1
|
Kocot J, Kosa P, Ashida S, Pirjanian NA, Goldbach-Mansky R, Peterson K, Fossati V, Holland SM, Bielekova B. Clemastine fumarate accelerates accumulation of disability in progressive multiple sclerosis by enhancing pyroptosis. J Clin Invest 2025; 135:e183941. [PMID: 40371642 PMCID: PMC12077908 DOI: 10.1172/jci183941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 03/12/2025] [Indexed: 05/16/2025] Open
Abstract
Multiple sclerosis (MS) is an immune-mediated demyelinating disease of the CNS. Clemastine fumarate, the over-the-counter antihistamine and muscarinic receptor blocker, has remyelinating potential in MS. A clemastine arm was added to an ongoing platform clinical trial, targeting residual activity by precision, biomarker-guided combination therapies of multiple sclerosis (TRAP-MS) (ClinicalTrials.gov NCT03109288), to identify a cerebrospinal fluid (CSF) remyelination signature and to collect safety data on clemastine in patients progressing independently of relapse activity (PIRA). The clemastine arm was stopped per protocol-defined criteria when 3 of 9 patients triggered individual safety stopping criteria. Clemastine-treated patients had significantly higher treatment-induced disability progression slopes compared with the remaining TRAP-MS participants. Quantification of approximately 7,000 proteins in CSF samples collected before and after clemastine treatment showed significant increases in purinergic signaling and pyroptosis. Mechanistic studies showed that clemastine with sublytic doses of extracellular adenosine triphosphate (ATP) activates inflammasome and induces pyroptotic cell death in macrophages. Clemastine with ATP also caused pyroptosis of induced pluripotent stem cell-derived human oligodendrocytes. Antagonist of the purinergic channel P2RX7, which is strongly expressed in oligodendrocytes and myeloid cells, blocked these toxic effects of clemastine. Finally, reanalysis of published single-nucleus RNA-Seq (snRNA-Seq) studies revealed increased P2RX7 expression and pyroptosis transcriptional signature in microglia and oligodendrocytes in the MS brain, especially in chronic active lesions. The CSF proteomic pyroptosis score was increased in untreated MS patients, was higher in patients with progressive than relapsing-remitting disease, and correlated significantly with the rates of MS progression. Collectively, this identifies pyroptosis as a likely mechanism of CNS injury underlying PIRA even outside of clemastine toxicity.
Collapse
Affiliation(s)
- Joanna Kocot
- Neuroimmunological Diseases Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Peter Kosa
- Neuroimmunological Diseases Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Shinji Ashida
- Neuroimmunological Diseases Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | | | - Raphaela Goldbach-Mansky
- Translational Autoinflammatory Diseases Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Karin Peterson
- Neuroimmunology Section, Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, NIH, Hamilton, Montana, USA
| | - Valentina Fossati
- The New York Stem Cell Foundation Research Institute, New York, New York, USA
| | - Steven M. Holland
- Immunopathogenesis Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Bibiana Bielekova
- Neuroimmunological Diseases Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| |
Collapse
|
2
|
Askari H, Rabiei F, Lohrasbi F, Ghadir S, Mehdipour Arbastan A, Ghasemi-Kasman M. AMP-activated protein kinase as a mediator of mitochondrial dysfunction of multiple sclerosis in animal models: A systematic review. J Cell Physiol 2024; 239:e31230. [PMID: 38403972 DOI: 10.1002/jcp.31230] [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: 11/14/2023] [Revised: 02/12/2024] [Accepted: 02/15/2024] [Indexed: 02/27/2024]
Abstract
Multiple sclerosis (MS) is a chronic central nervous system (CNS) disorder characterized by demyelination, neuronal damage, and oligodendrocyte depletion. Reliable biomarkers are essential for early diagnosis and disease management. Emerging research highlights the role of mitochondrial dysfunction and oxidative stress in CNS disorders, including MS, in which mitochondria are central to the degenerative process. Adenosine monophosphate-activated protein kinase (AMPK) regulates the mitochondrial energy balance and initiates responses in neurodegenerative conditions. This systematic review, following Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines, aimed to comprehensively assess the literature on AMPK pathways, mitochondrial dysfunction, and in vivo studies using MS animal models. The search strategy involved the use of AMPK syntaxes, MS syntaxes, and animal model syntaxes. The PubMed, Scopus, Web of Science, and Google Scholar databases were systematically searched on August 26, 2023 without publication year restrictions. The review identified and analyzed relevant papers to provide a comprehensive overview of the current state of related research. Eight studies utilizing various interventions and methodological approaches were included. Risk of bias assessment revealed some areas of low risk but lacked explicit reporting in others. These studies collectively revealed a complex relationship between AMPK, mitochondrial dysfunction, and MS pathogenesis, with both cuprizone and experimental autoimmune encephalomyelitis models demonstrating associations between AMPK and mitochondrial disorders, including oxidative stress and impaired expression of mitochondrial genes. These studies illuminate the multifaceted role of AMPK in MS animal models, involving energy metabolism, inflammatory processes, oxidative stress, and gene regulation leading to mitochondrial dysfunction. However, unanswered questions about its mechanisms and clinical applications underscore the need for further research to fully harness its potential in addressing MS-related mitochondrial dysfunction.
Collapse
Affiliation(s)
- Hamid Askari
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Fatemeh Rabiei
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Fatemeh Lohrasbi
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Sara Ghadir
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Ahmad Mehdipour Arbastan
- School of Medicine, Faculty of Medical Sciences, Islamic Azad University, Tonekabon Branch, Tonekabon, Iran
| | - Maryam Ghasemi-Kasman
- Cellular and Molecular Biology Research Center, Babol University of Medical Sciences, Babol, Iran
- Department of Physiology, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
3
|
LeVine SM. Exploring Potential Mechanisms Accounting for Iron Accumulation in the Central Nervous System of Patients with Alzheimer's Disease. Cells 2024; 13:689. [PMID: 38667304 PMCID: PMC11049304 DOI: 10.3390/cells13080689] [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: 04/04/2024] [Revised: 04/12/2024] [Accepted: 04/14/2024] [Indexed: 04/28/2024] Open
Abstract
Elevated levels of iron occur in both cortical and subcortical regions of the CNS in patients with Alzheimer's disease. This accumulation is present early in the disease process as well as in more advanced stages. The factors potentially accounting for this increase are numerous, including: (1) Cells increase their uptake of iron and reduce their export of iron, as iron becomes sequestered (trapped within the lysosome, bound to amyloid β or tau, etc.); (2) metabolic disturbances, such as insulin resistance and mitochondrial dysfunction, disrupt cellular iron homeostasis; (3) inflammation, glutamate excitotoxicity, or other pathological disturbances (loss of neuronal interconnections, soluble amyloid β, etc.) trigger cells to acquire iron; and (4) following neurodegeneration, iron becomes trapped within microglia. Some of these mechanisms are also present in other neurological disorders and can also begin early in the disease course, indicating that iron accumulation is a relatively common event in neurological conditions. In response to pathogenic processes, the directed cellular efforts that contribute to iron buildup reflect the importance of correcting a functional iron deficiency to support essential biochemical processes. In other words, cells prioritize correcting an insufficiency of available iron while tolerating deposited iron. An analysis of the mechanisms accounting for iron accumulation in Alzheimer's disease, and in other relevant neurological conditions, is put forward.
Collapse
Affiliation(s)
- Steven M LeVine
- Department of Cell Biology and Physiology, University of Kansas Medical Center, 3901 Rainbow Blvd., Mail Stop 3043, Kansas City, KS 66160, USA
| |
Collapse
|
4
|
Kocot J, Kosa P, Ashida S, Pirjanian N, Goldbach-Mansky R, Peterson K, Fossati V, Holland SM, Bielekova B. Clemastine fumarate accelerates accumulation of disability in progressive multiple sclerosis by enhancing pyroptosis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.04.09.24305506. [PMID: 39802756 PMCID: PMC11722480 DOI: 10.1101/2024.04.09.24305506] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/21/2025]
Abstract
Multiple sclerosis (MS) is an immune-mediated demyelinating disease of the central nervous system (CNS). Clemastine fumarate, the over-the-counter antihistamine and muscarinic receptor blocker, has remyelinating potential in MS. A clemastine arm was added to an ongoing platform clinical trial TRAP-MS (NCT03109288) to identify a cerebrospinal fluid (CSF) remyelination signature and to collect safety data on clemastine in patients progressing independently of relapse activity (PIRA). The clemastine arm was stopped per protocol-defined criteria when 3/9 patients triggered individual safety stopping criteria (χ2 p=0.00015 compared to remaining TRAP-MS treatments). Clemastine treated patients had significantly higher treatment-induced disability progression slopes compared to remaining TRAP-MS participants (p=0.0075). Quantification of ~7000 proteins in CSF samples collected before and after clemastine treatment showed significant increase in purinergic/ATP signaling and pyroptosis cell death. Mechanistic studies showed that clemastine with sub-lytic doses of extracellular ATP activates inflammasome and induces pyroptotic cell death in macrophages. Clemastine with ATP also caused pyroptosis of induced pluripotent stem cell-derived human oligodendrocytes. Antagonist of the purinergic channel P2RX7 that is strongly expressed in oligodendrocytes and myeloid cells, blocked these toxic effects of clemastine. Finally, re-analyses of published snRNAseq studies revealed increased P2RX7 expression and pyroptosis transcriptional signature in microglia and oligodendrocytes in MS brain, especially in chronic active lesions. CSF proteomic pyroptosis score was increased in untreated MS patients, was higher in patients with progressive than relapsing-remitting disease and correlated significantly with rates of MS progression. Thus, pyroptosis is likely first well-characterized mechanism of CNS injury underlying PIRA even outside of clemastine toxicity.
Collapse
Affiliation(s)
- Joanna Kocot
- Neuroimmunological Diseases Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
| | - Peter Kosa
- Neuroimmunological Diseases Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
| | - Shinji Ashida
- Neuroimmunological Diseases Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
| | - Nicolette Pirjanian
- The New York Stem Cell Foundation Research Institute; New York, NY 10019, USA
| | - Raphaela Goldbach-Mansky
- Translational Autoinflammatory Diseases Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
| | - Karin Peterson
- Neuroimmunology Section, Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Hamilton, MT, USA
| | - Valentina Fossati
- The New York Stem Cell Foundation Research Institute; New York, NY 10019, USA
| | - Steven M. Holland
- Immunopathogenesis Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
| | - Bibiana Bielekova
- Neuroimmunological Diseases Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
| |
Collapse
|
5
|
Varshney V, Kumar A, Parashar V, Kumar A, Goyal A, Garabadu D. Therapeutic Potential of Capsaicin in Various Neurodegenerative Diseases with Special Focus on Nrf2 Signaling. Curr Pharm Biotechnol 2024; 25:1693-1707. [PMID: 38173062 DOI: 10.2174/0113892010277933231122111244] [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: 09/02/2023] [Revised: 11/07/2023] [Accepted: 11/10/2023] [Indexed: 01/05/2024]
Abstract
Neurodegenerative disease is mainly characterized by the accumulation of misfolded proteins, contributing to mitochondrial impairments, increased production of proinflammatory cytokines and reactive oxygen species, and neuroinflammation resulting in synaptic loss and neuronal loss. These pathophysiological factors are a serious concern in the treatment of neurodegenerative diseases. Based on the symptoms of various neurodegenerative diseases, different treatments are available, but they have serious side effects and fail in clinical trials, too. Therefore, treatments for neurodegenerative diseases are still a challenge at present. Thus, it is important to study an alternative option. Capsaicin is a naturally occurring alkaloid found in capsicum. Besides the TRPV1 receptor activator in nociception, capsaicin showed a protective effect in brain-related disorders. Capsaicin also reduces the aggregation of misfolded proteins, improves mitochondrial function, and decreases ROS generation. Its antioxidant role is due to increased expression of an nrf2-mediated signaling pathway. Nrf2 is a nuclear erythroid 2-related factor, a transcription factor, which has a crucial role in maintaining the normal function of mitochondria and the cellular defense system against oxidative stress. Intriguingly, Nrf2 mediated pathway improved the upregulation of antioxidant genes and inhibition of microglial-induced inflammation, improved mitochondrial resilience and functions, leading to decreased ROS in neurodegenerative conditions, suggesting that Nrf2 activation could be a better therapeutic approach to target pathophysiology of neurodegenerative disease. Therefore, the present review has evaluated the potential role of capsaicin as a pharmacological agent for the treatment and management of various neurodegenerative diseases via the Nrf2-mediated signaling pathway.
Collapse
Affiliation(s)
- Vibhav Varshney
- Department of Pharmacology, Institute of Pharmaceutical Research, GLA University, Mathura-281406, Uttar Pradesh, India
| | - Abhishek Kumar
- Department of Pharmacology, Institute of Pharmaceutical Research, GLA University, Mathura-281406, Uttar Pradesh, India
| | - Vikas Parashar
- Department of Pharmacology, Institute of Pharmaceutical Research, GLA University, Mathura-281406, Uttar Pradesh, India
| | - Ankit Kumar
- Department of Pharmacology, Institute of Pharmaceutical Research, GLA University, Mathura-281406, Uttar Pradesh, India
| | - Ahsas Goyal
- Department of Pharmacology, Institute of Pharmaceutical Research, GLA University, Mathura-281406, Uttar Pradesh, India
| | - Debapriya Garabadu
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda- 151001, Punjab, India
| |
Collapse
|
6
|
Wang X, Yang W, Wang L, Zheng L, Choi WS. Platinum-based chemotherapy induces demyelination of Schwann cells in oral squamous cell carcinoma treatment. Toxicol Appl Pharmacol 2023; 481:116751. [PMID: 37944569 DOI: 10.1016/j.taap.2023.116751] [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: 08/17/2023] [Revised: 11/02/2023] [Accepted: 11/03/2023] [Indexed: 11/12/2023]
Abstract
BACKGROUND Cisplatin, carboplatin, and oxaliplatin are the only three platinum-based antineoplastic drugs that have been accepted worldwide for treating various cancers. Up to 83.6% of patients treated with platinum-based antineoplastic drugs will develop chemotherapy-induced peripheral neuropathy (CIPN), manifesting as sensory paresthesias, dysesthesias, and hypoesthesias that can cause significant adverse impact to daily activities. AIM To investigate how these three platinum-based drugs affect mitochondrial function and myelination state of Schwann cells and the signalling pathway involved. METHOD 2 μM Cisplatin, 20 μM carboplatin, and 1 μM oxaliplatin were used to inhibit the growth of CAL-27 by 20% respectively. These drugs were then used to induce chemotherapy-induced peripheral neuropathy in Rat Schwann Cells (RSC96). The changes in cell metabolism and myelin formation in RSC96 were investigated. RESULT Cisplatin and carboplatin, but not oxaliplatin increased intracellular and mitochondrial reactive oxygen species in RSC96. Only Cisplatin and carboplatin decreased mitochondrial membrane potential (ΔΨm) and ATP production in RSC96. Both Cisplatin and carboplatin led to demyelination of RSC96, characterized by increased expression of p75NTR and decreased expression of myelin protein zero (MPZ). CONCLUSION Cisplatin and carboplatin, but not oxaliplatin, caused mitochondrial dysfunction and induced demyelination in RSC96 while showing similar toxicity to head and neck cancer cells. Oxaliplatin may be a potential chemotherapy drug to prevent CIPN in patients with head and neck cancer.
Collapse
Affiliation(s)
- Xian Wang
- Division of Oral and Maxillofacial Surgery, Faculty of Dentistry, The University of Hong Kong, Hong Kong, SAR, China
| | - Weifa Yang
- Division of Oral and Maxillofacial Surgery, Faculty of Dentistry, The University of Hong Kong, Hong Kong, SAR, China
| | - Leilei Wang
- Division of Oral and Maxillofacial Surgery, Faculty of Dentistry, The University of Hong Kong, Hong Kong, SAR, China
| | - Liwu Zheng
- Division of Oral and Maxillofacial Surgery, Faculty of Dentistry, The University of Hong Kong, Hong Kong, SAR, China
| | - Wing Shan Choi
- Division of Oral and Maxillofacial Surgery, Faculty of Dentistry, The University of Hong Kong, Hong Kong, SAR, China.
| |
Collapse
|
7
|
Cabezas Perez RJ, Ávila Rodríguez MF, Rosero Salazar DH. Exogenous Antioxidants in Remyelination and Skeletal Muscle Recovery. Biomedicines 2022; 10:biomedicines10102557. [PMID: 36289819 PMCID: PMC9599955 DOI: 10.3390/biomedicines10102557] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 07/18/2022] [Accepted: 07/25/2022] [Indexed: 11/16/2022] Open
Abstract
Inflammatory, oxidative, and autoimmune responses cause severe damage to the nervous system inducing loss of myelin layers or demyelination. Even though demyelination is not considered a direct cause of skeletal muscle disease there is extensive damage in skeletal muscles following demyelination and impaired innervation. In vitro and in vivo evidence using exogenous antioxidants in models of demyelination is showing improvements in myelin formation alongside skeletal muscle recovery. For instance, exogenous antioxidants such as EGCG stimulate nerve structure maintenance, activation of glial cells, and reduction of oxidative stress. Consequently, this evidence is also showing structural and functional recovery of impaired skeletal muscles due to demyelination. Exogenous antioxidants mostly target inflammatory pathways and stimulate remyelinating mechanisms that seem to induce skeletal muscle regeneration. Therefore, the aim of this review is to describe recent evidence related to the molecular mechanisms in nerve and skeletal muscle regeneration induced by exogenous antioxidants. This will be relevant to identifying further targets to improve treatments of neuromuscular demyelinating diseases.
Collapse
|
8
|
Neurodegeneration in Multiple Sclerosis: The Role of Nrf2-Dependent Pathways. Antioxidants (Basel) 2022; 11:antiox11061146. [PMID: 35740042 PMCID: PMC9219619 DOI: 10.3390/antiox11061146] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/06/2022] [Accepted: 06/08/2022] [Indexed: 12/10/2022] Open
Abstract
Multiple sclerosis (MS) encompasses a chronic, irreversible, and predominantly immune-mediated disease of the central nervous system that leads to axonal degeneration, neuronal death, and several neurological symptoms. Although various immune therapies have reduced relapse rates and the severity of symptoms in relapsing-remitting MS, there is still no cure for this devastating disease. In this brief review, we discuss the role of mitochondria dysfunction in the progression of MS, focused on the possible role of Nrf2 signaling in orchestrating the impairment of critical cellular and molecular aspects such as reactive oxygen species (ROS) management, under neuroinflammation and neurodegeneration in MS. In this scenario, we propose a new potential downstream signaling of Nrf2 pathway, namely the opening of hemichannels and pannexons. These large-pore channels are known to modulate glial/neuronal function and ROS production as they are permeable to extracellular Ca2+ and release potentially harmful transmitters to the synaptic cleft. In this way, the Nrf2 dysfunction impairs not only the bioenergetics and metabolic properties of glial cells but also the proper antioxidant defense and energy supply that they provide to neurons.
Collapse
|
9
|
Liu J, Kelly E, Bielekova B. Current Status and Future Opportunities in Modeling Clinical Characteristics of Multiple Sclerosis. Front Neurol 2022; 13:884089. [PMID: 35720098 PMCID: PMC9198703 DOI: 10.3389/fneur.2022.884089] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 04/25/2022] [Indexed: 12/15/2022] Open
Abstract
Development of effective treatments requires understanding of disease mechanisms. For diseases of the central nervous system (CNS), such as multiple sclerosis (MS), human pathology studies and animal models tend to identify candidate disease mechanisms. However, these studies cannot easily link the identified processes to clinical outcomes, such as MS severity, required for causality assessment of candidate mechanisms. Technological advances now allow the generation of thousands of biomarkers in living human subjects, derived from genes, transcripts, medical images, and proteins or metabolites in biological fluids. These biomarkers can be assembled into computational models of clinical value, provided such models are generalizable. Reproducibility of models increases with the technical rigor of the study design, such as blinding, control implementation, the use of large cohorts that encompass the entire spectrum of disease phenotypes and, most importantly, model validation in independent cohort(s). To facilitate the growth of this important research area, we performed a meta-analysis of publications (n = 302) that model MS clinical outcomes extracting effect sizes, while also scoring the technical quality of the study design using predefined criteria. Finally, we generated a Shiny-App-based website that allows dynamic exploration of the data by selective filtering. On average, the published studies fulfilled only one of the seven criteria of study design rigor. Only 15.2% of the studies used any validation strategy, and only 8% used the gold standard of independent cohort validation. Many studies also used small cohorts, e.g., for magnetic resonance imaging (MRI) and blood biomarker predictors, the median sample size was <100 subjects. We observed inverse relationships between reported effect sizes and the number of study design criteria fulfilled, expanding analogous reports from non-MS fields, that studies that fail to limit bias overestimate effect sizes. In conclusion, the presented meta-analysis represents a useful tool for researchers, reviewers, and funders to improve the design of future modeling studies in MS and to easily compare new studies with the published literature. We expect that this will accelerate research in this important area, leading to the development of robust models with proven clinical value.
Collapse
Affiliation(s)
| | | | - Bibiana Bielekova
- Neuroimmunological Diseases Section (NDS), National Institute of Allergy and Infectious Diseases (NIAID) of the National Institutes of Health (NIH), Bethesda, MD, United States
| |
Collapse
|
10
|
Positron emission tomography in multiple sclerosis - straight to the target. Nat Rev Neurol 2021; 17:663-675. [PMID: 34545219 DOI: 10.1038/s41582-021-00537-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2021] [Indexed: 02/08/2023]
Abstract
Following the impressive progress in the treatment of relapsing-remitting multiple sclerosis (MS), the major challenge ahead is the development of treatments to prevent or delay the irreversible accumulation of clinical disability in progressive forms of the disease. The substrate of clinical progression is neuro-axonal degeneration, and a deep understanding of the mechanisms that underlie this process is a precondition for the development of therapies for progressive MS. PET imaging involves the use of radiolabelled compounds that bind to specific cellular and metabolic targets, thereby enabling direct in vivo measurement of several pathological processes. This approach can provide key insights into the clinical relevance of these processes and their chronological sequence during the disease course. In this Review, we focus on the contribution that PET is making to our understanding of extraneuronal and intraneuronal mechanisms that are involved in the pathogenesis of irreversible neuro-axonal damage in MS. We consider the major challenges with the use of PET in MS and the steps necessary to realize clinical benefits of the technique. In addition, we discuss the potential of emerging PET tracers and future applications of existing compounds to facilitate the identification of effective neuroprotective treatments for patients with MS.
Collapse
|
11
|
Vejux A, Ghzaiel I, Nury T, Schneider V, Charrière K, Sghaier R, Zarrouk A, Leoni V, Moreau T, Lizard G. Oxysterols and multiple sclerosis: Physiopathology, evolutive biomarkers and therapeutic strategy. J Steroid Biochem Mol Biol 2021; 210:105870. [PMID: 33684483 DOI: 10.1016/j.jsbmb.2021.105870] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 03/02/2021] [Indexed: 12/13/2022]
Abstract
Multiple sclerosis is an autoimmune disease that affects the central nervous system. Dysfunction of the immune system leads to lesions that cause motor, sensory, cognitive, visual and/or sphincter disturbances. In the long term, these disorders can progress towards an irreversible handicap. The diagnosis takes time because there are no specific criteria to diagnose multiple sclerosis. To realize the diagnosis, a combination of clinical, biological, and radiological arguments is therefore required. Hence, there is a need to identify multiple sclerosis biomarkers. Some biomarkers target immunity through the detection of oligoclonal bands, the measurement of the IgG index and cytokines. During the physiopathological process, the blood-brain barrier can be broken, and this event can be identified by measuring metalloproteinase activity and diffusion of gadolinium in the brain by magnetic resonance imaging. Markers of demyelination and of astrocyte and microglial activity may also be of interest as well as markers of neuronal damage and mitochondrial status. The measurement of different lipids in the plasma and cerebrospinal fluid can also provide suitable information. These different lipids include fatty acids, fatty acid peroxidation products, phospholipids as well as oxidized derivatives of cholesterol (oxysterols). Oxysterols could constitute new biomarkers providing information on the form of multiple sclerosis, the outcome of the disease and the answer to treatment.
Collapse
Affiliation(s)
- Anne Vejux
- Team Bio-PeroxIL, "Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism" (EA7270), University Bourgogne Franche-Comté, Inserm, Dijon, France.
| | - Imen Ghzaiel
- Team Bio-PeroxIL, "Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism" (EA7270), University Bourgogne Franche-Comté, Inserm, Dijon, France; Faculty of Medicine, LR12ES05, Lab-NAFS "Nutrition - Functional Food & Vascular Health", University of Monastir, Monastir, Tunisia
| | - Thomas Nury
- Team Bio-PeroxIL, "Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism" (EA7270), University Bourgogne Franche-Comté, Inserm, Dijon, France
| | - Vincent Schneider
- Team Bio-PeroxIL, "Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism" (EA7270), University Bourgogne Franche-Comté, Inserm, Dijon, France; University Hospital, Department of Neurology, Dijon, France
| | - Karine Charrière
- Centre Hospitalier Universitaire de Besançon, Centre d'Investigation Clinique, INSERM CIC 1431, 25030, Besançon Cedex, France
| | - Randa Sghaier
- Team Bio-PeroxIL, "Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism" (EA7270), University Bourgogne Franche-Comté, Inserm, Dijon, France
| | - Amira Zarrouk
- Faculty of Medicine, LR12ES05, Lab-NAFS "Nutrition - Functional Food & Vascular Health", University of Monastir, Monastir, Tunisia; Laboratory of Biochemistry, Faculty of Medicine, University of Sousse, Sousse, Tunisia
| | - Valerio Leoni
- Laboratory of Clinical Chemistry, Hospital of Varese, ASST-Settelaghi, Varese, Italy
| | - Thibault Moreau
- Team Bio-PeroxIL, "Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism" (EA7270), University Bourgogne Franche-Comté, Inserm, Dijon, France; University Hospital, Department of Neurology, Dijon, France
| | - Gérard Lizard
- Team Bio-PeroxIL, "Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism" (EA7270), University Bourgogne Franche-Comté, Inserm, Dijon, France.
| |
Collapse
|
12
|
Collorone S, Prados F, Kanber B, Cawley NM, Tur C, Grussu F, Solanky BS, Yiannakas M, Davagnanam I, Wheeler-Kingshott CAMG, Barkhof F, Ciccarelli O, Toosy AT. Brain microstructural and metabolic alterations detected in vivo at onset of the first demyelinating event. Brain 2021; 144:1409-1421. [PMID: 33903905 PMCID: PMC8219367 DOI: 10.1093/brain/awab043] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 11/03/2020] [Accepted: 12/03/2020] [Indexed: 12/22/2022] Open
Abstract
In early multiple sclerosis, a clearer understanding of normal-brain tissue microstructural and metabolic abnormalities will provide valuable insights into its pathophysiology. We used multi-parametric quantitative MRI to detect alterations in brain tissues of patients with their first demyelinating episode. We acquired neurite orientation dispersion and density imaging [to investigate morphology of neurites (dendrites and axons)] and 23Na MRI (to estimate total sodium concentration, a reflection of underlying changes in metabolic function). In this cross-sectional study, we enrolled 42 patients diagnosed with clinically isolated syndrome or multiple sclerosis within 3 months of their first demyelinating event and 16 healthy controls. Physical and cognitive scales were assessed. At 3 T, we acquired brain and spinal cord structural scans, and neurite orientation dispersion and density imaging. Thirty-two patients and 13 healthy controls also underwent brain 23Na MRI. We measured neurite density and orientation dispersion indices and total sodium concentration in brain normal-appearing white matter, white matter lesions, and grey matter. We used linear regression models (adjusting for brain parenchymal fraction and lesion load) and Spearman correlation tests (significance level P ≤ 0.01). Patients showed higher orientation dispersion index in normal-appearing white matter, including the corpus callosum, where they also showed lower neurite density index and higher total sodium concentration, compared with healthy controls. In grey matter, compared with healthy controls, patients demonstrated: lower orientation dispersion index in frontal, parietal and temporal cortices; lower neurite density index in parietal, temporal and occipital cortices; and higher total sodium concentration in limbic and frontal cortices. Brain volumes did not differ between patients and controls. In patients, higher orientation dispersion index in corpus callosum was associated with worse performance on timed walk test (P = 0.009, B = 0.01, 99% confidence interval = 0.0001 to 0.02), independent of brain and lesion volumes. Higher total sodium concentration in left frontal middle gyrus was associated with higher disability on Expanded Disability Status Scale (rs = 0.5, P = 0.005). Increased axonal dispersion was found in normal-appearing white matter, particularly corpus callosum, where there was also axonal degeneration and total sodium accumulation. The association between increased axonal dispersion in the corpus callosum and worse walking performance implies that morphological and metabolic alterations in this structure could mechanistically contribute to disability in multiple sclerosis. As brain volumes were neither altered nor related to disability in patients, our findings suggest that these two advanced MRI techniques are more sensitive at detecting clinically relevant pathology in early multiple sclerosis.
Collapse
Affiliation(s)
- Sara Collorone
- NMR Research Unit, Queen Square MS Centre, Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, UK
| | - Ferran Prados
- NMR Research Unit, Queen Square MS Centre, Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, UK.,Centre for Medical Image Computing (CMIC), Department of Medical Physics and Biomedical Engineering, University College London, London, UK.,Universitat Oberta de Catalunya, Barcelona, Spain
| | - Baris Kanber
- Centre for Medical Image Computing (CMIC), Department of Medical Physics and Biomedical Engineering, University College London, London, UK
| | - Niamh M Cawley
- NMR Research Unit, Queen Square MS Centre, Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, UK
| | - Carmen Tur
- NMR Research Unit, Queen Square MS Centre, Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, UK
| | - Francesco Grussu
- NMR Research Unit, Queen Square MS Centre, Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, UK.,Centre for Medical Image Computing (CMIC), Department of Computer Sciences, University College London, London, UK
| | - Bhavana S Solanky
- NMR Research Unit, Queen Square MS Centre, Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, UK
| | - Marios Yiannakas
- NMR Research Unit, Queen Square MS Centre, Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, UK
| | - Indran Davagnanam
- Department of Brain Repair and Rehabilitation, University College London Institute of Neurology, Faculty of Brain Sciences, UCL, London, UK
| | - Claudia A M Gandini Wheeler-Kingshott
- NMR Research Unit, Queen Square MS Centre, Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, UK.,Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy.,Brain MRI 3T Research Centre, IRCCS Mondino Foundation, Pavia, Italy
| | - Frederik Barkhof
- Centre for Medical Image Computing (CMIC), Department of Medical Physics and Biomedical Engineering, University College London, London, UK.,Department of Brain Repair and Rehabilitation, University College London Institute of Neurology, Faculty of Brain Sciences, UCL, London, UK.,Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Vrije Universiteit, The Netherlands.,National Institute for Health Research, University College London Hospitals, Biomedical Research Centre, London, UK
| | - Olga Ciccarelli
- NMR Research Unit, Queen Square MS Centre, Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, UK.,National Institute for Health Research, University College London Hospitals, Biomedical Research Centre, London, UK
| | - Ahmed T Toosy
- NMR Research Unit, Queen Square MS Centre, Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, UK
| |
Collapse
|
13
|
Ineichen BV, Zhu K, Carlström KE. Axonal mitochondria adjust in size depending on g-ratio of surrounding myelin during homeostasis and advanced remyelination. J Neurosci Res 2020; 99:793-805. [PMID: 33368634 PMCID: PMC7898477 DOI: 10.1002/jnr.24767] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 11/14/2020] [Indexed: 12/11/2022]
Abstract
Demyelinating pathology is common in many neurological diseases such as multiple sclerosis, stroke, and Alzheimer's disease and results in axonal energy deficiency, dysfunctional axonal propagation, and neurodegeneration. During myelin repair and also during myelin homeostasis, mutual regulative processes between axons and myelin sheaths are known to be essential. However, proficient tools are lacking to characterize axon‐myelin interdependence during (re)myelination. Thus, we herein investigated adaptions in myelin sheath g‐ratio as a proxy for myelin thickness and axon metabolic status during homeostasis and myelin repair, by using axonal mitochondrial size as a proxy for axonal metabolic status. We found that axons with thinner myelin sheaths had larger axonal mitochondria; this was true for across different central nervous system tracts as well as across species, including humans. The link between myelin sheath thickness and mitochondrial size was temporarily absent during demyelination but reestablished during advanced remyelination, as shown in two commonly used animal models of toxic demyelination. By further exploring this association in mice with either genetically induced mitochondrial or myelin dysfunction, we show that axonal mitochondrial size adjusts in response to the thickness of the myelin sheath but not vice versa. This pinpoints the relevance of mitochondrial adaptation upon myelin repair and might open a new therapeutic window for remyelinating therapies.
Collapse
Affiliation(s)
- Benjamin V Ineichen
- Department of Clinical Neurosciences, Karolinska Institutet, Center for Molecular Medicine, Karolinska Hospital at Solna, Stockholm, Sweden
| | - Keying Zhu
- Department of Clinical Neurosciences, Karolinska Institutet, Center for Molecular Medicine, Karolinska Hospital at Solna, Stockholm, Sweden
| | - Karl E Carlström
- Department of Clinical Neurosciences, Karolinska Institutet, Center for Molecular Medicine, Karolinska Hospital at Solna, Stockholm, Sweden.,Laboratory of Molecular Neurobiology, Department Medical Biochemistry and Biophysics, Biomedicum, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
14
|
Alcover-Sanchez B, Garcia-Martin G, Escudero-Ramirez J, Gonzalez-Riano C, Lorenzo P, Gimenez-Cassina A, Formentini L, de la Villa-Polo P, Pereira MP, Wandosell F, Cubelos B. Absence of R-Ras1 and R-Ras2 causes mitochondrial alterations that trigger axonal degeneration in a hypomyelinating disease model. Glia 2020; 69:619-637. [PMID: 33010069 DOI: 10.1002/glia.23917] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 08/18/2020] [Accepted: 09/21/2020] [Indexed: 12/11/2022]
Abstract
Fast synaptic transmission in vertebrates is critically dependent on myelin for insulation and metabolic support. Myelin is produced by oligodendrocytes (OLs) that maintain multilayered membrane compartments that wrap around axonal fibers. Alterations in myelination can therefore lead to severe pathologies such as multiple sclerosis. Given that hypomyelination disorders have complex etiologies, reproducing clinical symptoms of myelin diseases from a neurological perspective in animal models has been difficult. We recently reported that R-Ras1-/- and/or R-Ras2-/- mice, which lack GTPases essential for OL survival and differentiation processes, present different degrees of hypomyelination in the central nervous system with a compounded hypomyelination in double knockout (DKO) mice. Here, we discovered that the loss of R-Ras1 and/or R-Ras2 function is associated with aberrant myelinated axons with increased numbers of mitochondria, and a disrupted mitochondrial respiration that leads to increased reactive oxygen species levels. Consequently, aberrant myelinated axons are thinner with cytoskeletal phosphorylation patterns typical of axonal degeneration processes, characteristic of myelin diseases. Although we observed different levels of hypomyelination in a single mutant mouse, the combined loss of function in DKO mice lead to a compromised axonal integrity, triggering the loss of visual function. Our findings demonstrate that the loss of R-Ras function reproduces several characteristics of hypomyelinating diseases, and we therefore propose that R-Ras1-/- and R-Ras2-/- neurological models are valuable approaches for the study of these myelin pathologies.
Collapse
Affiliation(s)
- Berta Alcover-Sanchez
- Departamento de Biología Molecular and Centro Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid - Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Gonzalo Garcia-Martin
- Departamento de Biología Molecular and Centro Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid - Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Juan Escudero-Ramirez
- Departamento de Biología Molecular and Centro Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid - Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Carolina Gonzalez-Riano
- CEMBIO (Centre for Metabolomics and Bioanalysis), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Paz Lorenzo
- CEMBIO (Centre for Metabolomics and Bioanalysis), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Alfredo Gimenez-Cassina
- Departamento de Biología Molecular and Centro Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid - Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Laura Formentini
- Departamento de Biología Molecular and Centro Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid - Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Pedro de la Villa-Polo
- Departamento de Biología de Sistemas, Universidad de Alcalá, Madrid, Spain.,Grupo de Neurofisiología Visual, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Marta P Pereira
- Departamento de Biología Molecular and Centro Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid - Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Francisco Wandosell
- Departamento de Biología Molecular and Centro Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid - Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Beatriz Cubelos
- Departamento de Biología Molecular and Centro Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid - Consejo Superior de Investigaciones Científicas, Madrid, Spain
| |
Collapse
|
15
|
Holman SP, Lobo AS, Novorolsky RJ, Nichols M, Fiander MDJ, Konda P, Kennedy BE, Gujar S, Robertson GS. Neuronal mitochondrial calcium uniporter deficiency exacerbates axonal injury and suppresses remyelination in mice subjected to experimental autoimmune encephalomyelitis. Exp Neurol 2020; 333:113430. [PMID: 32745471 DOI: 10.1016/j.expneurol.2020.113430] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/05/2020] [Accepted: 07/28/2020] [Indexed: 12/11/2022]
Abstract
High-capacity mitochondrial calcium (Ca2+) uptake by the mitochondrial Ca2+ uniporter (MCU) is strategically positioned to support the survival and remyelination of axons in multiple sclerosis (MS) by undocking mitochondria, buffering Ca2+ and elevating adenosine triphosphate (ATP) synthesis at metabolically stressed sites. Respiratory chain deficits in MS are proposed to metabolically compromise axon survival and remyelination by suppressing MCU activity. In support of this hypothesis, clinical scores, mitochondrial dysfunction, myelin loss, axon damage and inflammation were elevated while remyelination was blocked in neuronal MCU deficient (Thy1-MCU Def) mice relative to Thy1 controls subjected to experimental autoimmune encephalomyelitis (EAE). At the first sign of walking deficits, mitochondria in EAE/Thy1 axons showed signs of activation. By contrast, cytoskeletal damage, fragmented mitochondria and large autophagosomes were seen in EAE/Thy1-MCU Def axons. As EAE severity increased, EAE/Thy1 axons were filled with massively swollen mitochondria with damaged cristae while EAE/Thy1-MCU Def axons were riddled with late autophagosomes. ATP concentrations and mitochondrial gene expression were suppressed while calpain activity, autophagy-related gene mRNA levels and autophagosome marker (LC3) co-localization in Thy1-expressing neurons were elevated in the spinal cords of EAE/Thy1-MCU Def compared to EAE/Thy1 mice. These findings suggest that MCU inhibition contributes to axonal damage that drives MS progression.
Collapse
Affiliation(s)
- Scott P Holman
- Department of Pharmacology, Brain Repair Centre, Dalhousie University, 1348 Summer Street, Life Sciences Research Institute, North Tower, Halifax B3H 4R2, Canada; Faculty of Medicine, Dalhousie University, 1348 Summer Street, Life Sciences Research Institute, North Tower, Halifax B3H 4R2, Canada
| | - Aurelio S Lobo
- Department of Pharmacology, Brain Repair Centre, Dalhousie University, 1348 Summer Street, Life Sciences Research Institute, North Tower, Halifax B3H 4R2, Canada; Faculty of Medicine, Dalhousie University, 1348 Summer Street, Life Sciences Research Institute, North Tower, Halifax B3H 4R2, Canada
| | - Robyn J Novorolsky
- Department of Pharmacology, Brain Repair Centre, Dalhousie University, 1348 Summer Street, Life Sciences Research Institute, North Tower, Halifax B3H 4R2, Canada; Faculty of Medicine, Dalhousie University, 1348 Summer Street, Life Sciences Research Institute, North Tower, Halifax B3H 4R2, Canada
| | - Matthew Nichols
- Department of Pharmacology, Brain Repair Centre, Dalhousie University, 1348 Summer Street, Life Sciences Research Institute, North Tower, Halifax B3H 4R2, Canada; Faculty of Medicine, Dalhousie University, 1348 Summer Street, Life Sciences Research Institute, North Tower, Halifax B3H 4R2, Canada
| | - Maximillian D J Fiander
- Department of Pharmacology, Brain Repair Centre, Dalhousie University, 1348 Summer Street, Life Sciences Research Institute, North Tower, Halifax B3H 4R2, Canada; Faculty of Medicine, Dalhousie University, 1348 Summer Street, Life Sciences Research Institute, North Tower, Halifax B3H 4R2, Canada
| | - Prathyusha Konda
- Department of Pathology, Faculty of Medicine, Dalhousie University, 1348 Summer Street, Life Sciences Research Institute, North Tower, Halifax B3H 4R2, Canada
| | - Barry E Kennedy
- Department of Pathology, Faculty of Medicine, Dalhousie University, 1348 Summer Street, Life Sciences Research Institute, North Tower, Halifax B3H 4R2, Canada
| | - Shashi Gujar
- Department of Pathology, Faculty of Medicine, Dalhousie University, 1348 Summer Street, Life Sciences Research Institute, North Tower, Halifax B3H 4R2, Canada
| | - George S Robertson
- Department of Pharmacology, Brain Repair Centre, Dalhousie University, 1348 Summer Street, Life Sciences Research Institute, North Tower, Halifax B3H 4R2, Canada; Faculty of Medicine, Dalhousie University, 1348 Summer Street, Life Sciences Research Institute, North Tower, Halifax B3H 4R2, Canada; Department of Psychiatry, 5909 Veterans' Memorial Lane, 8th Floor, Abbie J. Lane Memorial Building, QEII Health Sciences Centre, Halifax B3H 2E2, Canada.
| |
Collapse
|
16
|
Muke I, Sprenger A, Bobylev I, Wiemer V, Barham M, Neiss WF, Lehmann HC. Ultrastructural characterization of mitochondrial damage in experimental autoimmune neuritis. J Neuroimmunol 2020; 343:577218. [PMID: 32251941 DOI: 10.1016/j.jneuroim.2020.577218] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 03/16/2020] [Accepted: 03/18/2020] [Indexed: 12/11/2022]
Abstract
Data are sparse about mitochondrial damage in GBS and in its most frequently employed animal model, experimental autoimmune neuritis (EAN). We here characterized changes in mitochondrial content and morphology at different time points during EAN by use of ultrastructural imaging and immunofluorescent labelling. Histological examination revealed that demyelinated axons and their adjacent Schwann cells showed reduced mitochondrial content and remaining mitochondria appeared swollen with greater diameter in Schwann cells and unmyelinated axons. Our findings indicate that in EAN, particularly mitochondria in Schwann cells are damaged. Further studies are warranted to address whether these changes are amenable to novel, mitoprotective treatments.
Collapse
Affiliation(s)
- Ines Muke
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Germany
| | - Alina Sprenger
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Germany
| | - Ilja Bobylev
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Germany
| | - Valerie Wiemer
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, Germany
| | - Mohammed Barham
- Department of Anatomy I, Faculty of Medicine, University of Cologne, Germany
| | | | - Helmar Christoph Lehmann
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Germany.
| |
Collapse
|
17
|
Soni N, Vegh V, To XV, Mohamed AZ, Borges K, Nasrallah FA. Combined Diffusion Tensor Imaging and Quantitative Susceptibility Mapping Discern Discrete Facets of White Matter Pathology Post-injury in the Rodent Brain. Front Neurol 2020; 11:153. [PMID: 32210907 PMCID: PMC7067826 DOI: 10.3389/fneur.2020.00153] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 02/18/2020] [Indexed: 12/12/2022] Open
Abstract
Early loss of white matter microstructure integrity is a significant cause of long-term neurological disorders following traumatic brain injury (TBI). White matter abnormalities typically involve axonal loss and demyelination. In-vivo imaging tools to detect and differentiate such microstructural changes are not well-explored. This work utilizes the conjoint potential offered by advanced magnetic resonance imaging techniques, including quantitative susceptibility mapping (QSM) and diffusion tensor imaging (DTI), to discern the underlying white matter pathology at specific time points (5 h, 1, 3, 7, 14, and 30 days) post-injury in the controlled cortical impact mouse model. A total of 42 animals were randomized into six TBI groups (n = 6 per group) and one sham group (n = 6). Histopathology was performed to validate in-vivo findings by performing myelin basic protein (MBP) and glial fibrillary acidic protein (GFAP) immunostaining for the assessment of changes to myelin and astrocytes. After 5 h of injury radial diffusivity (RD) was increased in white matter without a significant change in axial diffusivity (AxD) and susceptibility values. After 1 day post-injury RD was decreased. AxD and susceptibility changes were seen after 3 days post-injury. Susceptibility increases in white matter were observed in both ipsilateral and contralateral regions and persisted for 30 days. In histology, an increase in GFAP immunoreactivity was observed after 3 days post-injury and remained high for 30 days in both ipsilateral and contralateral white matter regions. A loss in MBP signal was noted after 3 days post-injury that continued up to 30 days. In conclusion, these results demonstrate the complementary ability of DTI and QSM in discerning the micro-pathological processes triggered following TBI. While DTI revealed acute and focal white matter changes, QSM mirrored the temporal demyelination in the white matter tracts and diffuse regions at the chronic state.
Collapse
Affiliation(s)
- Neha Soni
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Viktor Vegh
- Center for Advanced Imaging, The University of Queensland, Brisbane, QLD, Australia
| | - Xuan Vinh To
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Abdalla Z Mohamed
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Karin Borges
- Faculty of Medicine, School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Fatima A Nasrallah
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
18
|
Tobore TO. Towards a comprehensive etiopathogenetic and pathophysiological theory of multiple sclerosis. Int J Neurosci 2019; 130:279-300. [PMID: 31588832 DOI: 10.1080/00207454.2019.1677648] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Background: Multiple sclerosis (MS) is a neurodegenerative disease caused by dysfunction of the immune system that affects the central nervous system (CNS). It is characterized by demyelination, chronic inflammation, neuronal and oligodendrocyte loss and reactive astrogliosis. It can result in physical disability and acute neurological and cognitive problems. Despite the gains in knowledge of immunology, cell biology, and genetics in the last five decades, the ultimate etiology or specific elements that trigger MS remain unknown. The objective of this review is to propose a theoretical basis for MS etiopathogenesis.Methods: Search was done by accessing PubMed/Medline, EBSCO, and PsycINFO databases. The search string used was "(multiple sclerosis* OR EAE) AND (pathophysiology* OR etiopathogenesis)". The electronic databases were searched for titles or abstracts containing these terms in all published articles between January 1, 1960, and June 30, 2019. The search was filtered down to 362 articles which were included in this review.Results: A framework to better understand the etiopathogenesis and pathophysiology of MS can be derived from four essential factors; mitochondria dysfunction (MtD) & oxidative stress (OS), vitamin D (VD), sex hormones and thyroid hormones. These factors play a direct role in MS etiopathogenesis and have a modulatory effect on many other factors involved in the disease.Conclusions: For better MS prevention and treatment outcomes, efforts should be geared towards treating thyroid problems, sex hormone alterations, VD deficiency, sleep problems and melatonin alterations. MS patients should be encouraged to engage in activities that boost total antioxidant capacity (TAC) including diet and regular exercise and discouraged from activities that promote OS including smoking and alcohol consumption.
Collapse
|
19
|
Tobore TO. On elucidation of the role of mitochondria dysfunction and oxidative stress in multiple sclerosis. ACTA ACUST UNITED AC 2019. [DOI: 10.1111/ncn3.12335] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
20
|
Podlecka-Piętowska A, Kacka A, Zakrzewska-Pniewska B, Nojszewska M, Zieminska E, Chalimoniuk M, Toczylowska B. Altered Cerebrospinal Fluid Concentrations of Hydrophobic and Hydrophilic Compounds in Early Stages of Multiple Sclerosis-Metabolic Profile Analyses. J Mol Neurosci 2019; 69:94-105. [PMID: 31134532 PMCID: PMC6689291 DOI: 10.1007/s12031-019-01336-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 05/07/2019] [Indexed: 11/29/2022]
Abstract
The lack of a single predictive or diagnostic test in multiple sclerosis (MS) remains a major obstacle in the patient’s care. The aim of this study was to investigate metabolic profiles, especially lipids in cerebrospinal fluid (CSF) using 1H-NMR spectroscopy and metabolomics analysis to discriminate MS patient group from the control ones. In this study, 19 MS patients and 19 controls, without neurological problems, patients were enrolled. To obtain the CSF metabolic profiles, NMR spectroscopy was used. Hydrophilic and hydrophobic compounds were analyzed using univariate and multivariate supervised analysis orthogonal partial least square discriminant analysis (OPLS-DA). Targeted OPLS-DA analysis of 32 hydrophilic and 17 hydrophobic compounds obtained 9 hydrophilic metabolites and 8 lipid functional groups which had the highest contribution to patient’s group separation. Lower concentrations of CSF hydrophilic and hydrophobic compounds were observed in MS patients as compared to control group. Acetone, choline, urea, 1,3-dimethylurate, creatinine, isoleucine, myo-inositol, leucine, and 3-OH butyrate; saturated and monounsaturated acyl groups of ω–9, ω–7, ω–6, ω–3, and fatty acid, triglycerides, 1,3-DG, 1-MG, and unassigned component signal at 3.33 ppm were the most important signal compounds in group separation. Analysis of metabolic profile of raw CSF and their lipid extract shows decreased levels of many compounds and led to the conclusion that MS patients could have a disturbance in many metabolic pathways perhaps leading to the decreased level of acetyl-CoA and/or inflammation. CSF metabolic profile analyses could be used as a fingerprint for early MS diagnosis.
Collapse
Affiliation(s)
- A Podlecka-Piętowska
- Department of Neurology, Medical University of Warsaw, Zwirki i Wigury 61, 02-091, Warsaw, Poland
| | - A Kacka
- Department of Anesthesiology, Medical University of Warsaw, Zwirki i Wigury 61, 02-091, Warsaw, Poland. .,Department of Anesthesiology and Intensive Care, The Maria Skłodowska Curie Memorial Cancer Centre and Institute of Oncology, WK Roentgena 5, 02-781, Warsaw, Poland.
| | - B Zakrzewska-Pniewska
- Department of Neurology, Medical University of Warsaw, Zwirki i Wigury 61, 02-091, Warsaw, Poland
| | - M Nojszewska
- Department of Neurology, Medical University of Warsaw, Zwirki i Wigury 61, 02-091, Warsaw, Poland
| | - E Zieminska
- Department of Neurochemistry, Mossakowski Medical Research Centre Polish Academy of Sciences, Pawinskiego Str. 5, 02-107, Warsaw, Poland
| | - M Chalimoniuk
- Department of Cellular Signaling, Mossakowski Medical Research Centre Polish Academy of Sciences, Pawinskiego Str. 5, 02-107, Warsaw, Poland.,Department of Tourism and Health in Biala Podlaska, Józef Piłsudski University of Physical Education in Warsaw, Marymoncka 34, 00-968, Warsaw, Poland
| | - B Toczylowska
- Institute of Biocybernetics and Biomedical Engineering, Trojdena Str. 4, 02-109, Warsaw, Poland.,NMR Laboratory, Institute of Biochemistry and Biophysics, Pawinskiego Str. 5A, 02-107, Warsaw, Poland
| |
Collapse
|
21
|
Zsurka G, Peeva V, Kotlyar A, Kunz WS. Is There Still Any Role for Oxidative Stress in Mitochondrial DNA-Dependent Aging? Genes (Basel) 2018; 9:genes9040175. [PMID: 29561808 PMCID: PMC5924517 DOI: 10.3390/genes9040175] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 03/09/2018] [Accepted: 03/16/2018] [Indexed: 11/21/2022] Open
Abstract
Recent deep sequencing data has provided compelling evidence that the spectrum of somatic point mutations in mitochondrial DNA (mtDNA) in aging tissues lacks G > T transversion mutations. This fact cannot, however, be used as an argument for the missing contribution of reactive oxygen species (ROS) to mitochondria-related aging because it is probably caused by the nucleotide selectivity of mitochondrial DNA polymerase γ (POLG). In contrast to point mutations, the age-dependent accumulation of mitochondrial DNA deletions is, in light of recent experimental data, still explainable by the segregation of mutant molecules generated by the direct mutagenic effects of ROS (in particular, of HO· radicals formed from H2O2 by a Fenton reaction). The source of ROS remains controversial, because the mitochondrial contribution to tissue ROS production is probably lower than previously thought. Importantly, in the discussion about the potential role of oxidative stress in mitochondria-dependent aging, ROS generated by inflammation-linked processes and the distribution of free iron also require careful consideration.
Collapse
Affiliation(s)
- Gábor Zsurka
- Institute of Experimental Epileptology and Neurocognition, University Bonn Medical Center, 53105 Bonn, Germany.
- Department of Epileptology, University Bonn Medical Center, 53105 Bonn, Germany.
| | - Viktoriya Peeva
- Institute of Experimental Epileptology and Neurocognition, University Bonn Medical Center, 53105 Bonn, Germany.
| | - Alexander Kotlyar
- Department of Biochemistry & Molecular Biology, Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel.
| | - Wolfram S Kunz
- Institute of Experimental Epileptology and Neurocognition, University Bonn Medical Center, 53105 Bonn, Germany.
- Department of Epileptology, University Bonn Medical Center, 53105 Bonn, Germany.
| |
Collapse
|
22
|
Understanding a role for hypoxia in lesion formation and location in the deep and periventricular white matter in small vessel disease and multiple sclerosis. Clin Sci (Lond) 2017; 131:2503-2524. [PMID: 29026001 DOI: 10.1042/cs20170981] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Revised: 08/01/2017] [Accepted: 08/15/2017] [Indexed: 12/28/2022]
Abstract
The deep and periventricular white matter is preferentially affected in several neurological disorders, including cerebral small vessel disease (SVD) and multiple sclerosis (MS), suggesting that common pathogenic mechanisms may be involved in this injury. Here we consider the potential pathogenic role of tissue hypoxia in lesion development, arising partly from the vascular anatomy of the affected white matter. Specifically, these regions are supplied by a sparse vasculature fed by long, narrow end arteries/arterioles that are vulnerable to oxygen desaturation if perfusion is reduced (as in SVD, MS and diabetes) or if the surrounding tissue is hypoxic (as in MS, at least). The oxygen crisis is exacerbated by a local preponderance of veins, as these can become highly desaturated 'sinks' for oxygen that deplete it from surrounding tissues. Additional haemodynamic deficiencies, including sluggish flow and impaired vasomotor reactivity and vessel compliance, further exacerbate oxygen insufficiency. The cells most vulnerable to hypoxic damage, including oligodendrocytes, die first, resulting in demyelination. Indeed, in preclinical models, demyelination is prevented if adequate oxygenation is maintained by raising inspired oxygen concentrations. In agreement with this interpretation, there is a predilection of lesions for the anterior and occipital horns of the lateral ventricles, namely regions located at arterial watersheds, or border zones, known to be especially susceptible to hypoperfusion and hypoxia. Finally, mitochondrial dysfunction due to genetic causes, as occurs in leucodystrophies or due to free radical damage, as occurs in MS, will compound any energy insufficiency resulting from hypoxia. Viewing lesion formation from the standpoint of tissue oxygenation not only reveals that lesion distribution is partly predictable, but may also inform new therapeutic strategies.
Collapse
|
23
|
Axonal transport deficits in multiple sclerosis: spiraling into the abyss. Acta Neuropathol 2017; 134:1-14. [PMID: 28315956 PMCID: PMC5486629 DOI: 10.1007/s00401-017-1697-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 03/13/2017] [Accepted: 03/14/2017] [Indexed: 12/16/2022]
Abstract
The transport of mitochondria and other cellular components along the axonal microtubule cytoskeleton plays an essential role in neuronal survival. Defects in this system have been linked to a large number of neurological disorders. In multiple sclerosis (MS) and associated models such as experimental autoimmune encephalomyelitis (EAE), alterations in axonal transport have been shown to exist before neurodegeneration occurs. Genome-wide association (GWA) studies have linked several motor proteins to MS susceptibility, while neuropathological studies have shown accumulations of proteins and organelles suggestive for transport deficits. A reduced effectiveness of axonal transport can lead to neurodegeneration through inhibition of mitochondrial motility, disruption of axoglial interaction or prevention of remyelination. In MS, demyelination leads to dysregulation of axonal transport, aggravated by the effects of TNF-alpha, nitric oxide and glutamate on the cytoskeleton. The combined effect of all these pathways is a vicious cycle in which a defective axonal transport system leads to an increase in ATP consumption through loss of membrane organization and a reduction in available ATP through inhibition of mitochondrial transport, resulting in even further inhibition of transport. The persistent activity of this positive feedback loop contributes to neurodegeneration in MS.
Collapse
|
24
|
Wood ET, Ercan E, Sati P, Cortese ICM, Ronen I, Reich DS. Longitudinal MR spectroscopy of neurodegeneration in multiple sclerosis with diffusion of the intra-axonal constituent N-acetylaspartate. Neuroimage Clin 2017; 15:780-788. [PMID: 28702353 PMCID: PMC5496488 DOI: 10.1016/j.nicl.2017.06.028] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 06/08/2017] [Accepted: 06/20/2017] [Indexed: 11/19/2022]
Abstract
Multiple sclerosis (MS) is a pathologically complex CNS disease: inflammation, demyelination, and neuroaxonal degeneration occur concurrently and may depend on one another. Current therapies are aimed at the immune-mediated, inflammatory destruction of myelin, whereas axonal degeneration is ongoing and not specifically targeted. Diffusion-weighted magnetic resonance spectroscopy can measure the diffusivity of metabolites in vivo, such as the axonal/neuronal constituent N-acetylaspartate, allowing compartment-specific assessment of disease-related changes. Previously, we found significantly lower N-acetylaspartate diffusivity in people with MS compared to healthy controls (Wood et al., 2012) suggesting that this technique can measure axonal degeneration and could be useful in developing neuroprotective agents. In this longitudinal study, we found that N-acetylaspartate diffusivity decreased by 8.3% (p < 0.05) over 6 months in participants who were experiencing clinical or MRI evidence of inflammatory activity (n = 13), whereas there was no significant change in N-acetylaspartate diffusivity in the context of clinical and radiological stability (n = 6). As N-acetylaspartate diffusivity measurements are thought to more specifically reflect the intra-axonal space, these data suggest that N-acetylaspartate diffusivity can report on axonal health on the background of multiple pathological processes in MS, both cross-sectionally and longitudinally.
Collapse
Key Words
- Axonopathy
- DW-MRS, diffusion-weighted magnetic resonance spectroscopy
- Diffusion-weighted magnetic resonance spectroscopy
- EDSS, Expanded Disability Scale Score
- HV, healthy volunteer
- ICV, intracranial volume
- MS, multiple sclerosis
- Multiple sclerosis
- NAA, N-acetylaspartate
- PASAT, Paced Auditory Symbol Addition Test
- T, Tesla
- VOI, volume of interest
- WM, white matter
- White matter
Collapse
Affiliation(s)
- Emily Turner Wood
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Ece Ercan
- C.J. Gorter Center for High Field MRI, Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
| | - Pascal Sati
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Irene C M Cortese
- Neuroimmunology Clinic, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Itamar Ronen
- C.J. Gorter Center for High Field MRI, Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
| | - Daniel S Reich
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA; Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
25
|
Yin X, Kidd GJ, Ohno N, Perkins GA, Ellisman MH, Bastian C, Brunet S, Baltan S, Trapp BD. Proteolipid protein-deficient myelin promotes axonal mitochondrial dysfunction via altered metabolic coupling. J Cell Biol 2017; 215:531-542. [PMID: 27872255 PMCID: PMC5119941 DOI: 10.1083/jcb.201607099] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 09/01/2016] [Accepted: 10/25/2016] [Indexed: 12/12/2022] Open
Abstract
The authors show that central nervous system myelin lacking proteolipid protein (PLP) induces mitochondrial dysfunction, including altered motility, degeneration, and ectopic smooth endoplasmic reticulum interactions, leading to axonal structural defects and degeneration. Mutated PLP occurs in hereditary spastic paraplegia, and these cellular effects provide potential insight into the pathology of the disease. Hereditary spastic paraplegia (HSP) is a neurological syndrome characterized by degeneration of central nervous system (CNS) axons. Mutated HSP proteins include myelin proteolipid protein (PLP) and axon-enriched proteins involved in mitochondrial function, smooth endoplasmic reticulum (SER) structure, and microtubule (MT) stability/function. We characterized axonal mitochondria, SER, and MTs in rodent optic nerves where PLP is replaced by the peripheral nerve myelin protein, P0 (P0-CNS mice). Mitochondrial pathology and degeneration were prominent in juxtaparanodal axoplasm at 1 mo of age. In wild-type (WT) optic nerve axons, 25% of mitochondria–SER associations occurred on extensions of the mitochondrial outer membrane. Mitochondria–SER associations were reduced by 86% in 1-mo-old P0-CNS juxtaparanodal axoplasm. 1-mo-old P0-CNS optic nerves were more sensitive to oxygen-glucose deprivation and contained less adenosine triphosphate (ATP) than WT nerves. MT pathology and paranodal axonal ovoids were prominent at 6 mo. These data support juxtaparanodal mitochondrial degeneration, reduced mitochondria–SER associations, and reduced ATP production as causes of axonal ovoid formation and axonal degeneration.
Collapse
Affiliation(s)
- Xinghua Yin
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Grahame J Kidd
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Nobuhiko Ohno
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Guy A Perkins
- National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, CA 92093
| | - Mark H Ellisman
- National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, CA 92093
| | - Chinthasagar Bastian
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Sylvain Brunet
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Selva Baltan
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Bruce D Trapp
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| |
Collapse
|
26
|
Bodini B, Branzoli F, Poirion E, García-Lorenzo D, Didier M, Maillart E, Socha J, Bera G, Lubetzki C, Ronen I, Lehericy S, Stankoff B. Dysregulation of energy metabolism in multiple sclerosis measured in vivo with diffusion-weighted spectroscopy. Mult Scler 2017; 24:313-321. [DOI: 10.1177/1352458517698249] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Objective: We employed diffusion-weighted magnetic resonance spectroscopy (DW-MRS), which allows to measure in vivo the diffusion properties of metabolites, to explore the functional neuro-axonal damage and the ongoing energetic dysregulation in multiple sclerosis (MS). Methods: Twenty-five patients with MS and 18 healthy controls (HC) underwent conventional magnetic resonance imaging (MRI) and DW-MRS. The apparent diffusion coefficient (ADC) of total N-acetyl-aspartate (tNAA) and creatine–phosphocreatine (tCr) were measured in the parietal normal-appearing white matter (NAWM) and in the thalamic grey matter (TGM). Multiple regressions were used to compare metabolite ADCs between groups and to explore clinical correlations. Results: In patients compared with HCs, we found a reduction in ADC(tNAA) in the TGM, reflecting functional and structural neuro-axonal damage, and in ADC(tCr) in both NAWM and TGM, possibly reflecting a reduction in energy supply in neurons and glial cells. Metabolite ADCs did not correlate with tissue atrophy, lesional volume or metabolite concentrations, while in TGM metabolite ADCs correlated with clinical scores. Conclusion: DW-MRS showed a reduction in tCr diffusivity in the normal-appearing brain of patients with MS, which might reflect a state of ongoing energy dysregulation affecting neurons and/or glial cells. Reversing this energy dysregulation before neuro-axonal degeneration arises may become a key objective in future neuroprotective strategies.
Collapse
Affiliation(s)
- Benedetta Bodini
- Institut du Cerveau et de la Moelle épinière (ICM), CNRS UMR 7225, INSERM, Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, Hôpital de la Pitié Salpêtrière, Paris, France/AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France/AP-HP, Hôpital Saint-Antoine, Paris, France
| | - Francesca Branzoli
- Institut du Cerveau et de la Moelle épinière (ICM), CNRS UMR 7225, INSERM, Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, Hôpital de la Pitié Salpêtrière, Paris, France/Centre de NeuroImagerie de Recherche – Institut du Cerveau et de la Moelle épinière (ICM), Paris, France
| | - Emilie Poirion
- Institut du Cerveau et de la Moelle épinière (ICM), CNRS UMR 7225, INSERM, Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Daniel García-Lorenzo
- Institut du Cerveau et de la Moelle épinière (ICM), CNRS UMR 7225, INSERM, Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Mélanie Didier
- Institut du Cerveau et de la Moelle épinière (ICM), CNRS UMR 7225, INSERM, Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, Hôpital de la Pitié Salpêtrière, Paris, France/Centre de NeuroImagerie de Recherche – Institut du Cerveau et de la Moelle épinière (ICM), Paris, France
| | | | - Julie Socha
- AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Geraldine Bera
- Institut du Cerveau et de la Moelle épinière (ICM), CNRS UMR 7225, INSERM, Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Catherine Lubetzki
- Institut du Cerveau et de la Moelle épinière (ICM), CNRS UMR 7225, INSERM, Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, Hôpital de la Pitié Salpêtrière, Paris, France/AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Itamar Ronen
- Department of Radiology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Stephane Lehericy
- Institut du Cerveau et de la Moelle épinière (ICM), CNRS UMR 7225, INSERM, Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, Hôpital de la Pitié Salpêtrière, Paris, France/Centre de NeuroImagerie de Recherche – Institut du Cerveau et de la Moelle épinière (ICM), Paris, France/AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Bruno Stankoff
- Institut du Cerveau et de la Moelle épinière (ICM), CNRS UMR 7225, INSERM, Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, Hôpital de la Pitié Salpêtrière, Paris, France/AP-HP, Hôpital Saint-Antoine, Paris, France
| |
Collapse
|
27
|
Luo F, Herrup K, Qi X, Yang Y. Inhibition of Drp1 hyper-activation is protective in animal models of experimental multiple sclerosis. Exp Neurol 2017; 292:21-34. [PMID: 28238799 DOI: 10.1016/j.expneurol.2017.02.015] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 02/21/2017] [Accepted: 02/21/2017] [Indexed: 12/20/2022]
Abstract
Multiple Sclerosis (MS), a leading neurological disorder of young adults, is characterized by the loss of oligodendrocytes (OLs), demyelination, inflammation and neuronal degeneration. Here we show that dynamin-related protein 1 (Drp1), a mitochondrial fission protein, is activated in primary OL cells exposed to TNF-α induced inflammation or oxidative stress, as well as in EAE-immunized and cuprizone toxicity-induced demyelinating mouse models. Inhibition of Drp1 hyper-activation by the selective inhibitor P110 abolishes Drp1 translocation to the mitochondria, reduces mitochondrial fragmentation and stems necrosis in primary OLs exposed to TNF-α and H2O2. Notably, in both types of mouse models, treatment with P110 significantly reduces the loss of mature OLs and demyelination, attenuates the number of active microglial cells and astrocytes, yet has no effect on the differentiation of oligodendrocyte precursor cells. Drp1 activation appears to be mediated through the RIPK1/RIPK3/MLKL/PGAM5 pathway during TNF-α-induced oligodendroglia necroptosis. Our results demonstrate a critical role of Drp1 hyper-activation in OL cell death and suggest that an inhibitor of Drp1 hyper-activation such as P110 is worth exploring for its ability to halt or slow the progression of MS.
Collapse
Affiliation(s)
- Fucheng Luo
- Department of Neurology, Case Western Reserve University School of Medicine, 10900 Euclid Ave., Cleveland, OH 44106, USA
| | - Karl Herrup
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| | - Xin Qi
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, 10900 Euclid Ave., Cleveland, OH 44106, USA; Center for Mitochondria Diseases, Case Western Reserve University School of Medicine, 10900 Euclid Ave., Cleveland, OH 44106, USA.
| | - Yan Yang
- Department of Neurology, Case Western Reserve University School of Medicine, 10900 Euclid Ave., Cleveland, OH 44106, USA; Center for Translational Neurosciences, Case Western Reserve University School of Medicine, 10900 Euclid Ave., Cleveland, OH 44106, USA.
| |
Collapse
|
28
|
Nimmagadda VKC, Makar TK, Chandrasekaran K, Sagi AR, Ray J, Russell JW, Bever CT. SIRT1 and NAD+ precursors: Therapeutic targets in multiple sclerosis a review. J Neuroimmunol 2016; 304:29-34. [PMID: 27474445 DOI: 10.1016/j.jneuroim.2016.07.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 07/06/2016] [Indexed: 12/16/2022]
Abstract
Neurodegeneration is an important determinant of disability in multiple sclerosis (MS) but while currently approved treatments reduce inflammation, they have not been shown to reduce neurodegeneration. SIRT1, a NAD dependent protein deacetylase, has been implicated in the pathogenesis of neurodegeneration in neurological diseases including MS. We have studied the role of SIRT1 in experimental autoimmune encephalomyelitis (EAE) and found evidence for a neuroprotective role. In this review we summarize the most recent findings from the use of SIRT1 activators and SIRT1 overexpression in transgenic mice. These data support provide a rational for the use of SIRT1 activators in MS.
Collapse
Affiliation(s)
- Vamshi K C Nimmagadda
- Department of Neurology, University of Maryland, Baltimore, MD 21201, USA; Research Service, VA Maryland Health Care System, Baltimore, MD 21201, USA
| | - Tapas K Makar
- Department of Neurology, University of Maryland, Baltimore, MD 21201, USA; Research Service, VA Maryland Health Care System, Baltimore, MD 21201, USA; VA Multiple Sclerosis Center of Excellence East, Baltimore, MD 21201, USA
| | | | - Avinash Rao Sagi
- Department of Neurology, University of Maryland, Baltimore, MD 21201, USA
| | - Jayanta Ray
- Department of Neurology, University of Maryland, Baltimore, MD 21201, USA
| | - James W Russell
- Department of Neurology, University of Maryland, Baltimore, MD 21201, USA; Research Service, VA Maryland Health Care System, Baltimore, MD 21201, USA
| | - Christopher T Bever
- Department of Neurology, University of Maryland, Baltimore, MD 21201, USA; Research Service, VA Maryland Health Care System, Baltimore, MD 21201, USA; VA Multiple Sclerosis Center of Excellence East, Baltimore, MD 21201, USA.
| |
Collapse
|
29
|
Desai RA, Davies AL, Tachrount M, Kasti M, Laulund F, Golay X, Smith KJ. Cause and prevention of demyelination in a model multiple sclerosis lesion. Ann Neurol 2016; 79:591-604. [PMID: 26814844 PMCID: PMC4949637 DOI: 10.1002/ana.24607] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Revised: 01/22/2016] [Accepted: 01/23/2016] [Indexed: 12/24/2022]
Abstract
Objective Demyelination is a cardinal feature of multiple sclerosis, but it remains unclear why new lesions form, and whether they can be prevented. Neuropathological evidence suggests that demyelination can occur in the relative absence of lymphocytes, and with distinctive characteristics suggestive of a tissue energy deficit. The objective was to examine an experimental model of the early multiple sclerosis lesion and identify pathogenic mechanisms and opportunities for therapy. Methods Demyelinating lesions were induced in the rat spinal dorsal column by microinjection of lipopolysaccharide, and examined immunohistochemically at different stages of development. The efficacy of treatment with inspired oxygen for 2 days following lesion induction was evaluated. Results Demyelinating lesions were not centered on the injection site, but rather formed 1 week later at the white–gray matter border, preferentially including the ventral dorsal column watershed. Lesion formation was preceded by a transient early period of hypoxia and increased production of superoxide and nitric oxide. Oligodendrocyte numbers decreased at the site shortly afterward, prior to demyelination. Lesions formed at a site of inherent susceptibility to hypoxia, as revealed by exposure of naive animals to a hypoxic environment. Notably, raising the inspired oxygen (80%, normobaric) during the hypoxic period significantly reduced or prevented the demyelination. Interpretation Demyelination characteristic of at least some early multiple sclerosis lesions can arise at a vascular watershed following activation of innate immune mechanisms that provoke hypoxia, and superoxide and nitric oxide formation, all of which can compromise cellular energy sufficiency. Demyelination can be reduced or eliminated by increasing inspired oxygen to alleviate the transient hypoxia. Ann Neurol 2016;79:591–604
Collapse
Affiliation(s)
- Roshni A Desai
- Department of Neuroinflammation and Queen Square Multiple Sclerosis Centre, UCL Institute of Neurology, London, United Kingdom
| | - Andrew L Davies
- Department of Neuroinflammation and Queen Square Multiple Sclerosis Centre, UCL Institute of Neurology, London, United Kingdom
| | - Mohamed Tachrount
- Department of Brain Repair and Rehabilitation, UCL Institute of Neurology, London, United Kingdom
| | - Marianne Kasti
- Department of Neuroinflammation and Queen Square Multiple Sclerosis Centre, UCL Institute of Neurology, London, United Kingdom
| | - Frida Laulund
- Department of Neuroinflammation and Queen Square Multiple Sclerosis Centre, UCL Institute of Neurology, London, United Kingdom
| | - Xavier Golay
- Department of Brain Repair and Rehabilitation, UCL Institute of Neurology, London, United Kingdom
| | - Kenneth J Smith
- Department of Neuroinflammation and Queen Square Multiple Sclerosis Centre, UCL Institute of Neurology, London, United Kingdom
| |
Collapse
|
30
|
Ayannuga OA, Shokunbi MT, Naicker TA. Myelin Sheath Injury in Kaolin-Induced Hydrocephalus: A Light and Electron Microscopy Study. Pediatr Neurosurg 2016; 51:61-8. [PMID: 26761462 DOI: 10.1159/000442212] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 11/04/2015] [Indexed: 11/19/2022]
Abstract
BACKGROUND In hydrocephalus, the impairment of cognitive and motor functions is thought to be partly due to injury to the myelin sheath of axons in the central nervous system. The exact nature of this injury is not completely understood. METHODS We induced hydrocephalus in 3-week-old rats with an intracisternal injection of kaolin suspension (0.04 ml of 200 mg/ml) and examined paraffin and ultrathin sections of the subcortical white matter from coronal slices of the cerebrum obtained at the level of the optic chiasm after sacrifice at weekly intervals for 4 weeks. RESULTS Over time, there was a progression of injury to the myelin sheath consisting of attenuation, lamella separation and accumulation of myelin debris, focal degeneration, and the appearance of casts and loops. CONCLUSION The results suggest that myelin injury in kaolin-induced hydrocephalus progresses with the duration and severity of ventriculomegaly.
Collapse
Affiliation(s)
- Olugbenga A Ayannuga
- Department of Anatomy, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | | | | |
Collapse
|
31
|
Praet J, Guglielmetti C, Berneman Z, Van der Linden A, Ponsaerts P. Cellular and molecular neuropathology of the cuprizone mouse model: clinical relevance for multiple sclerosis. Neurosci Biobehav Rev 2015; 47:485-505. [PMID: 25445182 DOI: 10.1016/j.neubiorev.2014.10.004] [Citation(s) in RCA: 301] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 09/18/2014] [Accepted: 10/01/2014] [Indexed: 01/30/2023]
Abstract
The cuprizone mouse model allows the investigation of the complex molecular mechanisms behind nonautoimmune-mediated demyelination and spontaneous remyelination. While it is generally accepted that oligodendrocytes are specifically vulnerable to cuprizone intoxication due to their high metabolic demands, a comprehensive overview of the etiology of cuprizone-induced pathology is still missing to date. In this review we extensively describe the physico-chemical mode of action of cuprizone and discuss the molecular and enzymatic mechanisms by which cuprizone induces metabolic stress, oligodendrocyte apoptosis, myelin degeneration and eventually axonal and neuronal pathology. In addition, we describe the dual effector function of the immune system which tightly controls demyelination by effective induction of oligodendrocyte apoptosis, but in contrast also paves the way for fast and efficient remyelination by the secretion of neurotrophic factors and the clearance of cellular and myelinic debris. Finally, we discuss the many clinical symptoms that can be observed following cuprizone treatment, and how these strengthened the cuprizone model as a useful tool to study human multiple sclerosis, schizophrenia and epilepsy.
Collapse
|
32
|
Abstract
Aspirin is widely used to lessen the risks of cardiovascular events. Some studies suggest that patients with multiple sclerosis have an increased risk for some cardiovascular events, for example, venous thromboembolism and perhaps ischemic strokes, raising the possibility that aspirin could lessen these increased risks in this population or subgroups (patients with limited mobility and/or antiphospholipid antibodies). However, aspirin causes a small increased risk of hemorrhagic stroke, which is a concern as it could potentially worsen a compromised blood-brain barrier. Aspirin has the potential to ameliorate the disease process in multiple sclerosis (for example, by limiting some components of inflammation), but aspirin also has the potential to inhibit mitochondrial complex I activity, which is already reduced in multiple sclerosis. In an experimental setting of a cerebral ischemic lesion, aspirin promoted the proliferation and/or differentiation of oligodendrocyte precursors, raising the possibility that aspirin could facilitate remyelination efforts in multiple sclerosis. Other actions by aspirin may lead to small improvements of some symptoms (for example, lessening fatigue). Here we consider potential benefits and risks of aspirin usage by patients with multiple sclerosis.
Collapse
Affiliation(s)
- Sheila Tsau
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, USA.
| | - Mitchell R Emerson
- Department of Pharmaceutical Sciences, College of Pharmacy-Glendale, Midwestern University, Glendale, AZ, USA.
| | - Sharon G Lynch
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA.
| | - Steven M LeVine
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
33
|
Leber hereditary optic neuropathy and multiple sclerosis: the mitochondrial connection. Can J Ophthalmol 2015; 50:e14-7. [DOI: 10.1016/j.jcjo.2014.10.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 10/22/2014] [Accepted: 10/28/2014] [Indexed: 11/23/2022]
|
34
|
Scola G, Andreazza AC. Oxidative Stress in Bipolar Disorder. OXIDATIVE STRESS IN APPLIED BASIC RESEARCH AND CLINICAL PRACTICE 2015. [DOI: 10.1007/978-1-4939-0440-2_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
35
|
Two-photon imaging of remyelination of spinal cord axons by engrafted neural precursor cells in a viral model of multiple sclerosis. Proc Natl Acad Sci U S A 2014; 111:E2349-55. [PMID: 24843159 DOI: 10.1073/pnas.1406658111] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Neural precursor cells (NPCs) offer a promising approach for treating demyelinating diseases. However, the cellular dynamics that underlie transplanted NPC-mediated remyelination have not been described. Using two-photon imaging of a newly developed ventral spinal cord preparation and a viral model of demyelination, we describe the motility and intercellular interactions of transplanted mouse NPCs expressing green fluorescent protein (GFP) with damaged axons expressing yellow fluorescent protein (YFP). Our findings reveal focal axonal degeneration that occurs in the ventral side of the spinal cord within 1 wk following intracranial instillation with the neurotropic JHM strain of mouse hepatitis virus (JHMV). Axonal damage precedes extensive demyelination and is characterized by swelling along the length of the axon, loss of YFP signal, and transected appearance. NPCs engrafted into spinal cords of JHMV-infected mice exhibited diminished migration velocities and increased proliferation compared with transplanted cells in noninfected mice. NPCs preferentially accumulated within areas of axonal damage, initiated direct contact with axons, and subsequently expressed the myelin proteolipid protein gene, initiating remyelination. These findings indicate that NPCs transplanted into an inflammatory demyelinating microenvironment participate directly in therapeutic outcome through the wrapping of myelin around damaged neurons.
Collapse
|
36
|
LeVine SM, Bilgen M, Lynch SG. Iron accumulation in multiple sclerosis: an early pathogenic event. Expert Rev Neurother 2013; 13:247-50. [PMID: 23448214 DOI: 10.1586/ern.13.14] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Iron has been shown to accumulate in deep gray matter structures in many forms of multiple sclerosis (MS), but detecting its presence early in the disease course (e.g., clinically isolated syndrome [CIS]) has been less clear. Here, we review a recent study where MRI scanning at 7 T together with susceptibility mapping was performed to assess iron deposition in CIS and control subjects. Susceptibility indicative of iron deposition was found to be increased in the globus pallidus, caudate, putamen and pulvinar of CIS patients compared with controls. The findings suggest that iron deposition is a pathological change that occurs early in the development of MS. Identifying the mechanisms of iron accumulation and determining whether iron promotes pathogenesis in MS are important areas of future research.
Collapse
Affiliation(s)
- Steven M LeVine
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, USA.
| | | | | |
Collapse
|
37
|
Degos B, Laforêt P, Jardel C, Sedel F, Jossay-Winter M, Romero NB, Lyon-Caen O, Tourbah A. POLG mutations associated with remitting/relapsing neurological events. J Clin Neurosci 2013; 21:186-8. [PMID: 23830586 DOI: 10.1016/j.jocn.2013.03.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Revised: 10/12/2012] [Accepted: 03/18/2013] [Indexed: 10/26/2022]
Abstract
Recent experimental data underline the relationship between mitochondria and immune function. Clinical reports of patients presenting with mitochondrial dysfunction associated with dysimmune responses in the central nervous system reinforce this new concept. We describe the first case of a woman presenting with symptoms related to a novel compound heterozygous mutation of the mitochondrial polymerase γ (POLG) gene, associated with neurological events suggestive of a demyelinating process. Clinical examination revealed bilateral ptosis, progressive external ophthalmoplegia and axonal sensitive polyneuropathy suggestive of a mitochondrial disease. In line with this, muscle biopsy showed ragged red fibers, and sequencing of POLG revealed two heterozygous mutations. In addition, the patient exhibited relapsing neurological symptoms, and cerebral and spinal MRI mimicking multiple sclerosis. This patient stresses the relationship between mitochondrial dysfunction and inflammation. Recent studies suggest that targeting mitochondrial dysfunction could provide benefits in treating some inflammatory diseases.
Collapse
Affiliation(s)
- Bertrand Degos
- Département de Neurologie, Hôpital Pitié-Salpêtrière, AP-HP, 47-83 boulevard de l'Hôpital, 75013 Paris, France; Université Pierre et Marie Curie (Paris VI), Paris, France.
| | - Pascal Laforêt
- Centre de référence de pathologie neuromusculaire Paris-Est, Hôpital Pitié-Salpêtrière, AP-HP, Paris, France; Institut de Myologie, Hôpital Pitié-Salpêtrière, AP-HP, Paris, France
| | - Claude Jardel
- Laboratoire de Biochimie Métabolique, Hôpital Pitié-Salpêtrière, AP-HP, Paris, France
| | - Frédéric Sedel
- Département de Neurologie, Hôpital Pitié-Salpêtrière, AP-HP, 47-83 boulevard de l'Hôpital, 75013 Paris, France; Université Pierre et Marie Curie (Paris VI), Paris, France
| | | | - Norma B Romero
- Département de Neurologie, Hôpital Pitié-Salpêtrière, AP-HP, 47-83 boulevard de l'Hôpital, 75013 Paris, France; Institut de Myologie, Unité de Morphologie Neuromusculaire, Inserm UMRS 974, CNRS UMR 7215, Hôpital Pitié-Salpêtrière, AP-HP, Paris, France
| | - Olivier Lyon-Caen
- Département de Neurologie, Hôpital Pitié-Salpêtrière, AP-HP, 47-83 boulevard de l'Hôpital, 75013 Paris, France; Université Pierre et Marie Curie (Paris VI), Paris, France
| | - Ayman Tourbah
- Service de Neurologie et Faculté de Médecine, Université Champagne-Ardenne CHU de Reims, Reims, France; Laboratoire de psychopathologie et Neuropsychologie, EA2027 Université Paris, France
| |
Collapse
|
38
|
Rice CM, Sun M, Kemp K, Gray E, Wilkins A, Scolding NJ. Mitochondrial sirtuins - a new therapeutic target for repair and protection in multiple sclerosis. Eur J Neurosci 2012; 35:1887-93. [DOI: 10.1111/j.1460-9568.2012.08150.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
39
|
Chang JR, Ghafouri M, Mukerjee R, Bagashev A, Chabrashvili T, Sawaya BE. Role of p53 in neurodegenerative diseases. NEURODEGENER DIS 2011; 9:68-80. [PMID: 22042001 DOI: 10.1159/000329999] [Citation(s) in RCA: 104] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2011] [Accepted: 06/09/2011] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND p53 plays an important role in many areas of cellular physiology and biology, ranging from cellular development and differentiation to cell cycle arrest and apoptosis. Many of its functions are attributed to its role in assuring proper cellular division. However, since the establishment of its role in cell cycle arrest, damage repair, and apoptosis (thus also establishing its importance in cancer development), numerous reports have demonstrated additional functions of p53 in various cells. In particular, p53 appears to have important functions as it relates to neurodegeneration and synaptic plasticity. OBJECTIVE In this review, we will address p53 functions as it relates to various neurodegenerative diseases, mainly its implications in the development of HIV-associated neurocognitive disorders. CONCLUSION p53 plays a pivotal role in the development of neurodegenerative diseases through its interaction with cellular factors, viral factors, and/or small RNAs that have the ability to promote the development of these diseases. Hence, inhibition of p53 may present an ideal target to restore neuronal functions.
Collapse
Affiliation(s)
- J Robert Chang
- Molecular Studies of Neurodegenerative Diseases Laboratory, Department of Neurology, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | | | | | | | | | | |
Collapse
|