1
|
Abasi M, Kianmehr A, Variji A, Sangali P, Mahrooz A. microRNAs as molecular tools for brain health: Neuroprotective potential in neurodegenerative disorders. Neuroscience 2025; 574:83-103. [PMID: 40210196 DOI: 10.1016/j.neuroscience.2025.04.012] [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: 09/15/2024] [Revised: 03/09/2025] [Accepted: 04/05/2025] [Indexed: 04/12/2025]
Abstract
As research on microRNAs (miRNAs) advances, it is becoming increasingly clear that these small molecules play crucial roles in the central nervous system (CNS). They are involved in various essential neuronal functions, with specific miRNAs preferentially expressed in different cell types within the nervous system. Notably, certain miRNAs are found at higher levels in the brain and spinal cord compared to other tissues, suggesting they may have specialized functions in the CNS. miRNAs associated with long-term neurodegenerative changes could serve as valuable tools for early treatment decisions and disease monitoring. The significance of miRNAs such as miR-320, miR-146 and miR-29 in the early diagnosis of neurodegenerative disorders becomes evident, especially considering that many neurological and physical symptoms manifest only after substantial degeneration of specific neurons. Interestingly, serum miRNA levels such as miR-92 and miR-486 may correlate with various MRI parameters in multiple sclerosis. Targeting miRNAs using antisense strategies, such as antisense miR-146 and miR-485, may provide advantages over targeting mRNAs, as a single anti-miRNA can regulate multiple disease-related genes. In the future, anti-miRNA-based therapeutic approaches could be integrated into the clinical management of neurological diseases. Certain miRNAs, including miR-223, miR-106, miR-181, and miR-146, contribute to the pathogenesis of various neurodegenerative diseases and thus warrant greater attention. This knowledge could pave the way for the identification of new diagnostic, prognostic, and theranostic biomarkers, and potentially guiding the development of RNA-based therapeutic strategies. This review highlights recent research on the roles of miRNAs in the nervous system, particularly their protective functions in neurodegenerative disorders.
Collapse
Affiliation(s)
- Mozhgan Abasi
- Department of Tissue Engineering and Applied Cell Sciences, Faculty of Advanced Technologies in Medicine, Mazandaran University of Medical Sciences, Sari, Iran; Immunogenetics Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Anvarsadat Kianmehr
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Athena Variji
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia; Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Parisa Sangali
- Department of Clinical Biochemistry and Medical Genetics, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Abdolkarim Mahrooz
- Immunogenetics Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
2
|
Ionescu C, Ghidersa M, Ciobica A, Mavroudis I, Kazis D, Petridis FE, Gorgan DL, Balmus IM. Potential Correlation Between Molecular Biomarkers and Oxidative Stress in Traumatic Brain Injury. Int J Mol Sci 2025; 26:3858. [PMID: 40332547 PMCID: PMC12027598 DOI: 10.3390/ijms26083858] [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: 03/14/2025] [Revised: 04/14/2025] [Accepted: 04/16/2025] [Indexed: 05/08/2025] Open
Abstract
Diagnosing traumatic brain injury (TBI) remains challenging due to an incomplete understanding of its neuropathological mechanisms. TBI is recognised as a complex condition involving both primary and secondary injuries. Although oxidative stress is a non-specific molecular phenomenon observed in various neuropathological conditions, it plays a crucial role in brain injury response and recovery. Due to these aspects, we aimed to evaluate the interaction between some known TBI molecular biomarkers and oxidative stress in providing evidence for its possible relevance in clinical diagnosis and outcome prediction. We found that while many of the currently validated molecular biomarkers interact with oxidative pathways, their patterns of variation could assist the diagnosis, prognosis, and outcomes prediction in TBI cases.
Collapse
Affiliation(s)
- Cătălina Ionescu
- Department of Biology, Faculty of Biology, “Alexandru Ioan Cuza” University of Iasi, 700505 Iasi, Romania; (C.I.); (M.G.); (A.C.); (D.L.G.)
| | - Madalina Ghidersa
- Department of Biology, Faculty of Biology, “Alexandru Ioan Cuza” University of Iasi, 700505 Iasi, Romania; (C.I.); (M.G.); (A.C.); (D.L.G.)
| | - Alin Ciobica
- Department of Biology, Faculty of Biology, “Alexandru Ioan Cuza” University of Iasi, 700505 Iasi, Romania; (C.I.); (M.G.); (A.C.); (D.L.G.)
- “Ioan Haulica” Institute, Apollonia University, 700511 Iasi, Romania
- Center of Biomedical Research, Romanian Academy, Iasi Branch, 2 Teodor Codrescu Street, 700481 Iasi, Romania
| | - Ioannis Mavroudis
- Academy of Romanian Scientists, 050094 Bucharest, Romania
- Department of Neurosciences, Leeds Teaching Hospitals, Leeds LS9 7TF, UK
| | - Dimitrios Kazis
- Third Department of Neurology, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (D.K.); (F.E.P.)
| | - Foivos E. Petridis
- Third Department of Neurology, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (D.K.); (F.E.P.)
| | - Dragoș Lucian Gorgan
- Department of Biology, Faculty of Biology, “Alexandru Ioan Cuza” University of Iasi, 700505 Iasi, Romania; (C.I.); (M.G.); (A.C.); (D.L.G.)
| | - Ioana-Miruna Balmus
- Department of Exact Sciences and Natural Sciences, Institute of Interdisciplinary Research, “Alexandru Ioan Cuza” University of Iasi, 700057 Iasi, Romania;
| |
Collapse
|
3
|
Schumacher N, Vandenbosch R, Franzen R. Peripheral myelin: From development to maintenance. J Neurochem 2025; 169:e16268. [PMID: 39655795 DOI: 10.1111/jnc.16268] [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: 05/15/2024] [Revised: 10/24/2024] [Accepted: 10/28/2024] [Indexed: 12/18/2024]
Abstract
Peripheral myelin is synthesized by glial cells called Schwann cells (SCs). SC development and differentiation must be tightly regulated to avoid any pathological consequence affecting peripheral nerve function. Neuropathic symptoms can arise from developmental issues in SCs, as well as in adult life through processes affecting mature SCs. In this review we focus on SC differentiation from the immature towards the myelinating and non-myelinating SC stages, defining molecular mechanisms outlining radial sorting, a multi-stepped event essential for immature SC differentiation and myelination. We also describe mechanisms regulating myelin sheath maintenance and SC homeostasis during aging. Finally, we will conclude with some remaining questions in the field of SC biology.
Collapse
Affiliation(s)
- Nathalie Schumacher
- Laboratory of Nervous System Disorders and Therapies, GIGA Institute, University of Liège, Liège, Belgium
| | - Renaud Vandenbosch
- Laboratory of Developmental Neurobiology, GIGA Institute, University of Liège, Liège, Belgium
| | - Rachelle Franzen
- Laboratory of Nervous System Disorders and Therapies, GIGA Institute, University of Liège, Liège, Belgium
| |
Collapse
|
4
|
Schwann cell functions in peripheral nerve development and repair. Neurobiol Dis 2023; 176:105952. [PMID: 36493976 DOI: 10.1016/j.nbd.2022.105952] [Citation(s) in RCA: 83] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/23/2022] [Accepted: 12/05/2022] [Indexed: 12/12/2022] Open
Abstract
The glial cell of the peripheral nervous system (PNS), the Schwann cell (SC), counts among the most multifaceted cells of the body. During development, SCs secure neuronal survival and participate in axonal path finding. Simultaneously, they orchestrate the architectural set up of the developing nerves, including the blood vessels and the endo-, peri- and epineurial layers. Perinatally, in rodents, SCs radially sort and subsequently myelinate individual axons larger than 1 μm in diameter, while small calibre axons become organised in non-myelinating Remak bundles. SCs have a vital role in maintaining axonal health throughout life and several specialized SC types perform essential functions at specific locations, such as terminal SC at the neuromuscular junction (NMJ) or SC within cutaneous sensory end organs. In addition, neural crest derived satellite glia maintain a tight communication with the soma of sensory, sympathetic, and parasympathetic neurons and neural crest derivatives are furthermore an indispensable part of the enteric nervous system. The remarkable plasticity of SCs becomes evident in the context of a nerve injury, where SC transdifferentiate into intriguing repair cells, which orchestrate a regenerative response that promotes nerve repair. Indeed, the multiple adaptations of SCs are captivating, but remain often ill-resolved on the molecular level. Here, we summarize and discuss the knowns and unknowns of the vast array of functions that this single cell type can cover in peripheral nervous system development, maintenance, and repair.
Collapse
|
5
|
Li JY, Li QQ, Sheng R. The role and therapeutic potential of exosomes in ischemic stroke. Neurochem Int 2021; 151:105194. [PMID: 34582960 DOI: 10.1016/j.neuint.2021.105194] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 09/05/2021] [Accepted: 09/25/2021] [Indexed: 01/08/2023]
Abstract
Ischemic stroke is a disease caused by insufficient blood and oxygen supply to the brain, which is mainly due to intracranial arterial stenosis and middle cerebral artery occlusion. Exosomes play an important role in cerebral ischemia. Nucleic acid substances such as miRNA, circRNA, lncRNA in exosomes can play communication roles and improve cerebral ischemia by regulating the development and regeneration of the nervous system, remodeling of blood vessels and inhibiting neuroinflammation. Furthermore, exosomes modulate stroke through various mechanisms, including improving neural communication, promoting the development of neuronal cells and myelin synapses, neurovascular unit remodeling and maintaining homeostasis of the nervous system. At the same time, exosomes are also a good carrier of bioactive substances, which can be modified and targeted to the lesion site. Here, we review the roles of exosomes in cerebral ischemia, and discuss the possible mechanisms and potentials of modification of exosomes for targeting stroke, providing a new idea for the prevention and treatment of cerebral ischemia.
Collapse
Affiliation(s)
- Jia-Ying Li
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, China
| | - Qi-Qi Li
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, China
| | - Rui Sheng
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, China.
| |
Collapse
|
6
|
Ikumi A, Gingery A, Toyoshima Y, Zhao C, Moran SL, Livia C, Rolland T, Peterson T, Sabbah MS, Boroumand S, Saffari TM, Behfar A, Shin AY, Amadio PC. Administration of Purified Exosome Product in a Rat Sciatic Serve Reverse Autograft Model. Plast Reconstr Surg 2021; 148:200e-211e. [PMID: 34153020 DOI: 10.1097/prs.0000000000008202] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND The nerve autograft remains the gold standard when reconstructing peripheral nerve defects. However, although autograft repair can result in useful functional recovery, poor outcomes are common, and better treatments are needed. The purpose of this study was to evaluate the effect of purified exosome product on functional motor recovery and nerve-related gene expression in a rat sciatic nerve reverse autograft model. METHODS Ninety-six Sprague-Dawley rats were divided into three experimental groups. In each group, a unilateral 10-mm sciatic nerve defect was created. The excised nerve was reversed and used to reconstruct the defect. Group I animals received the reversed autograft alone, group II animals received the reversed autograft with fibrin glue, and group III animals received the reversed autograft with purified exosome product suspended in the fibrin glue. The animals were killed at 3 and 7 days and 12 and 16 weeks after surgery. Evaluation included compound muscle action potentials, isometric tetanic force, tibialis anterior muscle wet weight, nerve regeneration-related gene expression, and nerve histomorphometry. RESULTS At 16 weeks, isometric tetanic force was significantly better in group III (p = 0.03). The average axon diameter of the peroneal nerve was significantly larger in group III at both 12 and 16 weeks (p = 0.015 at 12 weeks; p < 0.01 at 16 weeks). GAP43 and S100b gene expression was significantly up-regulated by purified exosome product. CONCLUSIONS Local administration of purified exosome product demonstrated improved nerve regeneration profiles in the reverse sciatic nerve autograft rat model. Thus, purified exosome product may have beneficial effects on nerve regeneration, gene profiles, and motor outcomes.
Collapse
Affiliation(s)
- Akira Ikumi
- From the Division of Orthopedic Research, Department of Orthopedic Surgery, the Department of Biochemistry and Molecular Biology, the Division of Plastic and Reconstructive Surgery, the Van Cleve Cardiac Regeneration Program, Center for Regenerative Medicine, the Division of Hand Surgery, Department of Orthopedic Surgery, and the Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic; and the Division of Cardiovascular Diseases, Department of Medicine, Mayo Clinic College of Medicine
| | - Anne Gingery
- From the Division of Orthopedic Research, Department of Orthopedic Surgery, the Department of Biochemistry and Molecular Biology, the Division of Plastic and Reconstructive Surgery, the Van Cleve Cardiac Regeneration Program, Center for Regenerative Medicine, the Division of Hand Surgery, Department of Orthopedic Surgery, and the Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic; and the Division of Cardiovascular Diseases, Department of Medicine, Mayo Clinic College of Medicine
| | - Yoichi Toyoshima
- From the Division of Orthopedic Research, Department of Orthopedic Surgery, the Department of Biochemistry and Molecular Biology, the Division of Plastic and Reconstructive Surgery, the Van Cleve Cardiac Regeneration Program, Center for Regenerative Medicine, the Division of Hand Surgery, Department of Orthopedic Surgery, and the Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic; and the Division of Cardiovascular Diseases, Department of Medicine, Mayo Clinic College of Medicine
| | - Chunfeng Zhao
- From the Division of Orthopedic Research, Department of Orthopedic Surgery, the Department of Biochemistry and Molecular Biology, the Division of Plastic and Reconstructive Surgery, the Van Cleve Cardiac Regeneration Program, Center for Regenerative Medicine, the Division of Hand Surgery, Department of Orthopedic Surgery, and the Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic; and the Division of Cardiovascular Diseases, Department of Medicine, Mayo Clinic College of Medicine
| | - Steven L Moran
- From the Division of Orthopedic Research, Department of Orthopedic Surgery, the Department of Biochemistry and Molecular Biology, the Division of Plastic and Reconstructive Surgery, the Van Cleve Cardiac Regeneration Program, Center for Regenerative Medicine, the Division of Hand Surgery, Department of Orthopedic Surgery, and the Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic; and the Division of Cardiovascular Diseases, Department of Medicine, Mayo Clinic College of Medicine
| | - Christopher Livia
- From the Division of Orthopedic Research, Department of Orthopedic Surgery, the Department of Biochemistry and Molecular Biology, the Division of Plastic and Reconstructive Surgery, the Van Cleve Cardiac Regeneration Program, Center for Regenerative Medicine, the Division of Hand Surgery, Department of Orthopedic Surgery, and the Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic; and the Division of Cardiovascular Diseases, Department of Medicine, Mayo Clinic College of Medicine
| | - Tyler Rolland
- From the Division of Orthopedic Research, Department of Orthopedic Surgery, the Department of Biochemistry and Molecular Biology, the Division of Plastic and Reconstructive Surgery, the Van Cleve Cardiac Regeneration Program, Center for Regenerative Medicine, the Division of Hand Surgery, Department of Orthopedic Surgery, and the Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic; and the Division of Cardiovascular Diseases, Department of Medicine, Mayo Clinic College of Medicine
| | - Timothy Peterson
- From the Division of Orthopedic Research, Department of Orthopedic Surgery, the Department of Biochemistry and Molecular Biology, the Division of Plastic and Reconstructive Surgery, the Van Cleve Cardiac Regeneration Program, Center for Regenerative Medicine, the Division of Hand Surgery, Department of Orthopedic Surgery, and the Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic; and the Division of Cardiovascular Diseases, Department of Medicine, Mayo Clinic College of Medicine
| | - Michael S Sabbah
- From the Division of Orthopedic Research, Department of Orthopedic Surgery, the Department of Biochemistry and Molecular Biology, the Division of Plastic and Reconstructive Surgery, the Van Cleve Cardiac Regeneration Program, Center for Regenerative Medicine, the Division of Hand Surgery, Department of Orthopedic Surgery, and the Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic; and the Division of Cardiovascular Diseases, Department of Medicine, Mayo Clinic College of Medicine
| | - Soulmaz Boroumand
- From the Division of Orthopedic Research, Department of Orthopedic Surgery, the Department of Biochemistry and Molecular Biology, the Division of Plastic and Reconstructive Surgery, the Van Cleve Cardiac Regeneration Program, Center for Regenerative Medicine, the Division of Hand Surgery, Department of Orthopedic Surgery, and the Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic; and the Division of Cardiovascular Diseases, Department of Medicine, Mayo Clinic College of Medicine
| | - Tiam M Saffari
- From the Division of Orthopedic Research, Department of Orthopedic Surgery, the Department of Biochemistry and Molecular Biology, the Division of Plastic and Reconstructive Surgery, the Van Cleve Cardiac Regeneration Program, Center for Regenerative Medicine, the Division of Hand Surgery, Department of Orthopedic Surgery, and the Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic; and the Division of Cardiovascular Diseases, Department of Medicine, Mayo Clinic College of Medicine
| | - Atta Behfar
- From the Division of Orthopedic Research, Department of Orthopedic Surgery, the Department of Biochemistry and Molecular Biology, the Division of Plastic and Reconstructive Surgery, the Van Cleve Cardiac Regeneration Program, Center for Regenerative Medicine, the Division of Hand Surgery, Department of Orthopedic Surgery, and the Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic; and the Division of Cardiovascular Diseases, Department of Medicine, Mayo Clinic College of Medicine
| | - Alexander Y Shin
- From the Division of Orthopedic Research, Department of Orthopedic Surgery, the Department of Biochemistry and Molecular Biology, the Division of Plastic and Reconstructive Surgery, the Van Cleve Cardiac Regeneration Program, Center for Regenerative Medicine, the Division of Hand Surgery, Department of Orthopedic Surgery, and the Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic; and the Division of Cardiovascular Diseases, Department of Medicine, Mayo Clinic College of Medicine
| | - Peter C Amadio
- From the Division of Orthopedic Research, Department of Orthopedic Surgery, the Department of Biochemistry and Molecular Biology, the Division of Plastic and Reconstructive Surgery, the Van Cleve Cardiac Regeneration Program, Center for Regenerative Medicine, the Division of Hand Surgery, Department of Orthopedic Surgery, and the Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic; and the Division of Cardiovascular Diseases, Department of Medicine, Mayo Clinic College of Medicine
| |
Collapse
|
7
|
Investigation of the miRNA146a and miRNA155 gene expression levels in patients with multiple sclerosis. J Clin Neurosci 2020; 78:189-193. [PMID: 32331943 DOI: 10.1016/j.jocn.2020.04.071] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 04/12/2020] [Indexed: 12/22/2022]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease and the most common neurodegenerative status. MicroRNAs play an important role in macrophage response to inflammatory processes, and alterations in miRNA levels trigger the inactivation of specific T lymphocytes. As a result, these factors can lead to autoimmune diseases such as MS. Therefore, to determine the role of MicroRNA-146a and MicroRNA-155 in MS patients, their expression levels in serum of MS patients were compared with healthy controls. In this study, the expression levels of MicroRNA-146a and MicroRNA-155 in 30 serum samples of MS and healthy patients as a control group. MicroRNA extraction and cDNA synthesis was performed according manufacture protocols. The expression levels of MicroRNAs were evaluated by Real Time-PCR. MicroRNA-146a and MicroRNA-155 levels were increased in patients with MS compared to controls. The results demonstrated that EDSS score are increased with increasing level of MicroRNA-146a and MicroRNA-155. ROC curve analysis showed that the area under curve (AUC) was significant for MicroRNA-146a and MicroRNA-155. Increased expression levels of MicroRNA-146a and MicroRNA-155 may be associated with the pathogenesis of MS disease. If this study is conducted in a larger sample population and the above results can be used to identify patients or control patients who are under medical care.
Collapse
|
8
|
Aprato J, Sock E, Weider M, Elsesser O, Fröb F, Wegner M. Myrf guides target gene selection of transcription factor Sox10 during oligodendroglial development. Nucleic Acids Res 2020; 48:1254-1270. [PMID: 31828317 PMCID: PMC7026603 DOI: 10.1093/nar/gkz1158] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 11/25/2019] [Accepted: 11/28/2019] [Indexed: 12/13/2022] Open
Abstract
Oligodendrocytes generate myelin in the vertebrate central nervous system and thus ensure rapid propagation of neuronal activity. Their development is controlled by a network of transcription factors that function as determinants of cell identity or as temporally restricted stage-specific regulators. The continuously expressed Sox10 and Myrf, a factor induced during late development, are particularly important for terminal differentiation. How these factors function together mechanistically and influence each other, is not well understood. Here we show that Myrf not only cooperates with Sox10 during the induction of genes required for differentiation and myelin formation. Myrf also inhibits the activity of Sox10 on genes that are essential during earlier phases of oligodendroglial development. By characterization of the exact DNA-binding requirements of Myrf, we furthermore show that cooperative activation is a consequence of joint binding of Sox10 and Myrf to the same regulatory regions. In contrast, inhibition of Sox10-dependent gene activation occurs on genes that lack Myrf binding sites and likely involves physical interaction between Myrf and Sox10 followed by sequestration. These two opposite activities allow Myrf to redirect Sox10 from genes that it activates in oligodendrocyte precursor cells to genes that need to be induced during terminal differentiation.
Collapse
Affiliation(s)
- Jessica Aprato
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Elisabeth Sock
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Matthias Weider
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Olga Elsesser
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Franziska Fröb
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Michael Wegner
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- To whom correspondence should be addressed. Tel: +49 9131 85 24620;
| |
Collapse
|
9
|
Pantera H, Shy ME, Svaren J. Regulating PMP22 expression as a dosage sensitive neuropathy gene. Brain Res 2019; 1726:146491. [PMID: 31586623 DOI: 10.1016/j.brainres.2019.146491] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 09/30/2019] [Accepted: 10/01/2019] [Indexed: 12/21/2022]
Abstract
Structural variation in the human genome has emerged as a major cause of disease as genomic data have accumulated. One of the most common structural variants associated with human disease causes the heritable neuropathy known as Charcot-Marie-Tooth (CMT) disease type 1A. This 1.4 Mb duplication causes nearly half of the CMT cases that are genetically diagnosed. The PMP22 gene is highly induced in Schwann cells during development, although its precise role in myelin formation and homeostasis is still under active investigation. The PMP22 gene can be considered as a nucleoprotein complex with enzymatic activity to produce the PMP22 transcript, and the complex is allosterically regulated by transcription factors that respond to intracellular signals and epigenomic modifications. The control of PMP22 transcript levels has been one of the major therapeutic targets of therapy development, and this review summarizes those approaches as well as efforts to characterize the regulation of the PMP22 gene.
Collapse
Affiliation(s)
- Harrison Pantera
- Molecular and Cellular Pharmacology Training Program, University of Wisconsin, Madison, WI, USA
| | - Michael E Shy
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - John Svaren
- Waisman Center and Department of Comparative Biosciences, University of Wisconsin, Madison, WI, USA.
| |
Collapse
|
10
|
Sock E, Wegner M. Transcriptional control of myelination and remyelination. Glia 2019; 67:2153-2165. [PMID: 31038810 DOI: 10.1002/glia.23636] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 04/01/2019] [Accepted: 04/11/2019] [Indexed: 12/11/2022]
Abstract
Myelination is an evolutionary recent differentiation program that has been independently acquired in vertebrates by Schwann cells in the peripheral nervous system and oligodendrocytes in the central nervous system. Therefore, it is not surprising that regulating transcription factors differ substantially between both cell types. However, overall principles are similar as transcriptional control in Schwann cells and oligodendrocytes combines lineage determining and stage-specific factors in complex regulatory networks. Myelination does not only occur during development, but also as remyelination in the adult. In line with the different conditions during developmental myelination and remyelination and the distinctive properties of Schwann cells and oligodendrocytes, transcriptional regulation of remyelination exhibits unique features and differs between the two cell types. This review gives an overview of the current state in the field.
Collapse
Affiliation(s)
- Elisabeth Sock
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Michael Wegner
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
11
|
Faroni A, Melfi S, Castelnovo LF, Bonalume V, Colleoni D, Magni P, Araúzo-Bravo MJ, Reinbold R, Magnaghi V. GABA-B1 Receptor-Null Schwann Cells Exhibit Compromised In Vitro Myelination. Mol Neurobiol 2018; 56:1461-1474. [PMID: 29948947 DOI: 10.1007/s12035-018-1158-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 05/28/2018] [Indexed: 12/11/2022]
Abstract
GABA-B receptors are important for Schwann cell (SC) commitment to a non-myelinating phenotype during development. However, the P0-GABA-B1fl/fl conditional knockout mice, lacking the GABA-B1 receptor specifically in SCs, also presented axon modifications, suggesting SC non-autonomous effects through the neuronal compartment. In this in vitro study, we evaluated whether the specific deletion of the GABA-B1 receptor in SCs may induce autonomous or non-autonomous cross-changes in sensory dorsal root ganglia (DRG) neurons. To this end, we performed an in vitro biomolecular and transcriptomic analysis of SC and DRG neuron primary cultures from P0-GABA-B1fl/fl mice. We found that cells from conditional P0-GABA-B1fl/fl mice exhibited proliferative, migratory and myelinating alterations. Moreover, we found transcriptomic changes in novel molecules that are involved in peripheral neuron-SC interaction.
Collapse
Affiliation(s)
- Alessandro Faroni
- Blond McIndoe Laboratories, Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Simona Melfi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via G. Balzaretti 9, 20133, Milan, Italy
| | - Luca Franco Castelnovo
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via G. Balzaretti 9, 20133, Milan, Italy
| | - Veronica Bonalume
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via G. Balzaretti 9, 20133, Milan, Italy
| | - Deborah Colleoni
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via G. Balzaretti 9, 20133, Milan, Italy
| | - Paolo Magni
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via G. Balzaretti 9, 20133, Milan, Italy
| | - Marcos J Araúzo-Bravo
- Computational Biology and Systems Biomedicine, Biodonostia Health Research Institute, San Sebastián, Spain.,Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Rolland Reinbold
- Institute of Biomedical Technologies, National Research Council, Milan, Italy
| | - Valerio Magnaghi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via G. Balzaretti 9, 20133, Milan, Italy.
| |
Collapse
|
12
|
Lin HP, Oksuz I, Svaren J, Awatramani R. Egr2-dependent microRNA-138 is dispensable for peripheral nerve myelination. Sci Rep 2018; 8:3817. [PMID: 29491350 PMCID: PMC5830491 DOI: 10.1038/s41598-018-22010-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 02/14/2018] [Indexed: 12/27/2022] Open
Abstract
Recent studies have elucidated the crucial role for microRNAs in peripheral nerve myelination by ablating components of the microRNA synthesis machinery. Few studies have focused on the role of individual microRNAs. To fill this gap, we focused this study on miR-138, which was shown to be drastically reduced in Dicer1 and Dgcr8 knockout mice with hypomyelinating phenotypes and to potentially target the negative regulators of Schwann cell differentiation. Here, we show that of two miR-138 encoding loci, mir-138-1 is the predominant locus transcribed in Schwann cells. mir-138-1 is transcriptionally upregulated during myelination and downregulated upon nerve injury. EGR2 is required for mir-138-1 transcription during development, and both SOX10 and EGR2 bind to an active enhancer near the mir-138-1 locus. Based on expression analyses, we hypothesized that miR-138 facilitates the transition between undifferentiated Schwann cells and myelinating Schwann cells. However, in conditional knockouts, we could not detect significant changes in Schwann cell proliferation, cell cycle exit, or myelination. Overall, our results demonstrate that miR-138 is an Egr2-dependent microRNA but is dispensable for Schwann cell myelination.
Collapse
Affiliation(s)
- Hsin-Pin Lin
- Department of Neurology and Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Idil Oksuz
- Department of Neurology and Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - John Svaren
- Waisman Center and Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Rajeshwar Awatramani
- Department of Neurology and Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA.
| |
Collapse
|
13
|
Abstract
The journey of Schwann cells from their origin in the neural crest to their ensheathment and myelination of peripheral nerves is a remarkable one. Their apparent static function in enabling saltatory conduction of mature nerve is not only vital for long-term health of peripheral nerve but also belies an innate capacity of terminally differentiated Schwann cells to radically alter their differentiation status in the face of nerve injury. The transition from migrating neural crest cells to nerve ensheathment, and then myelination of large diameter axons has been characterized extensively and several of the transcriptional networks have been identified. However, transcription factors must also modify chromatin structure during Schwann cell maturation and this review will focus on chromatin modification machinery that is involved in promoting the transition to, and maintenance of, myelinating Schwann cells. In addition, Schwann cells are known to play important regenerative roles after peripheral nerve injury, and information on epigenomic reprogramming of the Schwann cell genome has emerged. Characterization of epigenomic requirements for myelin maintenance and Schwann cell responses to injury will be vital in understanding how the various Schwann cell functions can be optimized to maintain and repair peripheral nerve function.
Collapse
Affiliation(s)
- Ki H Ma
- 1 Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - John Svaren
- 1 Waisman Center, University of Wisconsin-Madison, Madison, WI, USA.,2 Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
14
|
Ghibaudi M, Boido M, Vercelli A. Functional integration of complex miRNA networks in central and peripheral lesion and axonal regeneration. Prog Neurobiol 2017; 158:69-93. [PMID: 28779869 DOI: 10.1016/j.pneurobio.2017.07.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 07/24/2017] [Accepted: 07/28/2017] [Indexed: 01/06/2023]
Abstract
New players are emerging in the game of peripheral and central nervous system injury since their physiopathological mechanisms remain partially elusive. These mechanisms are characterized by several molecules whose activation and/or modification following a trauma is often controlled at transcriptional level. In this scenario, microRNAs (miRNAs/miRs) have been identified as main actors in coordinating important molecular pathways in nerve or spinal cord injury (SCI). miRNAs are small non-coding RNAs whose functionality at network level is now emerging as a new level of complexity. Indeed they can act as an organized network to provide a precise control of several biological processes. Here we describe the functional synergy of some miRNAs in case of SCI and peripheral damage. In particular we show how several small RNAs can cooperate in influencing simultaneously the molecular pathways orchestrating axon regeneration, inflammation, apoptosis and remyelination. We report about the networks for which miRNA-target bindings have been experimentally demonstrated or inferred based on target prediction data: in both cases, the connection between one miRNA and its downstream pathway is derived from a validated observation or is predicted from the literature. Hence, we discuss the importance of miRNAs in some pathological processes focusing on their functional structure as participating in a cooperative and/or convergence network.
Collapse
Affiliation(s)
- M Ghibaudi
- Department of Neuroscience "Rita Levi Montalcini", Neuroscience Institute Cavalieri Ottolenghi, University of Torino, Italian Institute of Neuroscience, Italy.
| | - M Boido
- Department of Neuroscience "Rita Levi Montalcini", Neuroscience Institute Cavalieri Ottolenghi, University of Torino, Italian Institute of Neuroscience, Italy
| | - A Vercelli
- Department of Neuroscience "Rita Levi Montalcini", Neuroscience Institute Cavalieri Ottolenghi, University of Torino, Italian Institute of Neuroscience, Italy
| |
Collapse
|
15
|
Song J, Li X, Li Y, Che J, Li X, Zhao X, Chen Y, Zheng X, Yuan W. Biodegradable and biocompatible cationic polymer delivering microRNA-221/222 promotes nerve regeneration after sciatic nerve crush. Int J Nanomedicine 2017; 12:4195-4208. [PMID: 28652727 PMCID: PMC5473607 DOI: 10.2147/ijn.s132190] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
MicroRNA (miRNA) has great potential to treat a wide range of illnesses by regulating the expression of eukaryotic genes. Biomaterials with high transfection efficiency and low toxicity are needed to deliver miRNA to target cells. In this study, a biodegradable and biocompatible cationic polymer (PDAPEI) was synthetized from low molecular weight polyethyleneimine (PEI1.8kDa) cross-linked with 2,6-pyridinedicarboxaldehyde. PDAPEI showed a lower cytotoxicity and higher transfection efficiency than PEI25kDa in transfecting miR-221/222 into rat Schwann cells (SCs). The upregulation of miR-221/222 in SCs promoted the expression of nerve growth factor and myelin basic protein in vitro. The mouse sciatic nerve crush injury model was used to evaluate the effectiveness of PDAPEI/miR-221/222 complexes for nerve regeneration in vivo. The results of electrophysiological tests, functional assessments, and histological and immunohistochemistry analyses demonstrated that PDAPEI/miR-221/222 complexes significantly promoted nerve regeneration after sciatic nerve crush, specifically enhancing remyelination. All these results show that the use of PDAPEI to deliver miR-221/222 may provide a safe therapeutic means of treating nerve crush injury and may help to overcome the barrier of biomaterial toxicity and low efficiency often encountered during medical intervention.
Collapse
Affiliation(s)
- Jialin Song
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital
- Department of Orthopedics, Shanghai University of Medicine and Health, Shanghai, Sixth People’s Hospital East Campus, Shanghai
| | - Xueyang Li
- Department of Plastic and Reconstructive Surgery, Xuzhou Medical College Affiliated Hospital, Xuzhou, Jiangsu
| | - Yingli Li
- Department of Plastic Surgery, The General Hospital of Jinan Military Command, Jinan, Shandong
- Department of Plastic Surgery, Chang Hai Hospital, Second Military Medical University
| | - Junyi Che
- School of Pharmacy, Shanghai Jiao Tong University
| | - Xiaoming Li
- School of Pharmacy, Shanghai Jiao Tong University
| | | | - Yinghui Chen
- Department of Neurology, Jinshan Hospital, Fudan University, JinShan District, Shanghai, People’s Republic of China
| | - Xianyou Zheng
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital
| | - Weien Yuan
- School of Pharmacy, Shanghai Jiao Tong University
| |
Collapse
|
16
|
Jeon SJ, Ryu JH, Bahn GH. Altered Translational Control of Fragile X Mental Retardation Protein on Myelin Proteins in Neuropsychiatric Disorders. Biomol Ther (Seoul) 2017; 25:231-238. [PMID: 27829268 PMCID: PMC5424632 DOI: 10.4062/biomolther.2016.042] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Revised: 06/28/2016] [Accepted: 07/28/2016] [Indexed: 01/07/2023] Open
Abstract
Myelin is a specialized structure of the nervous system that both enhances electrical conductance and insulates neurons from external risk factors. In the central nervous system, polarized oligodendrocytes form myelin by wrapping processes in a spiral pattern around neuronal axons through myelin-related gene regulation. Since these events occur at a distance from the cell body, post-transcriptional control of gene expression has strategic advantage to fine-tune the overall regulation of protein contents in situ. Therefore, many research interests have been focused to identify RNA binding proteins and their regulatory mechanism in myelinating compartments. Fragile X mental retardation protein (FMRP) is one such RNA binding protein, regulating its target expression by translational control. Although the majority of works on FMRP have been performed in neurons, it is also found in the developing or mature glial cells including oligodendrocytes, where its function is not well understood. Here, we will review evidences suggesting abnormal translational regulation of myelin proteins with accompanying white matter problem and neurological deficits in fragile X syndrome, which can have wider mechanistic and pathological implication in many other neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Se Jin Jeon
- Department of Life and Nanopharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jong Hoon Ryu
- Department of Life and Nanopharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Geon Ho Bahn
- Department of Neuropsychiatry, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
17
|
Castelnovo LF, Bonalume V, Melfi S, Ballabio M, Colleoni D, Magnaghi V. Schwann cell development, maturation and regeneration: a focus on classic and emerging intracellular signaling pathways. Neural Regen Res 2017; 12:1013-1023. [PMID: 28852375 PMCID: PMC5558472 DOI: 10.4103/1673-5374.211172] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The development, maturation and regeneration of Schwann cells (SCs), the main glial cells of the peripheral nervous system, require the coordinate and complementary interaction among several factors, signals and intracellular pathways. These regulatory molecules consist of integrins, neuregulins, growth factors, hormones, neurotransmitters, as well as entire intracellular pathways including protein-kinase A, C, Akt, Erk/MAPK, Hippo, mTOR, etc. For instance, Hippo pathway is overall involved in proliferation, apoptosis, regeneration and organ size control, being crucial in cancer proliferation process. In SCs, Hippo is linked to merlin and YAP/TAZ signaling and it seems to respond to mechanic/physical challenges. Recently, among factors regulating SCs, also the signaling intermediates Src tyrosine kinase and focal adhesion kinase (FAK) proved relevant for SC fate, participating in the regulation of adhesion, motility, migration and in vitro myelination. In SCs, the factors Src and FAK are regulated by the neuroactive steroid allopregnanolone, thus corroborating the importance of this steroid in the control of SC maturation. In this review, we illustrate some old and novel signaling pathways modulating SC biology and functions during the different developmental, mature and regenerative states.
Collapse
Affiliation(s)
- Luca Franco Castelnovo
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Veronica Bonalume
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Simona Melfi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Marinella Ballabio
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Deborah Colleoni
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Valerio Magnaghi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
18
|
Dickey LL, Worne CL, Glover JL, Lane TE, O’Connell RM. MicroRNA-155 enhances T cell trafficking and antiviral effector function in a model of coronavirus-induced neurologic disease. J Neuroinflammation 2016; 13:240. [PMID: 27604627 PMCID: PMC5015201 DOI: 10.1186/s12974-016-0699-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Accepted: 08/20/2016] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND MicroRNAs (miRNAs) are noncoding RNAs that modulate cellular gene expression, primarily at the post-transcriptional level. We sought to examine the functional role of miR-155 in a model of viral-induced neuroinflammation. METHODS Acute encephalomyelitis and immune-mediated demyelination were induced by intracranial injection with the neurotropic JHM strain of mouse hepatitis virus (JHMV) into C57BL/6 miR-155 (+/+) wildtype (WT) mice or miR-155 (-/-) mice. Morbidity and mortality, viral load and immune cell accumulation in the CNS, and spinal cord demyelination were assessed at defined points post-infection. T cells harvested from infected mice were used to examine cytolytic activity, cytokine activity, and expression of certain chemokine receptors. To determine the impact of miR-155 on trafficking, T cells from infected WT or miR-155 (-/-) mice were adoptively transferred into RAG1 (-/-) mice, and T cell accumulation into the CNS was assessed using flow cytometry. Statistical significance was determined using the Mantel-Cox log-rank test or Student's T tests. RESULTS Compared to WT mice, JHMV-infected miR-155 (-/-) mice developed exacerbated disease concomitant with increased morbidity/mortality and an inability to control viral replication within the CNS. In corroboration with increased susceptibility to disease, miR-155 (-/-) mice had diminished CD8(+) T cell responses in terms of numbers, cytolytic activity, IFN-γ secretion, and homing to the CNS that corresponded with reduced expression of the chemokine receptor CXCR3. Both IFN-γ secretion and trafficking were impaired in miR-155 (-/-) , virus-specific CD4(+) T cells; however, expression of the chemokine homing receptors analyzed on CD4(+) cells was not affected. Except for very early during infection, there were not significant differences in macrophage infiltration into the CNS between WT and miR-155 (-/-) JHMV-infected mice, and the severity of demyelination was similar at 14 days p.i. between WT and miR-155 (-/-) JHMV-infected mice. CONCLUSIONS These findings support a novel role for miR-155 in host defense in a model of viral-induced encephalomyelitis. Specifically, miR-155 enhances antiviral T cell responses including cytokine secretion, cytolytic activity, and homing to the CNS in response to viral infection. Further, miR-155 can play either a host-protective or host-damaging role during neuroinflammation depending on the disease trigger.
Collapse
Affiliation(s)
- Laura L. Dickey
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112 USA
| | - Colleen L. Worne
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112 USA
| | - Jessica L. Glover
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112 USA
| | - Thomas E. Lane
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112 USA
| | - Ryan M. O’Connell
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112 USA
| |
Collapse
|
19
|
Wheeler NA, Fuss B. Extracellular cues influencing oligodendrocyte differentiation and (re)myelination. Exp Neurol 2016; 283:512-30. [PMID: 27016069 PMCID: PMC5010977 DOI: 10.1016/j.expneurol.2016.03.019] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 03/03/2016] [Accepted: 03/18/2016] [Indexed: 02/07/2023]
Abstract
There is an increasing number of neurologic disorders found to be associated with loss and/or dysfunction of the CNS myelin sheath, ranging from the classic demyelinating disease, multiple sclerosis, through CNS injury, to neuropsychiatric diseases. The disabling burden of these diseases has sparked a growing interest in gaining a better understanding of the molecular mechanisms regulating the differentiation of the myelinating cells of the CNS, oligodendrocytes (OLGs), and the process of (re)myelination. In this context, the importance of the extracellular milieu is becoming increasingly recognized. Under pathological conditions, changes in inhibitory as well as permissive/promotional cues are thought to lead to an overall extracellular environment that is obstructive for the regeneration of the myelin sheath. Given the general view that remyelination is, even though limited in human, a natural response to demyelination, targeting pathologically 'dysregulated' extracellular cues and their downstream pathways is regarded as a promising approach toward the enhancement of remyelination by endogenous (or if necessary transplanted) OLG progenitor cells. In this review, we will introduce the extracellular cues that have been implicated in the modulation of (re)myelination. These cues can be soluble, part of the extracellular matrix (ECM) or mediators of cell-cell interactions. Their inhibitory and permissive/promotional roles with regard to remyelination as well as their potential for therapeutic intervention will be discussed.
Collapse
Affiliation(s)
- Natalie A Wheeler
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, United States
| | - Babette Fuss
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, United States.
| |
Collapse
|
20
|
Zappulli V, Friis KP, Fitzpatrick Z, Maguire CA, Breakefield XO. Extracellular vesicles and intercellular communication within the nervous system. J Clin Invest 2016; 126:1198-207. [PMID: 27035811 DOI: 10.1172/jci81134] [Citation(s) in RCA: 183] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Extracellular vesicles (EVs, including exosomes) are implicated in many aspects of nervous system development and function, including regulation of synaptic communication, synaptic strength, and nerve regeneration. They mediate the transfer of packets of information in the form of nonsecreted proteins and DNA/RNA protected within a membrane compartment. EVs are essential for the packaging and transport of many cell-fate proteins during development as well as many neurotoxic misfolded proteins during pathogenesis. This form of communication provides another dimension of cellular crosstalk, with the ability to assemble a "kit" of directional instructions made up of different molecular entities and address it to specific recipient cells. This multidimensional form of communication has special significance in the nervous system. How EVs help to orchestrate the wiring of the brain while allowing for plasticity associated with learning and memory and contribute to regeneration and degeneration are all under investigation. Because they carry specific disease-related RNAs and proteins, practical applications of EVs include potential uses as biomarkers and therapeutics. This Review describes our current understanding of EVs and serves as a springboard for future advances, which may reveal new important mechanisms by which EVs in coordinate brain and body function and dysfunction.
Collapse
|
21
|
Tomassy GS, Dershowitz LB, Arlotta P. Diversity Matters: A Revised Guide to Myelination. Trends Cell Biol 2016; 26:135-147. [PMID: 26442841 PMCID: PMC4727993 DOI: 10.1016/j.tcb.2015.09.002] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 08/28/2015] [Accepted: 09/01/2015] [Indexed: 11/28/2022]
Abstract
The evolutionary success of the vertebrate nervous system is largely due to a unique structural feature--the myelin sheath, a fatty envelope that surrounds the axons of neurons. By increasing the speed by which electrical signals travel along axons, myelin facilitates neuronal communication between distant regions of the nervous system. We review the cellular and molecular mechanisms that regulate the development of myelin as well as its homeostasis in adulthood. We discuss how finely tuned neuron-oligodendrocyte interactions are central to myelin formation during development and in the adult, and how these interactions can have profound implications for the plasticity of the adult brain. We also speculate how the functional diversity of both neurons and oligodendrocytes may impact on the myelination process in both health and disease.
Collapse
Affiliation(s)
- Giulio Srubek Tomassy
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA.
| | - Lori Bowe Dershowitz
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Paola Arlotta
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
22
|
Dionne N, Dib S, Finsen B, Denarier E, Kuhlmann T, Drouin R, Kokoeva M, Hudson TJ, Siminovitch K, Friedman HC, Peterson AC. Functional organization of anMbpenhancer exposes striking transcriptional regulatory diversity within myelinating glia. Glia 2015; 64:175-94. [DOI: 10.1002/glia.22923] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 09/04/2015] [Accepted: 09/09/2015] [Indexed: 11/11/2022]
Affiliation(s)
- Nancy Dionne
- Laboratory of Developmental Biology; Ludmer Research and Training Building, McGill University; Montreal Quebec Canada
| | - Samar Dib
- Laboratory of Developmental Biology; Ludmer Research and Training Building, McGill University; Montreal Quebec Canada
| | - Bente Finsen
- Department of Neurobiology Research; Institute of Molecular Medicine, University of Southern Denmark; Odense Denmark
| | - Eric Denarier
- Institut National De La Santé Et De La Recherche Médicale, U836-GIN iRTSV-GPC; Site Santé La Tronche, BP170 Grenoble Cedex 9 France
| | - Tanja Kuhlmann
- Institute of Neuropathology, University Hospital, Münster; Pottkamp 2 Münster Germany
| | - Régen Drouin
- Division of Genetics, Department of Pediatrics, Faculty of Medicine and Health Sciences; Université De Sherbrooke; Sherbrooke Quebec Canada
| | - Maia Kokoeva
- Department of Medicine; McGill University/MUHC Research Institute; Montreal Quebec Canada
| | - Thomas J. Hudson
- Ontario Institute for Cancer Research, MaRS Centre; South Tower Toronto Ontario Canada
| | - Kathy Siminovitch
- Department of Medicine; University of Toronto, Samuel Lunenfeld and Toronto General Research Institutes; Toronto Ontario Canada
- Department of Immunology and Molecular Genetics; University of Toronto; Toronto Ontario Canada
| | - Hana C Friedman
- Laboratory of Developmental Biology; Ludmer Research and Training Building, McGill University; Montreal Quebec Canada
| | - Alan C. Peterson
- Laboratory of Developmental Biology; Ludmer Research and Training Building, McGill University; Montreal Quebec Canada
| |
Collapse
|
23
|
The Lin28/let-7 axis is critical for myelination in the peripheral nervous system. Nat Commun 2015; 6:8584. [PMID: 26466203 PMCID: PMC4634210 DOI: 10.1038/ncomms9584] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 09/04/2015] [Indexed: 12/31/2022] Open
Abstract
MicroRNAs (miRNAs) are crucial regulators of myelination in the peripheral nervous system (PNS). However, the miRNAs species involved and the underlying mechanisms are largely unknown. We found that let-7 miRNAs are highly abundant during PNS myelination and that their levels are inversely correlated to the expression of lin28 homolog B (Lin28B), an antagonist of let-7 accumulation. Sustained expression of Lin28B and consequently reduced levels of let-7 miRNAs results in a failure of Schwann cell myelination in transgenic mouse models and in cell culture. Subsequent analyses revealed that let-7 miRNAs promote expression of the myelination-driving master transcription factor Krox20 (also known as Egr2) through suppression of myelination inhibitory Notch signalling. We conclude that the Lin28B/let-7 axis acts as a critical driver of PNS myelination, in particular by regulating myelination onset, identifying this pathway also as a potential therapeutic target in demyelinating diseases.
Collapse
|
24
|
MicroRNA delivery for regenerative medicine. Adv Drug Deliv Rev 2015; 88:108-22. [PMID: 26024978 DOI: 10.1016/j.addr.2015.05.014] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 05/13/2015] [Accepted: 05/21/2015] [Indexed: 12/26/2022]
Abstract
MicroRNA (miRNA) directs post-transcriptional regulation of a network of genes by targeting mRNA. Although relatively recent in development, many miRNAs direct differentiation of various stem cells including induced pluripotent stem cells (iPSCs), a major player in regenerative medicine. An effective and safe delivery of miRNA holds the key to translating miRNA technologies. Both viral and nonviral delivery systems have seen success in miRNA delivery, and each approach possesses advantages and disadvantages. A number of studies have demonstrated success in augmenting osteogenesis, improving cardiogenesis, and reducing fibrosis among many other tissue engineering applications. A scaffold-based approach with the possibility of local and sustained delivery of miRNA is particularly attractive since the physical cues provided by the scaffold may synergize with the biochemical cues induced by miRNA therapy. Herein, we first briefly cover the application of miRNA to direct stem cell fate via replacement and inhibition therapies, followed by the discussion of the promising viral and nonviral delivery systems. Next we present the unique advantages of a scaffold-based delivery in achieving lineage-specific differentiation and tissue development.
Collapse
|
25
|
Abstract
Peripheral nerve injuries remain problematic to treat, with poor functional recovery commonly observed. Injuries resulting in a nerve gap create specific difficulties for axonal regeneration. Approaches to address these difficulties include autologous nerve grafts (which are currently the gold standard treatment) and synthetic conduits, with the latter option being able to be impregnated with Schwann cells or stem cells which provide an appropriate micro-environment for neuronal regeneration to occur. Transplanting stem cells, however, infers additional risk of malignant transformation as well as manufacturing difficulties and ethical concerns, and the use of autologous nerve grafts and Schwann cells requires the sacrifice of a functioning nerve. A new approach utilizing exosomes, secreted extracellular vesicles, could avoid these complications. In this review, we summarize the current literature on exosomes, and suggest how they could help to improve axonal regeneration following peripheral nerve injury.
Collapse
Affiliation(s)
- Rosanna C Ching
- Department of Integrative Medical Biology, Umeå University, Umeå, SE-901 87, Sweden ; Department of Surgical & Perioperative Sciences, Umeå University, Umeå, SE-901 87, Sweden
| | - Paul J Kingham
- Department of Integrative Medical Biology, Umeå University, Umeå, SE-901 87, Sweden
| |
Collapse
|