9. [PMID: 22331573 DOI: 10.1002/jnr.23010] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2011] [Revised: 09/23/2011] [Accepted: 11/18/2011] [Indexed: 12/23/2022]
Abstract
Sox11 is a high-mobility group (HMG)-containing transcription factor that is significantly elevated in peripheral neurons in response to nerve injury. In vitro and in vivo studies support a central role for Sox11 in adult neuron growth and survival following injury. Brain-derived neurotrophic factor (BDNF) is a pleiotropic growth factor that has effects on neuronal survival, differentiation, synaptic plasticity, and regeneration. BDNF transcription is elevated in the dorsal root ganglia (DRG) following nerve injury in parallel with Sox11, allowing for the possible regulation by Sox11. To begin to assess the possible influence of Sox11, we used reverse transcriptase PCR assays to determine the relative expression of the nine (I-IXa) noncoding exons and one coding exon (exon IX) of the BDNF gene after sciatic nerve axotomy in the mouse. Exons with upstream promoter regions containing the Sox binding motif 5'-AACAAAG-3' (I, IV, VII, and VIII) were increased at 1 or 3 days following axotomy. Exons 1 and IV showed the greatest increase, and only exon 1 remained elevated at 3 days. Luciferase assays showed that Sox11 could activate the most highly regulated exons, I and IV, and that this activation was reduced by mutation of putative Sox binding sites. Exon expression in injured DRG neurons had some overlap with Neuro2a cells that overexpress Sox11, showing elevation in exon IV and VII transcripts. These findings indicate cell type and contextual specificity of Sox11 in modulation of BDNF transcription.
Collapse
Affiliation(s)
- Kathleen M Salerno
- Department of Medicine, Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | | | | | | | | |
Collapse
|
25
|
Pereira JA, Lebrun-Julien F, Suter U. Molecular mechanisms regulating myelination in the peripheral nervous system. Trends Neurosci 2011; 35:123-34. [PMID: 22192173 DOI: 10.1016/j.tins.2011.11.006] [Citation(s) in RCA: 187] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2011] [Revised: 11/17/2011] [Accepted: 11/18/2011] [Indexed: 12/21/2022]
Abstract
Glial cells and neurons are engaged in a continuous and highly regulated bidirectional dialog. A remarkable example is the control of myelination. Oligodendrocytes in the central nervous system (CNS) and Schwann cells (SCs) in the peripheral nervous system (PNS) wrap their plasma membranes around axons to organize myelinated nerve fibers that allow rapid saltatory conduction. The functionality of this system is critical, as revealed by numerous neurological diseases that result from deregulation of the system, including multiple sclerosis and peripheral neuropathies. In this review we focus on PNS myelination and present a conceptual framework that integrates crucial signaling mechanisms with basic SC biology. We will highlight signaling hubs and overarching molecular mechanisms, including genetic, epigenetic, and post-translational controls, which together regulate the interplay between SCs and axons, extracellular signals, and the transcriptional network.
Collapse
Affiliation(s)
- Jorge A Pereira
- Institute of Cell Biology, Department of Biology, Swiss Federal Institute of Technology, ETH Zürich, Zürich, Switzerland
| | | | | |
Collapse
|