1
|
Yang H, Mo N, Tong L, Dong J, Fan Z, Jia M, Yue J, Wang Y. Microglia lactylation in relation to central nervous system diseases. Neural Regen Res 2025; 20:29-40. [PMID: 38767474 PMCID: PMC11246148 DOI: 10.4103/nrr.nrr-d-23-00805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 08/09/2023] [Accepted: 01/08/2024] [Indexed: 05/22/2024] Open
Abstract
The development of neurodegenerative diseases is closely related to the disruption of central nervous system homeostasis. Microglia, as innate immune cells, play important roles in the maintenance of central nervous system homeostasis, injury response, and neurodegenerative diseases. Lactate has been considered a metabolic waste product, but recent studies are revealing ever more of the physiological functions of lactate. Lactylation is an important pathway in lactate function and is involved in glycolysis-related functions, macrophage polarization, neuromodulation, and angiogenesis and has also been implicated in the development of various diseases. This review provides an overview of the lactate metabolic and homeostatic regulatory processes involved in microglia lactylation, histone versus non-histone lactylation, and therapeutic approaches targeting lactate. Finally, we summarize the current research on microglia lactylation in central nervous system diseases. A deeper understanding of the metabolic regulatory mechanisms of microglia lactylation will provide more options for the treatment of central nervous system diseases.
Collapse
Affiliation(s)
- Hui Yang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang Province, China
| | - Nan Mo
- Department of Clinical Laboratory, The Fourth Clinical Medical College of Zhejiang University of Traditional Chinese Medicine (Hangzhou First People’s Hospital), Hangzhou, Zhejiang Province, China
| | - Le Tong
- College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Jianhong Dong
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang Province, China
| | - Ziwei Fan
- Department of Orthopedics (Spine Surgery), the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Mengxian Jia
- Department of Orthopedics (Spine Surgery), the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Juanqing Yue
- Department of Pathology, Affiliated Hangzhou First People’s Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Ying Wang
- Department of Clinical Research Center, Affiliated Hangzhou First People’s Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang Province, China
| |
Collapse
|
2
|
Amatruda M, Marechal D, Gacias M, Wentling M, Turpin-Nolan S, Morstein J, Moniruzzaman M, Brüning JC, Haughey NJ, Trauner DH, Casaccia P. Neuroprotective effect of neuron-specific deletion of the C16 ceramide synthetic enzymes in an animal model of multiple sclerosis. Glia 2024. [PMID: 39489703 DOI: 10.1002/glia.24631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 09/05/2024] [Accepted: 10/10/2024] [Indexed: 11/05/2024]
Abstract
Ceramide C16 is a sphingolipid detected at high levels in several neurodegenerative disorders, including multiple sclerosis (MS). It can be generated de novo or from the hydrolysis of other sphingolipids, such as sphingomyelin or through the recycling of sphingosine, in what is known as the salvage pathway. While the myelin damage occurring in MS suggests the importance of the hydrolytic and salvage pathways, the growing interest on the importance of diet in demyelinating disorders, prompted us to investigate the involvement of de novo ceramide C16 synthesis on disease severity. A diet rich in saturated fats such as palmitic acid, as found in many highly processed foods, provides substrates for the ceramide C16 synthetic enzymes ceramide synthase 6 (CERS6) and 5 (CERS5), which are expressed in the central nervous system. Using the experimental autoimmune encephalomyelitis (EAE) model of inflammatory demyelination, we show here that mice with CamK2a+ neuronal specific deletion of both CerS6 and CerS5 show a milder course of EAE than wild type mice, even when fed a diet enriched in palmitic acid. At a cellular level, neurons lacking both CerS6 and CerS5 are protected from the mitochondrial dysfunction arising from exposure to oxidative stress and palmitic acid in the medium. These data underscore the importance of a healthy diet avoiding processed foods for demyelinating disorders and identifies endogenous neuronal synthesis of ceramide C16 as an important determinant of disease severity.
Collapse
Affiliation(s)
- Mario Amatruda
- Neuroscience Initiative, Advanced Science Research Center, CUNY, New York, New York, USA
| | - Damien Marechal
- Neuroscience Initiative, Advanced Science Research Center, CUNY, New York, New York, USA
| | - Mar Gacias
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Maureen Wentling
- Neuroscience Initiative, Advanced Science Research Center, CUNY, New York, New York, USA
| | - Sarah Turpin-Nolan
- Max Planck Institute for Metabolism Research, Cologne, Germany
- Cellular and Molecular Metabolism Laboratory, Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Johannes Morstein
- Department of Chemistry, New York University, Silver Center, New York, New York, USA
- Department of Cellular and Molecular Pharmacology, UCSF, San Francisco, California, USA
| | - Mohammed Moniruzzaman
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jens C Brüning
- Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Norman J Haughey
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Dirk H Trauner
- Department of Chemistry, New York University, Silver Center, New York, New York, USA
- Chemistry and Translational Therapeutics and Systems Pharmacology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Patrizia Casaccia
- Neuroscience Initiative, Advanced Science Research Center, CUNY, New York, New York, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Program in Biochemistry, The Graduate Center of The City University of New York, New York, New York, USA
- Program in Biology, The Graduate Center of The City University of New York, New York, New York, USA
| |
Collapse
|
3
|
Fang Y, Min S, Shen H. The role of mitochondrial fusion and fission in immune-mediated inflammatory diseases. Cell Immunol 2024; 403-404:104864. [PMID: 39190985 DOI: 10.1016/j.cellimm.2024.104864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 08/29/2024]
Abstract
Mitochondria are highly dynamic organelles that maintain their homeostasis through mitochondrial dynamics. Mitochondrial fusion and fission are two important processes of mitochondrial dynamics. There is accumulating evidence that mitochondrial fusion and fission play an important role in the development of immune-mediated inflammatory diseases. This article provides a brief review of the essential role of mitochondrial fusion and fission in immune-mediated inflammatory diseases. It will provide a novel perspective and direction for the elucidation of the pathogenesis and treatment of immune-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Yulai Fang
- Nanjing University of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China; Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing 210029, China
| | - Shichen Min
- Nanjing University of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Hong Shen
- Nanjing University of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China; Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing 210029, China.
| |
Collapse
|
4
|
Clayton BLL, Barbar L, Sapar M, Kalpana K, Rao C, Migliori B, Rusielewicz T, Paull D, Brenner K, Moroziewicz D, Sand IK, Casaccia P, Tesar PJ, Fossati V. Patient iPSC models reveal glia-intrinsic phenotypes in multiple sclerosis. Cell Stem Cell 2024:S1934-5909(24)00288-1. [PMID: 39191254 DOI: 10.1016/j.stem.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 06/17/2024] [Accepted: 08/05/2024] [Indexed: 08/29/2024]
Abstract
Multiple sclerosis (MS) is an inflammatory and neurodegenerative disease of the central nervous system (CNS), resulting in neurological disability that worsens over time. While progress has been made in defining the immune system's role in MS pathophysiology, the contribution of intrinsic CNS cell dysfunction remains unclear. Here, we generated a collection of induced pluripotent stem cell (iPSC) lines from people with MS spanning diverse clinical subtypes and differentiated them into glia-enriched cultures. Using single-cell transcriptomic profiling and orthogonal analyses, we observed several distinguishing characteristics of MS cultures pointing to glia-intrinsic disease mechanisms. We found that primary progressive MS-derived cultures contained fewer oligodendrocytes. Moreover, MS-derived oligodendrocyte lineage cells and astrocytes showed increased expression of immune and inflammatory genes, matching those of glia from MS postmortem brains. Thus, iPSC-derived MS models provide a unique platform for dissecting glial contributions to disease phenotypes independent of the peripheral immune system and identify potential glia-specific targets for therapeutic intervention.
Collapse
Affiliation(s)
- Benjamin L L Clayton
- Institute for Glial Sciences, Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Lilianne Barbar
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, USA
| | - Maria Sapar
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, USA
| | - Kriti Kalpana
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, USA
| | - Chandrika Rao
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, USA
| | - Bianca Migliori
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, USA
| | - Tomasz Rusielewicz
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, USA
| | - Daniel Paull
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, USA
| | - Katie Brenner
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, USA
| | - Dorota Moroziewicz
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, USA
| | - Ilana Katz Sand
- Corinne Goldsmith Dickinson Center for Multiple Sclerosis, Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10129, USA
| | - Patrizia Casaccia
- Neuroscience Initiative, Advanced Science Research Center at CUNY, New York, NY 10031, USA
| | - Paul J Tesar
- Institute for Glial Sciences, Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| | - Valentina Fossati
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, USA.
| |
Collapse
|
5
|
Zivadinov R, Schweser F, Jakimovski D, Bergsland N, Dwyer MG. Decoding Gray Matter Involvement in Multiple Sclerosis via Imaging. Neuroimaging Clin N Am 2024; 34:453-468. [PMID: 38942527 DOI: 10.1016/j.nic.2024.03.007] [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] [Indexed: 06/30/2024]
Abstract
Multiple sclerosis (MS) is increasingly understood not only as a white matter disease but also involving both the deep and cortical gray matter (GM). GM pathology in people with MS (pwMS) includes the presence of lesions, leptomeningeal inflammation, atrophy, altered iron concentration, and microstructural changes. Studies using 7T and 3T MR imaging with optimized protocols established that GM damage is a principal driver of disease progression in pwMS. Future work is needed to incorporate the assessment of these GM imaging biomarkers into the clinical workup of pwMS and the assessment of treatment efficacy.
Collapse
Affiliation(s)
- Robert Zivadinov
- Department of Neurology, Buffalo Neuroimaging Analysis Center, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA; Center for Biomedical Imaging at Clinical Translational Science Institute, University at Buffalo, State University of New York, Buffalo, NY, USA.
| | - Ferdinand Schweser
- Department of Neurology, Buffalo Neuroimaging Analysis Center, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA; Center for Biomedical Imaging at Clinical Translational Science Institute, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Dejan Jakimovski
- Department of Neurology, Buffalo Neuroimaging Analysis Center, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Niels Bergsland
- Department of Neurology, Buffalo Neuroimaging Analysis Center, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Michael G Dwyer
- Department of Neurology, Buffalo Neuroimaging Analysis Center, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA; Center for Biomedical Imaging at Clinical Translational Science Institute, University at Buffalo, State University of New York, Buffalo, NY, USA
| |
Collapse
|
6
|
Clayton BL, Barbar L, Sapar M, Rusielewicz T, Kalpana K, Migliori B, Paull D, Brenner K, Moroziewicz D, Sand IK, Casaccia P, Tesar PJ, Fossati V. Patient iPSC models reveal glia-intrinsic phenotypes in multiple sclerosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.01.551553. [PMID: 37577713 PMCID: PMC10418164 DOI: 10.1101/2023.08.01.551553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Multiple sclerosis (MS) is considered an inflammatory and neurodegenerative disease of the central nervous system, typically resulting in significant neurological disability that worsens over time. While considerable progress has been made in defining the immune system's role in MS pathophysiology, the contribution of intrinsic CNS-cell dysfunction remains unclear. Here, we generated the largest reported collection of iPSC lines from people with MS spanning diverse clinical subtypes and differentiated them into glia-enriched cultures. Using single-cell transcriptomic profiling, we observed several distinguishing characteristics of MS cultures pointing to glia-intrinsic disease mechanisms. We found that iPSC-derived cultures from people with primary progressive MS contained fewer oligodendrocytes. Moreover, iPSC-oligodendrocyte lineage cells and astrocytes from people with MS showed increased expression of immune and inflammatory genes that match those of glial cells from MS postmortem brains. Thus, iPSC-derived MS models provide a unique platform for dissecting glial contributions to disease phenotypes independent of the peripheral immune system and identify potential glia-specific targets for therapeutic intervention.
Collapse
Affiliation(s)
- Benjamin L.L. Clayton
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
- These authors contributed equally
| | - Lilianne Barbar
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, USA
- Current affiliation: Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, 63105, USA
- These authors contributed equally
| | - Maria Sapar
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, USA
| | - Tomasz Rusielewicz
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, USA
| | - Kriti Kalpana
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, USA
| | - Bianca Migliori
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, USA
| | | | - Daniel Paull
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, USA
| | - Katie Brenner
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, USA
| | - Dorota Moroziewicz
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, USA
| | - Ilana Katz Sand
- Corinne Goldsmith Dickinson Center for Multiple Sclerosis, Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10129, USA
| | | | - Paul J. Tesar
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Valentina Fossati
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, USA
| |
Collapse
|
7
|
Tokarska N, Naniong JMA, Johnston JM, Salapa HE, Muir GD, Levin MC, Popescu BF, Verge VMK. Acute intermittent hypoxia alters disease course and promotes CNS repair including resolution of inflammation and remyelination in the experimental autoimmune encephalomyelitis model of MS. Glia 2023; 71:2045-2066. [PMID: 37132422 DOI: 10.1002/glia.24381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 04/09/2023] [Accepted: 04/15/2023] [Indexed: 05/04/2023]
Abstract
Remyelination and neurodegeneration prevention mitigate disability in Multiple Sclerosis (MS). We have shown acute intermittent hypoxia (AIH) is a novel, non-invasive and effective therapy for peripheral nerve repair, including remyelination. Thus, we posited AIH would improve repair following CNS demyelination and address the paucity of MS repair treatments. AIH's capacity to enhance intrinsic repair, functional recovery and alter disease course in the experimental autoimmune encephalomyelitis (EAE) model of MS was assessed. EAE was induced by MOG35-55 immunization in C57BL/6 female mice. EAE mice received either AIH (10 cycles-5 min 11% oxygen alternating with 5 min 21% oxygen) or Normoxia (control; 21% oxygen for same duration) once daily for 7d beginning at near peak EAE disease score of 2.5. Mice were followed post-treatment for an additional 7d before assessing histopathology or 14d to examine maintenance of AIH effects. Alterations in histopathological correlates of multiple repair indices were analyzed quantitatively in focally demyelinated ventral lumbar spinal cord areas to assess AIH impacts. AIH begun at near peak disease significantly improved daily clinical scores/functional recovery and associated histopathology relative to Normoxia controls and the former were maintained for at least 14d post-treatment. AIH enhanced correlates of myelination, axon protection and oligodendrocyte precursor cell recruitment to demyelinated areas. AIH also effected a dramatic reduction in inflammation, while polarizing remaining macrophages/microglia toward a pro-repair state. Collectively, this supports a role for AIH as a novel non-invasive therapy to enhance CNS repair and alter disease course following demyelination and holds promise as a neuroregenerative MS strategy.
Collapse
Affiliation(s)
- Nataliya Tokarska
- Department of Anatomy, Physiology and Pharmacology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Cameco MS Neuroscience Research Centre, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Justin M A Naniong
- Department of Anatomy, Physiology and Pharmacology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Cameco MS Neuroscience Research Centre, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Jayne M Johnston
- Department of Anatomy, Physiology and Pharmacology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Cameco MS Neuroscience Research Centre, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Hannah E Salapa
- Cameco MS Neuroscience Research Centre, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Office of the Saskatchewan Multiple Sclerosis Clinical Research Chair, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- College of Medicine, Neurology Division, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Gillian D Muir
- Cameco MS Neuroscience Research Centre, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Department of Biomedical Sciences, WCVM, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Michael C Levin
- Cameco MS Neuroscience Research Centre, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Office of the Saskatchewan Multiple Sclerosis Clinical Research Chair, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- College of Medicine, Neurology Division, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Bogdan F Popescu
- Department of Anatomy, Physiology and Pharmacology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Cameco MS Neuroscience Research Centre, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Valerie M K Verge
- Department of Anatomy, Physiology and Pharmacology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Cameco MS Neuroscience Research Centre, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
8
|
Virupakshaiah A, Ladakis DC, Nourbakhsh B, Bhargava P, Dilwali S, Schoeps V, Borkowski K, Newman JW, Waubant E. Several serum lipid metabolites are associated with relapse risk in pediatric-onset multiple sclerosis. Mult Scler 2023; 29:936-944. [PMID: 37199529 PMCID: PMC10524330 DOI: 10.1177/13524585231171517] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
BACKGROUND The circulating metabolome is altered in multiple sclerosis (MS), but its prognostic capabilities have not been extensively explored. Lipid metabolites might be of particular interest due to their multiple roles in the brain, as they can serve as structural components, energy sources, and bioactive molecules. Gaining a deeper understanding of the disease may be possible by examining the lipid metabolism in the periphery, which serves as the primary source of lipids for the brain. OBJECTIVE To determine if altered serum lipid metabolites are associated with the risk of relapse and disability in children with MS. METHODS We collected serum samples from 61 participants with pediatric-onset MS within 4 years of disease onset. Prospective longitudinal relapse data and cross-sectional disability measures (Expanded Disability Status Scale [EDSS]) were collected. Serum metabolomics was performed using untargeted liquid chromatography and mass spectrometry. Individual lipid metabolites were clustered into pre-defined pathways. The associations between clusters of metabolites and relapse rate and EDSS score were estimated utilizing negative binomial and linear regression models, respectively. RESULTS We found that serum acylcarnitines (relapse rate: normalized enrichment score [NES] = 2.1, q = 1.03E-04; EDSS: NES = 1.7, q = 0.02) and poly-unsaturated fatty acids (relapse rate: NES = 1.6, q = 0.047; EDSS: NES = 1.9, q = 0.005) were associated with higher relapse rates and EDSS, while serum phosphatidylethanolamines (relapse rate: NES = -2.3, q = 0.002; EDSS: NES = -2.1, q = 0.004), plasmalogens (relapse rate: NES = -2.5, q = 5.81E-04; EDSS: NES = -2.1, q = 0.004), and primary bile acid metabolites (relapse rate: NES = -2.0, q = 0.02; EDSS: NES = -1.9, q = 0.02) were associated with lower relapse rates and lower EDSS. CONCLUSION This study supports the role of some lipid metabolites in pediatric MS relapses and disability.
Collapse
Affiliation(s)
- Akash Virupakshaiah
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Dimitrios C Ladakis
- Division of Neuroimmunology, Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Bardia Nourbakhsh
- Division of Neuroimmunology, Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Pavan Bhargava
- Division of Neuroimmunology, Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Sonam Dilwali
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Vinicius Schoeps
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Kamil Borkowski
- West Coast Metabolomics Center, University of California Davis, Davis, CA, USA
| | - John W Newman
- West Coast Metabolomics Center, University of California Davis, Davis, CA, USA United States Department of Agriculture, Agricultural Research Service, Western Human Nutrition Research Center, Davis, CA, USA Department of Nutrition, University of California Davis, Davis, CA, USA
| | - Emmanuelle Waubant
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
9
|
Herman S, Arvidsson McShane S, Zjukovskaja C, Khoonsari PE, Svenningsson A, Burman J, Spjuth O, Kultima K. Disease phenotype prediction in multiple sclerosis. iScience 2023; 26:106906. [PMID: 37332601 PMCID: PMC10275960 DOI: 10.1016/j.isci.2023.106906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 03/09/2023] [Accepted: 05/12/2023] [Indexed: 06/20/2023] Open
Abstract
Progressive multiple sclerosis (PMS) is currently diagnosed retrospectively. Here, we work toward a set of biomarkers that could assist in early diagnosis of PMS. A selection of cerebrospinal fluid metabolites (n = 15) was shown to differentiate between PMS and its preceding phenotype in an independent cohort (AUC = 0.93). Complementing the classifier with conformal prediction showed that highly confident predictions could be made, and that three out of eight patients developing PMS within three years of sample collection were predicted as PMS at that time point. Finally, this methodology was applied to PMS patients as part of a clinical trial for intrathecal treatment with rituximab. The methodology showed that 68% of the patients decreased their similarity to the PMS phenotype one year after treatment. In conclusion, the inclusion of confidence predictors contributes with more information compared to traditional machine learning, and this information is relevant for disease monitoring.
Collapse
Affiliation(s)
- Stephanie Herman
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | | | | | - Payam Emami Khoonsari
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
- Department of Biochemistry and Biophysics, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Stockholm University, Box 1031, 17121 Solna, Sweden
| | - Anders Svenningsson
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Joachim Burman
- Department of Neuroscience, Neurology, Uppsala University, Uppsala, Sweden
| | - Ola Spjuth
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Kim Kultima
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
| |
Collapse
|
10
|
Mormone E, Iorio EL, Abate L, Rodolfo C. Sirtuins and redox signaling interplay in neurogenesis, neurodegenerative diseases, and neural cell reprogramming. Front Neurosci 2023; 17:1073689. [PMID: 36816109 PMCID: PMC9929468 DOI: 10.3389/fnins.2023.1073689] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 01/13/2023] [Indexed: 02/04/2023] Open
Abstract
Since the discovery of Neural Stem Cells (NSCs) there are still mechanism to be clarified, such as the role of mitochondrial metabolism in the regulation of endogenous adult neurogenesis and its implication in neurodegeneration. Although stem cells require glycolysis to maintain their stemness, they can perform oxidative phosphorylation and it is becoming more and more evident that mitochondria are central players, not only for ATP production but also for neuronal differentiation's steps regulation, through their ability to handle cellular redox state, intracellular signaling, epigenetic state of the cell, as well as the gut microbiota-brain axis, upon dietary influences. In this scenario, the 8-oxoguanine DNA glycosylase (OGG1) repair system would link mitochondrial DNA integrity to the modulation of neural differentiation. On the other side, there is an increasing interest in NSCs generation, from induced pluripotent stem cells, as a clinical model for neurodegenerative diseases (NDs), although this methodology still presents several drawbacks, mainly related to the reprogramming process. Indeed, high levels of reactive oxygen species (ROS), associated with telomere shortening, genomic instability, and defective mitochondrial dynamics, lead to pluripotency limitation and reprogramming efficiency's reduction. Moreover, while a physiological or moderate ROS increase serves as a signaling mechanism, to activate differentiation and suppress self-renewal, excessive oxidative stress is a common feature of NDs and aging. This ROS-dependent regulatory effect might be modulated by newly identified ROS suppressors, including the NAD+-dependent deacetylase enzymes family called Sirtuins (SIRTs). Recently, the importance of subcellular localization of NAD synthesis has been coupled to different roles for NAD in chromatin stability, DNA repair, circadian rhythms, and longevity. SIRTs have been described as involved in the control of both telomere's chromatin state and expression of nuclear gene involved in the regulation of mitochondrial gene expression, as well as in several NDs and aging. SIRTs are ubiquitously expressed in the mammalian brain, where they play important roles. In this review we summarize the current knowledge on how SIRTs-dependent modulation of mitochondrial metabolism could impact on neurogenesis and neurodegeneration, focusing mainly on ROS function and their role in SIRTs-mediated cell reprogramming and telomere protection.
Collapse
Affiliation(s)
- Elisabetta Mormone
- Unitá Produttiva per Terapie Avanzate, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy,*Correspondence: Elisabetta Mormone, ;
| | | | - Lucrezia Abate
- Unitá Produttiva per Terapie Avanzate, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Carlo Rodolfo
- Department of Biology, University of Rome Tor Vergata, Rome, Italy,Department of Paediatric Onco-Haematology and Cell and Gene Therapy, IRCCS Bambino Gesù Children’s Hospital, Rome, Italy,Carlo Rodolfo,
| |
Collapse
|
11
|
Tonietto M, Poirion E, Lazzarotto A, Ricigliano V, Papeix C, Bottlaender M, Bodini B, Stankoff B. Periventricular remyelination failure in multiple sclerosis: a substrate for neurodegeneration. Brain 2023; 146:182-194. [PMID: 36097347 DOI: 10.1093/brain/awac334] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 07/26/2022] [Accepted: 08/18/2022] [Indexed: 01/11/2023] Open
Abstract
In multiple sclerosis, spontaneous remyelination is generally incomplete and heterogeneous across patients. A high heterogeneity in remyelination may also exist across lesions within the same individual, suggesting the presence of local factors interfering with myelin regeneration. In this study we explored in vivo the regional distribution of myelin repair and investigated its relationship with neurodegeneration. We first took advantage of the myelin binding property of the amyloid radiotracer 11C-PiB to conduct a longitudinal 11C-PiB PET study in an original cohort of 19 participants with a relapsing-remitting form of multiple sclerosis, followed-up over a period of 1-4 months. We then replicated our results on an independent cohort of 40 people with multiple sclerosis followed-up over 1 year with magnetization transfer imaging, an MRI metrics sensitive to myelin content. For each imaging method, voxel-wise maps of myelin content changes were generated according to modality-specific thresholds. We demonstrated a selective failure of remyelination in periventricular white matter lesions of people with multiple sclerosis in both cohorts. In both the original and the replication cohort, we estimated that the probability of demyelinated voxels to remyelinate over the follow-up increased significantly as a function of the distance from ventricular CSF. Enlarged choroid plexus, a recently discovered biomarker linked to neuroinflammation, was found to be associated with the periventricular failure of remyelination in the two cohorts (r = -0.79, P = 0.0018; r = -0.40, P = 0.045, respectively), suggesting a role of the brain-CSF barrier in affecting myelin repair in surrounding tissues. In both cohorts, the failure of remyelination in periventricular white matter lesions was associated with lower thalamic volume (r = 0.86, P < 0.0001; r = 0.33; P = 0.069, respectively), an imaging marker of neurodegeneration. Interestingly, we also showed an association between the periventricular failure of remyelination and regional cortical atrophy that was mediated by the number of cortex-derived tracts passing through periventricular white matter lesions, especially in patients at the relapsing-remitting stage. Our findings demonstrate that lesion proximity to ventricles is associated with a failure of myelin repair and support the hypothesis that a selective periventricular remyelination failure in combination with the large number of tracts connecting periventricular lesions with cortical areas is a key mechanism contributing to cortical damage in multiple sclerosis.
Collapse
Affiliation(s)
- Matteo Tonietto
- Paris Brain Institute, Sorbonne Université, ICM, CNRS, Inserm, Paris, France.,Service Hospitalier Frédéric Joliot, Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Orsay, France
| | - Emilie Poirion
- Paris Brain Institute, Sorbonne Université, ICM, CNRS, Inserm, Paris, France
| | - Andrea Lazzarotto
- Paris Brain Institute, Sorbonne Université, ICM, CNRS, Inserm, Paris, France.,Neurology Department, St Antoine Hospital, APHP, Paris, France
| | - Vito Ricigliano
- Paris Brain Institute, Sorbonne Université, ICM, CNRS, Inserm, Paris, France.,Neurology Department, St Antoine Hospital, APHP, Paris, France
| | - Caroline Papeix
- Paris Brain Institute, Sorbonne Université, ICM, CNRS, Inserm, Paris, France.,Neurology Department, Pitié-Salpêtrière Hospital, APHP, Paris, France
| | - Michel Bottlaender
- Service Hospitalier Frédéric Joliot, Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Orsay, France
| | - Benedetta Bodini
- Paris Brain Institute, Sorbonne Université, ICM, CNRS, Inserm, Paris, France.,Neurology Department, St Antoine Hospital, APHP, Paris, France
| | - Bruno Stankoff
- Paris Brain Institute, Sorbonne Université, ICM, CNRS, Inserm, Paris, France.,Neurology Department, St Antoine Hospital, APHP, Paris, France
| |
Collapse
|
12
|
Blood Metabolomics May Discriminate a Sub-Group of Patients with First Demyelinating Episode in the Context of RRMS with Increased Disability and MRI Characteristics Indicative of Poor Prognosis. Int J Mol Sci 2022; 23:ijms232314578. [PMID: 36498904 PMCID: PMC9735785 DOI: 10.3390/ijms232314578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/08/2022] [Accepted: 11/17/2022] [Indexed: 11/24/2022] Open
Abstract
Biomarker research across the health-to-disease continuum is being increasingly applied. We applied blood-based metabolomics in order to identify patient clusters with a first demyelinating episode, and explored the prognostic potential of the method by thoroughly characterizing each cluster in terms of clinical, laboratory and MRI markers of established prognostic potential for Multiple Sclerosis (MS). Recruitment consisted of 11 patients with Clinically Isolated Syndrome (CIS), 37 patients with a first demyelinating episode in the context of Relapsing-Remitting MS (RRMS) and 11 control participants. Blood-based metabolomics and hierarchical clustering analysis (HCL) were applied. Constructed OPLS-DA models illustrated a discrimination between patients with CIS and the controls (p = 0.0014), as well as between patients with RRMS and the controls (p = 1 × 10−5). Hierarchical clustering analysis (HCL) for patients with RRMS identified three clusters. RRMS-patients-cluster-3 exhibited higher mean cell numbers in the Cerebro-spinal Fluid (CSF) compared to patients with CIS (18.17 ± 6.3 vs. 1.09 ± 0.41, p = 0.004). Mean glucose CSF/serum ratio and infratentorial lesion burden significantly differed across CIS- and HCL-derived RRMS-patient clusters (F = 14.95, p < 0.001 and F = 6.087, p = 0.002, respectively), mainly due to increased mean values for patients with RRMS-cluster-3. HCL discriminated a cluster of patients with a first demyelinating episode in the context of RRMS with increased disability, laboratory findings linked with increased pathology burden and MRI markers of poor prognosis.
Collapse
|
13
|
Borziak K, Finkelstein J. X-linked genetic risk factors that promote autoimmunity and dampen remyelination are associated with multiple sclerosis susceptibility. Mult Scler Relat Disord 2022; 66:104065. [PMID: 35905688 DOI: 10.1016/j.msard.2022.104065] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/14/2022] [Accepted: 07/17/2022] [Indexed: 01/21/2023]
Abstract
BACKGROUND Multiple sclerosis (MS) is a chronic neurodegenerative disease, which has a strong genetic component and is more prevalent in women. MS is caused by an autoimmunity initiated inflammatory response which leads to axon demyelination, followed by axon loss, plaque formation and neurodegeneration. The goal of this article was to explore X-linked genetic factors that are associated with MS susceptibility. METHODS Using UK Biobank microarray, we analyzed the prevalence of alleles on the X chromosome to identify variants potentially involved in MS. Overall, 488,225 patients across 18,857 markers were analyzed using PLINK. RESULTS Our results identify 20 SNPs that are significantly more abundant in persons with MS. The genes associated with these SNPs belong to immunity (LAMP2, AVPR2, MTMR8, F8, BCOR, PORCN, and ELF4) and remyelination (NSDHL, HS6ST2, RBM10, TAZ, and AR) pathways that are potentially of great significance for understanding the onset and progression of multiple sclerosis. We further identified a significant 20-fold increase in incidence of MS cases in women with co-occurrences of SNPs associated with myelination and immunity functions. CONCLUSIONS Our analysis provides novel insights into the roles of X-linked genes in the onset and presentation of multiple sclerosis, identifying 20 SNPs in 14 genes involved primarily in immunity and myelination functions that are significantly more abundant in persons with MS. Our co-occurrence analysis suggests that concurrent disruption of both myelination and immune systems significantly increases the risk of MS onset in women.
Collapse
Affiliation(s)
- Kirill Borziak
- Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 United States.
| | - Joseph Finkelstein
- Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 United States
| |
Collapse
|
14
|
Beirowski B. Emerging evidence for compromised axonal bioenergetics and axoglial metabolic coupling as drivers of neurodegeneration. Neurobiol Dis 2022; 170:105751. [PMID: 35569720 DOI: 10.1016/j.nbd.2022.105751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/20/2022] [Accepted: 05/09/2022] [Indexed: 10/18/2022] Open
Abstract
Impaired bioenergetic capacity of the nervous system is thought to contribute to the pathogenesis of many neurodegenerative diseases (NDD). Since neuronal synapses are believed to be the major energy consumers in the nervous system, synaptic derangements resulting from energy deficits have been suggested to play a central role for the development of many of these disorders. However, long axons constitute the largest compartment of the neuronal network, require large amounts of energy, are metabolically and structurally highly vulnerable, and undergo early injurious stresses in many NDD. These stresses likely impose additional energy demands for continuous adaptations and repair processes, and may eventually overwhelm axonal maintenance mechanisms. Indeed, pathological axon degeneration (pAxD) is now recognized as an etiological focus in a wide array of NDD associated with bioenergetic abnormalities. In this paper I first discuss the recognition that a simple experimental model for pAxD is regulated by an auto-destruction program that exhausts distressed axons energetically. Provision of the energy substrate pyruvate robustly counteracts this axonal breakdown. Importantly, energy decline in axons is not only a consequence but also an initiator of this program. This opens the intriguing possibility that axon dysfunction and pAxD can be suppressed by preemptively energizing distressed axons. Second, I focus on the emerging concept that axons communicate energetically with their flanking glia. This axoglial metabolic coupling can help offset the axonal energy decline that activates the pAxD program but also jeopardize axon integrity as a result of perturbed glial metabolism. Third, I present compelling evidence that abnormal axonal energetics and compromised axoglial metabolic coupling accompany the activation of the pAxD auto-destruction pathway in models of glaucoma, a widespread neurodegenerative condition with pathogenic overlap to other common NDD. In conclusion, I propose a novel conceptual framework suggesting that therapeutic interventions focused on bioenergetic support of the nervous system should also address axons and their metabolic interactions with glia.
Collapse
Affiliation(s)
- Bogdan Beirowski
- Institute for Myelin and Glia Exploration, New York State Center of Excellence in Bioinformatics & Life Sciences (CBLS), University at Buffalo, Buffalo, NY 14203, USA; Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14214, USA.
| |
Collapse
|
15
|
Iram T, Kern F, Kaur A, Myneni S, Morningstar AR, Shin H, Garcia MA, Yerra L, Palovics R, Yang AC, Hahn O, Lu N, Shuken SR, Haney MS, Lehallier B, Iyer M, Luo J, Zetterberg H, Keller A, Zuchero JB, Wyss-Coray T. Young CSF restores oligodendrogenesis and memory in aged mice via Fgf17. Nature 2022; 605:509-515. [PMID: 35545674 PMCID: PMC9377328 DOI: 10.1038/s41586-022-04722-0] [Citation(s) in RCA: 104] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 04/04/2022] [Indexed: 12/15/2022]
Abstract
Recent understanding of how the systemic environment shapes the brain throughout life has led to numerous intervention strategies to slow brain ageing1-3. Cerebrospinal fluid (CSF) makes up the immediate environment of brain cells, providing them with nourishing compounds4,5. We discovered that infusing young CSF directly into aged brains improves memory function. Unbiased transcriptome analysis of the hippocampus identified oligodendrocytes to be most responsive to this rejuvenated CSF environment. We further showed that young CSF boosts oligodendrocyte progenitor cell (OPC) proliferation and differentiation in the aged hippocampus and in primary OPC cultures. Using SLAMseq to metabolically label nascent mRNA, we identified serum response factor (SRF), a transcription factor that drives actin cytoskeleton rearrangement, as a mediator of OPC proliferation following exposure to young CSF. With age, SRF expression decreases in hippocampal OPCs, and the pathway is induced by acute injection with young CSF. We screened for potential SRF activators in CSF and found that fibroblast growth factor 17 (Fgf17) infusion is sufficient to induce OPC proliferation and long-term memory consolidation in aged mice while Fgf17 blockade impairs cognition in young mice. These findings demonstrate the rejuvenating power of young CSF and identify Fgf17 as a key target to restore oligodendrocyte function in the ageing brain.
Collapse
Affiliation(s)
- Tal Iram
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA.,Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, California, USA,Correspondence to or
| | - Fabian Kern
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA.,Chair for Clinical Bioinformatics, Saarland University, 66123 Saarbrücken, Germany.,Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), Saarland University Campus E8.1, Saarbrücken, Germany
| | - Achint Kaur
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA.,Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, California, USA
| | - Saket Myneni
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA.,Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, California, USA
| | - Allison R. Morningstar
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA.,Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, California, USA
| | - Heather Shin
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA.,Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, California, USA
| | - Miguel A. Garcia
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, CA 94305, USA
| | - Lakshmi Yerra
- Palo Alto Veterans Institute for Research, Palo Alto, CA 94304
| | - Robert Palovics
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA.,Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, California, USA
| | - Andrew C. Yang
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA.,Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, California, USA
| | - Oliver Hahn
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA.,Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, California, USA
| | - Nannan Lu
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA.,Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, California, USA
| | - Steven R. Shuken
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA.,Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, California, USA,Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Michael s. Haney
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA.,Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, California, USA
| | - Benoit Lehallier
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA.,Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, California, USA
| | - Manasi Iyer
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, CA 94305, USA
| | - Jian Luo
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA.,Palo Alto Veterans Institute for Research, Palo Alto, CA 94304
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK; UK Dementia Research Institute at UCL, London, UK
| | - Andreas Keller
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA.,Chair for Clinical Bioinformatics, Saarland University, 66123 Saarbrücken, Germany.,Center for Bioinformatics, Saarland Informatics Campus, 66123 Saarbrücken, Germany
| | - J. Bradley Zuchero
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, CA 94305, USA
| | - Tony Wyss-Coray
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA.,Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, California, USA,Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, California, USA.,Correspondence to or
| |
Collapse
|
16
|
Ntranos A, Park HJ, Wentling M, Tolstikov V, Amatruda M, Inbar B, Kim-Schulze S, Frazier C, Button J, Kiebish MA, Lublin F, Edwards K, Casaccia P. Bacterial neurotoxic metabolites in multiple sclerosis cerebrospinal fluid and plasma. Brain 2022; 145:569-583. [PMID: 34894211 PMCID: PMC10060700 DOI: 10.1093/brain/awab320] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 07/14/2021] [Accepted: 08/01/2021] [Indexed: 11/14/2022] Open
Abstract
The identification of intestinal dysbiosis in patients with neurological and psychiatric disorders has highlighted the importance of gut-brain communication, and yet the question regarding the identity of the components responsible for this cross-talk remains open. We previously reported that relapsing remitting multiple sclerosis patients treated with dimethyl fumarate have a prominent depletion of the gut microbiota, thereby suggesting that studying the composition of plasma and CSF samples from these patients may help to identify microbially derived metabolites. We used a functional xenogeneic assay consisting of cultured rat neurons exposed to CSF samples collected from multiple sclerosis patients before and after dimethyl fumarate treatment to assess neurotoxicity and then conducted a metabolomic analysis of plasma and CSF samples to identify metabolites with differential abundance. A weighted correlation network analysis allowed us to identify groups of metabolites, present in plasma and CSF samples, whose abundance correlated with the neurotoxic potential of the CSF. This analysis identified the presence of phenol and indole group metabolites of bacterial origin (e.g. p-cresol sulphate, indoxyl sulphate and N-phenylacetylglutamine) as potentially neurotoxic and decreased by treatment. Chronic exposure of cultured neurons to these metabolites impaired their firing rate and induced axonal damage, independent from mitochondrial dysfunction and oxidative stress, thereby identifying a novel pathway of neurotoxicity. Clinical, radiological and cognitive test metrics were also collected in treated patients at follow-up visits. Improved MRI metrics, disability and cognition were only detected in dimethyl fumarate-treated relapsing remitting multiple sclerosis patients. The levels of the identified metabolites of bacterial origin (p-cresol sulphate, indoxyl sulphate and N-phenylacetylglutamine) were inversely correlated to MRI measurements of cortical volume and directly correlated to the levels of neurofilament light chain, an established biomarker of neurodegeneration. Our data suggest that phenol and indole derivatives from the catabolism of tryptophan and phenylalanine are microbially derived metabolites, which may mediate gut-brain communication and induce neurotoxicity in multiple sclerosis.
Collapse
Affiliation(s)
- Achilles Ntranos
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Advanced Science Research Center at the Graduate Center of the City University of New York, New York, NY 10031, USA
| | - Hye-Jin Park
- Advanced Science Research Center at the Graduate Center of the City University of New York, New York, NY 10031, USA
| | - Maureen Wentling
- Advanced Science Research Center at the Graduate Center of the City University of New York, New York, NY 10031, USA
| | | | - Mario Amatruda
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Advanced Science Research Center at the Graduate Center of the City University of New York, New York, NY 10031, USA
| | - Benjamin Inbar
- Advanced Science Research Center at the Graduate Center of the City University of New York, New York, NY 10031, USA
| | - Seunghee Kim-Schulze
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Carol Frazier
- Multiple Sclerosis Center of Northeastern New York, Latham, NY 12110, USA
| | - Judy Button
- Multiple Sclerosis Center of Northeastern New York, Latham, NY 12110, USA
| | | | - Fred Lublin
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Keith Edwards
- Multiple Sclerosis Center of Northeastern New York, Latham, NY 12110, USA
| | - Patrizia Casaccia
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Advanced Science Research Center at the Graduate Center of the City University of New York, New York, NY 10031, USA
- Graduate Program in Biology and Biochemistry at the Graduate Center of the City University of New York, New York, NY, USA
| |
Collapse
|
17
|
Ghirotto B, Oliveira DF, Cipelli M, Basso PJ, de Lima J, Breda CNS, Ribeiro HC, Silva CCC, Sertié AL, Oliveira AER, Hiyane MI, Caldini EG, Sussulini A, Nakaya HI, Kowaltowski AJ, Oliveira EML, Zatz M, Câmara NOS. MS-driven metabolic alterations are recapitulated in iPSC-derived astrocytes. Ann Neurol 2022; 91:652-669. [PMID: 35226368 PMCID: PMC9310856 DOI: 10.1002/ana.26336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 02/17/2022] [Accepted: 02/18/2022] [Indexed: 11/20/2022]
Abstract
Objective Astrocytes play a significant role in the pathology of multiple sclerosis (MS). Nevertheless, for ethical reasons, most studies in these cells were performed using the Experimental Autoimmune Encephalomyelitis model. As there are significant differences between human and mouse cells, we aimed here to better characterize astrocytes from patients with MS (PwMS), focusing mainly on mitochondrial function and cell metabolism. Methods We obtained and characterized induced pluripotent stem cell (iPSC)‐derived astrocytes from three PwMS and three unaffected controls, and performed electron microscopy, flow cytometry, cytokine and glutamate measurements, gene expression, in situ respiration, and metabolomics. We validated our findings using a single‐nuclei RNA sequencing dataset. Results We detected several differences in MS astrocytes including: (i) enrichment of genes associated with neurodegeneration, (ii) increased mitochondrial fission, (iii) increased production of superoxide and MS‐related proinflammatory chemokines, (iv) impaired uptake and enhanced release of glutamate, (v) increased electron transport capacity and proton leak, in line with the increased oxidative stress, and (vi) a distinct metabolic profile, with a deficiency in amino acid catabolism and increased sphingolipid metabolism, which have already been linked to MS. Interpretation Here we describe the metabolic profile of iPSC‐derived astrocytes from PwMS and validate this model as a very powerful tool to study disease mechanisms and to perform non‐invasive drug targeting assays in vitro. Our findings recapitulate several disease features described in patients and provide new mechanistic insights into the metabolic rewiring of astrocytes in MS, which could be targeted in future therapeutic studies. ANN NEUROL 2022;91:652–669
Collapse
Affiliation(s)
- Bruno Ghirotto
- Transplantation Immunobiology Laboratory, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, 05508-900, Brazil
| | - Danyllo F Oliveira
- Human Genome and Stem Cell Research Center, Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of São Paulo, São Paulo, SP, 05508-090, Brazil
| | - Marcella Cipelli
- Transplantation Immunobiology Laboratory, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, 05508-900, Brazil
| | - Paulo J Basso
- Transplantation Immunobiology Laboratory, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, 05508-900, Brazil
| | - Jean de Lima
- Transplantation Immunobiology Laboratory, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, 05508-900, Brazil
| | - Cristiane N S Breda
- Transplantation Immunobiology Laboratory, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, 05508-900, Brazil
| | - Henrique C Ribeiro
- Laboratory of Bioanalytics and Integrated Omics, Department of Analytical Chemistry, Institute of Chemistry, State University of Campinas, Campinas, SP, 13083-970, Brazil
| | - Camille C C Silva
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP, 05508-000, Brazil
| | - Andrea L Sertié
- Hospital Israelita Albert Einstein, São Paulo, SP, 05652-900, Brazil
| | - Antonio Edson R Oliveira
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, SP, 05508-000, Brazil
| | - Meire I Hiyane
- Transplantation Immunobiology Laboratory, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, 05508-900, Brazil
| | - Elia G Caldini
- Cell Biology Laboratory, Department of Pathology, School of Medicine, University of São Paulo, São Paulo, SP, 01246903, Brazil
| | - Alessandra Sussulini
- Laboratory of Bioanalytics and Integrated Omics, Department of Analytical Chemistry, Institute of Chemistry, State University of Campinas, Campinas, SP, 13083-970, Brazil
| | - Helder I Nakaya
- Hospital Israelita Albert Einstein, São Paulo, SP, 05652-900, Brazil.,Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, SP, 05508-000, Brazil
| | - Alicia J Kowaltowski
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP, 05508-000, Brazil
| | - Enedina M L Oliveira
- Neuroimmunology Clinic, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, 04039-002, Brazil
| | - Mayana Zatz
- Human Genome and Stem Cell Research Center, Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of São Paulo, São Paulo, SP, 05508-090, Brazil
| | - Niels O S Câmara
- Transplantation Immunobiology Laboratory, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, 05508-900, Brazil
| |
Collapse
|
18
|
Gianferrara T, Cescon E, Grieco I, Spalluto G, Federico S. Glycogen Synthase Kinase 3β Involvement in Neuroinflammation and Neurodegenerative Diseases. Curr Med Chem 2022; 29:4631-4697. [PMID: 35170406 DOI: 10.2174/0929867329666220216113517] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 11/24/2021] [Accepted: 12/19/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND GSK-3β activity has been strictly related to neuroinflammation and neurodegeneration. Alzheimer's disease is the most studied neurodegenerative disease, but GSK-3β seems to be involved in almost all neurodegenerative diseases including Parkinson's disease, amyotrophic lateral sclerosis, frontotemporal dementia, Huntington's disease and the autoimmune disease multiple sclerosis. OBJECTIVE The aim of this review is to help researchers both working on this research topic or not to have a comprehensive overview on GSK-3β in the context of neuroinflammation and neurodegeneration. METHOD Literature has been searched using PubMed and SciFinder databases by inserting specific keywords. A total of more than 500 articles have been discussed. RESULTS First of all, the structure and regulation of the kinase were briefly discussed and then, specific GSK-3β implications in neuroinflammation and neurodegenerative diseases were illustrated also with the help of figures, to conclude with a comprehensive overview on the most important GSK-3β and multitarget inhibitors. For all discussed compounds, the structure and IC50 values at the target kinase have been reported. CONCLUSION GSK-3β is involved in several signaling pathways both in neurons as well as in glial cells and immune cells. The fine regulation and interconnection of all these pathways are at the base of the rationale use of GSK-3β inhibitors in neuroinflammation and neurodegeneration. In fact, some compounds are now under clinical trials. Despite this, pharmacodynamic and ADME/Tox profiles of the compounds were often not fully characterized and this is deleterious in such a complex system.
Collapse
Affiliation(s)
- Teresa Gianferrara
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Trieste, Italy
| | - Eleonora Cescon
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Trieste, Italy
| | - Ilenia Grieco
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Trieste, Italy
| | - Giampiero Spalluto
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Trieste, Italy
| | - Stephanie Federico
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Trieste, Italy
| |
Collapse
|
19
|
Relapse rates and risk factors for unfavorable neurological prognosis of transverse myelitis in systemic lupus erythematosus: A systematic review and meta-analysis. Autoimmun Rev 2021; 21:102996. [PMID: 34798313 DOI: 10.1016/j.autrev.2021.102996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 11/14/2021] [Indexed: 11/20/2022]
Abstract
BACKGROUND Transverse myelitis (TM) is a rare but severe systemic lupus erythematosus (SLE) manifestation. To date, the prognostic factors for SLE-associated TM have been far less well-studied. There are also controversial data on the association of antiphospholipid antibodies (aPLs), Systemic Lupus Erythematosus Disease Activity Index (SLEDAI) score, longitudinal extensive transverse myelitis (LETM), and decreased complement levels with the outcome of TM. We aimed to review the potential prognostic factors and integrate relapse rates of observational studies for SLE-associated TM. METHOD To review the prognosis for SLE-associated TM, relevant articles published up to July 30, 2021, were comprehensively and systematically identified from PubMed, EMBASE and Web of Science databases. Five studies encompassing 283 patients with SLE-related TM were included in this meta-analysis; raw data were obtained from three studies. RESULTS The risk factors for unfavorable neurological outcome included demographic features, clinical characteristics, laboratory data, among which a grade of A, B or C on the American Spinal Injury Association Impairment Scale (AIS) at the onset of TM was associated with poor prognosis (OR: 56.05, 95% CI: 6.29-499.25, P < 0.001). The presence of hypoglycorrhachia was also correlated with a worse prognosis (OR: 10.78, 95% CI: 3.74-31.07, P < 0.001). No noticeable correlation was revealed between a poor outcome and positive aPLs and different aPLs profiles (anticardiolipin antibody [aCL], anti-β2-glycoprotein I (anti-β2GPI], lupus anticoagulant [LA]). The pooled 1-, 3- and 5-year relapse rates were 22% (95% CI: 0.13-0.31), 34% (95% CI: 0.22-0.47) and 36% (95% CI: 0.14-0.58), respectively. No significant publication bias was found. CONCLUSION A grade of A, B, or C on the AIS at initial TM and the presence of hypoglycorrhachia were found to be related to a worse prognosis in patients with SLE-associated TM. Notably, aPLs and different aPLs profiles may not suggest poor neurological outcome. The long-term relapse rate of patients with SLE-associated TM was relatively high. We recommend that treatment be stratified based on the initial severity of myelitis. For patients with severe myelitis, early intensive therapy may be initiated as soon as possible.
Collapse
|
20
|
Ricigliano VAG, Morena E, Colombi A, Tonietto M, Hamzaoui M, Poirion E, Bottlaender M, Gervais P, Louapre C, Bodini B, Stankoff B. Choroid Plexus Enlargement in Inflammatory Multiple Sclerosis: 3.0-T MRI and Translocator Protein PET Evaluation. Radiology 2021; 301:166-177. [PMID: 34254858 DOI: 10.1148/radiol.2021204426] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Background Choroid plexuses (CPs) have been suggested as a key gateway for inflammation in experimental autoimmune encephalitis, but in vivo evidence of their involvement in multiple sclerosis (MS) is lacking. Purpose To assess CP volumetric and inflammatory changes in patients with MS versus healthy control participants. Materials and Methods This was a secondary analysis of 97 patients (61 with relapsing-remitting MS [RRMS] and 36 with progressive MS) and 44 healthy control participants who participated in three prospective 3.0-T brain MRI studies between May 2009 and September 2017. A subgroup of 37 patients and 19 healthy control participants also underwent translocator protein fluorine 18 (18F)-DPA-714 PET for neuroinflammation. Relapses and disability scores were collected at baseline and over 2 years. CPs were manually segmented on three-dimensional T1-weighted images; other brain volumes were additionally segmented. Volumes were expressed as a ratio of intracranial volume. The 18F-DPA-714 distribution volume ratio was quantified in parenchymal regions, whereas standardized uptake value was used for CP inflammation. Multivariable linear regression analyses were performed to assess CP volumetric and inflammatory differences between patients with MS and healthy control participants and correlations between CP volume and lesion load, brain volumes, 18F-DPA-714 uptake, and annualized relapse rate. Results Ninety-seven patients with MS (mean age, 42 years ± 12 [standard deviation]; 49 women) and 44 healthy control participants (mean age, 39 years ± 14; 23 women) underwent MRI. Thirty-seven patients with MS and 19 healthy control participants underwent PET. CPs were 35% larger in patients with MS (mean value, 15.9 × 10-4 ± 4.5) than in healthy control participants (mean value, 11.8 × 10-4 ± 3.8; P = .004). Subgroup analysis confirmed greater CP volume in patients with RRMS (mean value, 15.5 × 10-4 ± 4.6; P = .008) than in healthy control participants. CP enlargement was greater in patients with active lesions at MRI (mean volume, 18.2 × 10-4 ± 4.9 in patients with lesions that enhanced with gadolinium vs 14.9 × 10-4 ± 4 in patients with lesions that did not enhance with gadolinium; P < .001) and correlated with white matter lesion load (r = 0.39; 95% CI: 0.20, 0.55; P < .001) and 18F-DPA-714 binding in the thalami (r = 0.44; 95% CI: 0.22, 0.72; P = .04) and normal-appearing white matter (r = 0.5; 95% CI: 0.20, 0.71; P = .005). Moreover, it correlated with annualized relapse rate in patients with RRMS (r = 0.37; 95% CI: 0.1, 0.55; P = .005). Finally, patients with MS showed 18.5% higher CP 18F-DPA-714 uptake than control participants (mean value, 0.778 ± 0.23 vs 0.635 ± 0.15, respectively; P = .01). CP volume in patients with RRMS (r = 0.57; 95% CI: 0.37, 0.73; P = .009) correlated with higher 18F-DPA-714 uptake. Conclusion Choroid plexuses (CPs) are enlarged and inflamed in patients with multiple sclerosis (MS), particularly in those with relapsing-remitting MS with inflammatory profiles; CP volumetric analysis could represent an MS imaging marker. © RSNA, 2021 EudraCT no. 2008-004174-40; clinical trial registration nos. NCT02305264 and NCT01651520 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Vito A G Ricigliano
- From the Sorbonne Université, Paris Brain Institute, Institut du Cerveau, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Paris, France (V.A.G.R., E.M., A.C., M.T., M.H., E.P., C.L., B.B., B.S.); Université Paris-Saclay, Commissariat à l'énergie atomique et aux énergies alternatives, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, BioMaps, Service Hospitalier Frédéric Joliot, Orsay, France (M.T., M.B., P.G.); Service d'Imagerie Médicale, Hôpital Fondation Adolphe de Rothschild, Paris, France (E.P.); Department of Neurology, Pitié-Salpêtrière Hospital, Assistance Publique des Hôpitaux de Paris, Paris, France (C.L.); and Department of Neurology, St Antoine Hospital, 184, rue du Faubourg St Antoine, Assistance Publique des Hôpitaux de Paris, 75571 Paris, France (B.B., B.S.)
| | - Emanuele Morena
- From the Sorbonne Université, Paris Brain Institute, Institut du Cerveau, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Paris, France (V.A.G.R., E.M., A.C., M.T., M.H., E.P., C.L., B.B., B.S.); Université Paris-Saclay, Commissariat à l'énergie atomique et aux énergies alternatives, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, BioMaps, Service Hospitalier Frédéric Joliot, Orsay, France (M.T., M.B., P.G.); Service d'Imagerie Médicale, Hôpital Fondation Adolphe de Rothschild, Paris, France (E.P.); Department of Neurology, Pitié-Salpêtrière Hospital, Assistance Publique des Hôpitaux de Paris, Paris, France (C.L.); and Department of Neurology, St Antoine Hospital, 184, rue du Faubourg St Antoine, Assistance Publique des Hôpitaux de Paris, 75571 Paris, France (B.B., B.S.)
| | - Annalisa Colombi
- From the Sorbonne Université, Paris Brain Institute, Institut du Cerveau, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Paris, France (V.A.G.R., E.M., A.C., M.T., M.H., E.P., C.L., B.B., B.S.); Université Paris-Saclay, Commissariat à l'énergie atomique et aux énergies alternatives, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, BioMaps, Service Hospitalier Frédéric Joliot, Orsay, France (M.T., M.B., P.G.); Service d'Imagerie Médicale, Hôpital Fondation Adolphe de Rothschild, Paris, France (E.P.); Department of Neurology, Pitié-Salpêtrière Hospital, Assistance Publique des Hôpitaux de Paris, Paris, France (C.L.); and Department of Neurology, St Antoine Hospital, 184, rue du Faubourg St Antoine, Assistance Publique des Hôpitaux de Paris, 75571 Paris, France (B.B., B.S.)
| | - Matteo Tonietto
- From the Sorbonne Université, Paris Brain Institute, Institut du Cerveau, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Paris, France (V.A.G.R., E.M., A.C., M.T., M.H., E.P., C.L., B.B., B.S.); Université Paris-Saclay, Commissariat à l'énergie atomique et aux énergies alternatives, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, BioMaps, Service Hospitalier Frédéric Joliot, Orsay, France (M.T., M.B., P.G.); Service d'Imagerie Médicale, Hôpital Fondation Adolphe de Rothschild, Paris, France (E.P.); Department of Neurology, Pitié-Salpêtrière Hospital, Assistance Publique des Hôpitaux de Paris, Paris, France (C.L.); and Department of Neurology, St Antoine Hospital, 184, rue du Faubourg St Antoine, Assistance Publique des Hôpitaux de Paris, 75571 Paris, France (B.B., B.S.)
| | - Mariem Hamzaoui
- From the Sorbonne Université, Paris Brain Institute, Institut du Cerveau, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Paris, France (V.A.G.R., E.M., A.C., M.T., M.H., E.P., C.L., B.B., B.S.); Université Paris-Saclay, Commissariat à l'énergie atomique et aux énergies alternatives, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, BioMaps, Service Hospitalier Frédéric Joliot, Orsay, France (M.T., M.B., P.G.); Service d'Imagerie Médicale, Hôpital Fondation Adolphe de Rothschild, Paris, France (E.P.); Department of Neurology, Pitié-Salpêtrière Hospital, Assistance Publique des Hôpitaux de Paris, Paris, France (C.L.); and Department of Neurology, St Antoine Hospital, 184, rue du Faubourg St Antoine, Assistance Publique des Hôpitaux de Paris, 75571 Paris, France (B.B., B.S.)
| | - Emilie Poirion
- From the Sorbonne Université, Paris Brain Institute, Institut du Cerveau, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Paris, France (V.A.G.R., E.M., A.C., M.T., M.H., E.P., C.L., B.B., B.S.); Université Paris-Saclay, Commissariat à l'énergie atomique et aux énergies alternatives, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, BioMaps, Service Hospitalier Frédéric Joliot, Orsay, France (M.T., M.B., P.G.); Service d'Imagerie Médicale, Hôpital Fondation Adolphe de Rothschild, Paris, France (E.P.); Department of Neurology, Pitié-Salpêtrière Hospital, Assistance Publique des Hôpitaux de Paris, Paris, France (C.L.); and Department of Neurology, St Antoine Hospital, 184, rue du Faubourg St Antoine, Assistance Publique des Hôpitaux de Paris, 75571 Paris, France (B.B., B.S.)
| | - Michel Bottlaender
- From the Sorbonne Université, Paris Brain Institute, Institut du Cerveau, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Paris, France (V.A.G.R., E.M., A.C., M.T., M.H., E.P., C.L., B.B., B.S.); Université Paris-Saclay, Commissariat à l'énergie atomique et aux énergies alternatives, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, BioMaps, Service Hospitalier Frédéric Joliot, Orsay, France (M.T., M.B., P.G.); Service d'Imagerie Médicale, Hôpital Fondation Adolphe de Rothschild, Paris, France (E.P.); Department of Neurology, Pitié-Salpêtrière Hospital, Assistance Publique des Hôpitaux de Paris, Paris, France (C.L.); and Department of Neurology, St Antoine Hospital, 184, rue du Faubourg St Antoine, Assistance Publique des Hôpitaux de Paris, 75571 Paris, France (B.B., B.S.)
| | - Philippe Gervais
- From the Sorbonne Université, Paris Brain Institute, Institut du Cerveau, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Paris, France (V.A.G.R., E.M., A.C., M.T., M.H., E.P., C.L., B.B., B.S.); Université Paris-Saclay, Commissariat à l'énergie atomique et aux énergies alternatives, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, BioMaps, Service Hospitalier Frédéric Joliot, Orsay, France (M.T., M.B., P.G.); Service d'Imagerie Médicale, Hôpital Fondation Adolphe de Rothschild, Paris, France (E.P.); Department of Neurology, Pitié-Salpêtrière Hospital, Assistance Publique des Hôpitaux de Paris, Paris, France (C.L.); and Department of Neurology, St Antoine Hospital, 184, rue du Faubourg St Antoine, Assistance Publique des Hôpitaux de Paris, 75571 Paris, France (B.B., B.S.)
| | - Céline Louapre
- From the Sorbonne Université, Paris Brain Institute, Institut du Cerveau, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Paris, France (V.A.G.R., E.M., A.C., M.T., M.H., E.P., C.L., B.B., B.S.); Université Paris-Saclay, Commissariat à l'énergie atomique et aux énergies alternatives, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, BioMaps, Service Hospitalier Frédéric Joliot, Orsay, France (M.T., M.B., P.G.); Service d'Imagerie Médicale, Hôpital Fondation Adolphe de Rothschild, Paris, France (E.P.); Department of Neurology, Pitié-Salpêtrière Hospital, Assistance Publique des Hôpitaux de Paris, Paris, France (C.L.); and Department of Neurology, St Antoine Hospital, 184, rue du Faubourg St Antoine, Assistance Publique des Hôpitaux de Paris, 75571 Paris, France (B.B., B.S.)
| | - Benedetta Bodini
- From the Sorbonne Université, Paris Brain Institute, Institut du Cerveau, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Paris, France (V.A.G.R., E.M., A.C., M.T., M.H., E.P., C.L., B.B., B.S.); Université Paris-Saclay, Commissariat à l'énergie atomique et aux énergies alternatives, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, BioMaps, Service Hospitalier Frédéric Joliot, Orsay, France (M.T., M.B., P.G.); Service d'Imagerie Médicale, Hôpital Fondation Adolphe de Rothschild, Paris, France (E.P.); Department of Neurology, Pitié-Salpêtrière Hospital, Assistance Publique des Hôpitaux de Paris, Paris, France (C.L.); and Department of Neurology, St Antoine Hospital, 184, rue du Faubourg St Antoine, Assistance Publique des Hôpitaux de Paris, 75571 Paris, France (B.B., B.S.)
| | - Bruno Stankoff
- From the Sorbonne Université, Paris Brain Institute, Institut du Cerveau, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Paris, France (V.A.G.R., E.M., A.C., M.T., M.H., E.P., C.L., B.B., B.S.); Université Paris-Saclay, Commissariat à l'énergie atomique et aux énergies alternatives, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, BioMaps, Service Hospitalier Frédéric Joliot, Orsay, France (M.T., M.B., P.G.); Service d'Imagerie Médicale, Hôpital Fondation Adolphe de Rothschild, Paris, France (E.P.); Department of Neurology, Pitié-Salpêtrière Hospital, Assistance Publique des Hôpitaux de Paris, Paris, France (C.L.); and Department of Neurology, St Antoine Hospital, 184, rue du Faubourg St Antoine, Assistance Publique des Hôpitaux de Paris, 75571 Paris, France (B.B., B.S.)
| |
Collapse
|
21
|
Elevated Mitochondrial Reactive Oxygen Species within Cerebrospinal Fluid as New Index in the Early Detection of Lumbar Spinal Stenosis. Diagnostics (Basel) 2021; 11:diagnostics11050748. [PMID: 33922090 PMCID: PMC8143471 DOI: 10.3390/diagnostics11050748] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/14/2021] [Accepted: 04/20/2021] [Indexed: 01/08/2023] Open
Abstract
Lumbar spinal stenosis (LSS) is a common neurodegenerative condition. However, how neurogenic claudication develops with severe leg pain has not yet been clearly elucidated. Moreover, cerebrospinal fluid (CSF) physiology at the lumbosacral level is poorly understood because of the difficulties involved in quantification and visualization. Recent studies have suggested that assessment of mitochondrial function in CSF provides an indirect way to assess neurological disorders and an important feature of disease progression. In this study, we assessed the relevance of endogenous extracellular mitochondria in the CSF of rats after LSS. Mitochondrial changes within the CSF were analyzed following LSS at 1 week using flow cytometry. An increase in cell size and number was observed in CSF with LSS, and reactive oxygen species (ROS) levels were also increased within the CSF at 1 week in the LSS group. Elevated mitochondrial ROS and functional changes in the CSF are hallmarks of LSS. The present study is the first to demonstrate that elevated mitochondrial ROS within the CSF is a new index for the early detection of LSS. Moreover, it may represent a potential novel treatment target for LSS.
Collapse
|
22
|
Pardini M, Brown JWL, Magliozzi R, Reynolds R, Chard DT. Surface-in pathology in multiple sclerosis: a new view on pathogenesis? Brain 2021; 144:1646-1654. [PMID: 33876200 DOI: 10.1093/brain/awab025] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 11/03/2020] [Accepted: 11/17/2020] [Indexed: 11/12/2022] Open
Abstract
While multiple sclerosis can affect any part of the CNS, it does not do so evenly. In white matter it has long been recognized that lesions tend to occur around the ventricles, and grey matter lesions mainly accrue in the outermost (subpial) cortex. In cortical grey matter, neuronal loss is greater in the outermost layers. This cortical gradient has been replicated in vivo with magnetization transfer ratio and similar gradients in grey and white matter magnetization transfer ratio are seen around the ventricles, with the most severe abnormalities abutting the ventricular surface. The cause of these gradients remains uncertain, though soluble factors released from meningeal inflammation into the CSF has the most supporting evidence. In this Update, we review this 'surface-in' spatial distribution of multiple sclerosis abnormalities and consider the implications for understanding pathogenic mechanisms and treatments designed to slow or stop them.
Collapse
Affiliation(s)
- Matteo Pardini
- NMR Research Unit, Queen Square Multiple Sclerosis Centre, University College London (UCL) Institute of Neurology, London, UK.,Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, and IRCCS AOU San Martino-IST, Genoa, Italy
| | - J William L Brown
- NMR Research Unit, Queen Square Multiple Sclerosis Centre, University College London (UCL) Institute of Neurology, London, UK.,Department of Clinical Neurosciences, University of Cambridge, Box 165, Cambridge Biomedical Campus, Cambridge, UK.,Clinical Outcomes Research Unit (CORe), University of Melbourne, Melbourne, Australia
| | - Roberta Magliozzi
- Department of Neuroscience, Biomedicine and Movement Science, University of Verona, Verona, Italy.,Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK
| | - Richard Reynolds
- Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK.,Centre for Molecular Neuropathology, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Declan T Chard
- NMR Research Unit, Queen Square Multiple Sclerosis Centre, University College London (UCL) Institute of Neurology, London, UK.,National Institute for Health Research (NIHR) University College London Hospitals (UCLH) Biomedical Research Centre, UK
| |
Collapse
|
23
|
Fettig NM, Osborne LC. Direct and indirect effects of microbiota-derived metabolites on neuroinflammation in multiple sclerosis. Microbes Infect 2021; 23:104814. [PMID: 33775860 DOI: 10.1016/j.micinf.2021.104814] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 03/06/2021] [Accepted: 03/09/2021] [Indexed: 12/11/2022]
Abstract
Multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE) are highly influenced by changes in the microbiota and of microbiota-derived metabolites, including short chain fatty acids, bile acids, and tryptophan derivatives. This review will discuss the effects of microbiota-derived metabolites on neuroinflammation driven by central nervous system-resident cells and peripheral immune cells, and their influence on outcomes of EAE and MS.
Collapse
Affiliation(s)
- Naomi M Fettig
- Department of Microbiology & Immunology, Life Sciences Institute, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Lisa C Osborne
- Department of Microbiology & Immunology, Life Sciences Institute, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
24
|
Ng Kee Kwong KC, Harbham PK, Selvaraj BT, Gregory JM, Pal S, Hardingham GE, Chandran S, Mehta AR. 40 Years of CSF Toxicity Studies in ALS: What Have We Learnt About ALS Pathophysiology? Front Mol Neurosci 2021; 14:647895. [PMID: 33815058 PMCID: PMC8012723 DOI: 10.3389/fnmol.2021.647895] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 02/24/2021] [Indexed: 12/11/2022] Open
Abstract
Based on early evidence of in vitro neurotoxicity following exposure to serum derived from patients with amyotrophic lateral sclerosis (ALS), several studies have attempted to explore whether cerebrospinal fluid (CSF) obtained from people with ALS could possess similar properties. Although initial findings proved inconclusive, it is now increasingly recognized that ALS-CSF may exert toxicity both in vitro and in vivo. Nevertheless, the mechanism underlying CSF-induced neurodegeneration remains unclear. This review aims to summarize the 40-year long history of CSF toxicity studies in ALS, while discussing the various mechanisms that have been proposed, including glutamate excitotoxicity, proteotoxicity and oxidative stress. Furthermore, we consider the potential implications of a toxic CSF circulatory system in the pathophysiology of ALS, and also assess its significance in the context of current ALS research.
Collapse
Affiliation(s)
| | - Pratap K. Harbham
- West Midlands Academic Foundation Programme, University of Birmingham, Birmingham, United Kingdom
| | - Bhuvaneish T. Selvaraj
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
- UK Dementia Research Institute at University of Edinburgh, Edinburgh, United Kingdom
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Jenna M. Gregory
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
- UK Dementia Research Institute at University of Edinburgh, Edinburgh, United Kingdom
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, United Kingdom
- MRC Edinburgh Brain Bank, Academic Department of Neuropathology, University of Edinburgh, Edinburgh, United Kingdom
- Edinburgh Pathology, University of Edinburgh, Edinburgh, United Kingdom
| | - Suvankar Pal
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, United Kingdom
- Anne Rowling Regenerative Neurology Clinic, University of Edinburgh, Edinburgh, United Kingdom
| | - Giles E. Hardingham
- UK Dementia Research Institute at University of Edinburgh, Edinburgh, United Kingdom
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Siddharthan Chandran
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
- UK Dementia Research Institute at University of Edinburgh, Edinburgh, United Kingdom
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, United Kingdom
- Anne Rowling Regenerative Neurology Clinic, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Brain Development and Repair, InStem, Bengaluru, India
| | - Arpan R. Mehta
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
- UK Dementia Research Institute at University of Edinburgh, Edinburgh, United Kingdom
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, United Kingdom
- Anne Rowling Regenerative Neurology Clinic, University of Edinburgh, Edinburgh, United Kingdom
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
25
|
de Oliveira LG, Angelo YDS, Iglesias AH, Peron JPS. Unraveling the Link Between Mitochondrial Dynamics and Neuroinflammation. Front Immunol 2021; 12:624919. [PMID: 33796100 PMCID: PMC8007920 DOI: 10.3389/fimmu.2021.624919] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 02/25/2021] [Indexed: 12/13/2022] Open
Abstract
Neuroinflammatory and neurodegenerative diseases are a major public health problem worldwide, especially with the increase of life-expectancy observed during the last decades. For many of these diseases, we still lack a full understanding of their etiology and pathophysiology. Nonetheless their association with mitochondrial dysfunction highlights this organelle as an important player during CNS homeostasis and disease. Markers of Parkinson (PD) and Alzheimer (AD) diseases are able to induce innate immune pathways induced by alterations in mitochondrial Ca2+ homeostasis leading to neuroinflammation. Additionally, exacerbated type I IFN responses triggered by mitochondrial DNA (mtDNA), failures in mitophagy, ER-mitochondria communication and mtROS production promote neurodegeneration. On the other hand, regulation of mitochondrial dynamics is essential for CNS health maintenance and leading to the induction of IL-10 and reduction of TNF-α secretion, increased cell viability and diminished cell injury in addition to reduced oxidative stress. Thus, although previously solely seen as power suppliers to organelles and molecular processes, it is now well established that mitochondria have many other important roles, including during immune responses. Here, we discuss the importance of these mitochondrial dynamics during neuroinflammation, and how they correlate either with the amelioration or worsening of CNS disease.
Collapse
Affiliation(s)
- Lilian Gomes de Oliveira
- Neuroimmune Interactions Laboratory, Immunology Department - Institute of Biomedical Sciences (ICB) IV, University of São Paulo (USP), São Paulo, Brazil
- Neuroimmunology of Arboviruses Laboratory, Scientific Platform Pasteur-USP, University of São Paulo (USP), São Paulo, Brazil
| | - Yan de Souza Angelo
- Neuroimmune Interactions Laboratory, Immunology Department - Institute of Biomedical Sciences (ICB) IV, University of São Paulo (USP), São Paulo, Brazil
- Neuroimmunology of Arboviruses Laboratory, Scientific Platform Pasteur-USP, University of São Paulo (USP), São Paulo, Brazil
| | - Antonio H Iglesias
- Loyola University Medical Center, Stritch School of Medicine, Loyola University Chicago, Chicago, IL, United States
| | - Jean Pierre Schatzmann Peron
- Neuroimmune Interactions Laboratory, Immunology Department - Institute of Biomedical Sciences (ICB) IV, University of São Paulo (USP), São Paulo, Brazil
- Neuroimmunology of Arboviruses Laboratory, Scientific Platform Pasteur-USP, University of São Paulo (USP), São Paulo, Brazil
- Loyola University Medical Center, Stritch School of Medicine, Loyola University Chicago, Chicago, IL, United States
| |
Collapse
|
26
|
Kleerekooper I, Petzold A, Trip SA. Anterior visual system imaging to investigate energy failure in multiple sclerosis. Brain 2020; 143:1999-2008. [PMID: 32163545 DOI: 10.1093/brain/awaa049] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 11/26/2019] [Accepted: 12/24/2019] [Indexed: 12/12/2022] Open
Abstract
Mitochondrial failure and hypoxia are key contributors to multiple sclerosis pathophysiology. Importantly, improving mitochondrial function holds promise as a new therapeutic strategy in multiple sclerosis. Currently, studying mitochondrial changes in multiple sclerosis is hampered by a paucity of non-invasive techniques to investigate mitochondrial function of the CNS in vivo. It is against this backdrop that the anterior visual system provides new avenues for monitoring of mitochondrial changes. The retina and optic nerve are among the metabolically most active structures in the human body and are almost always affected to some degree in multiple sclerosis. Here, we provide an update on emerging technologies that have the potential to indirectly monitor changes of metabolism and mitochondrial function. We report on the promising work with optical coherence tomography, showing structural changes in outer retinal mitochondrial signal bands, and with optical coherence angiography, quantifying retinal perfusion at the microcapillary level. We show that adaptive optics scanning laser ophthalmoscopy can visualize live perfusion through microcapillaries and structural changes at the level of single photoreceptors and neurons. Advantages and limitations of these techniques are summarized with regard to future research into the pathology of the disease and as trial outcome measures.
Collapse
Affiliation(s)
- Iris Kleerekooper
- Department of Neuroinflammation, UCL Institute of Neurology, Queen Square, London, UK.,The National Hospital for Neurology and Neurosurgery, Queen Square, London, UK.,Moorfields Eye Hospital, City Road, London, UK
| | - Axel Petzold
- Department of Neuroinflammation, UCL Institute of Neurology, Queen Square, London, UK.,The National Hospital for Neurology and Neurosurgery, Queen Square, London, UK.,Moorfields Eye Hospital, City Road, London, UK.,Dutch Expertise Centre Neuro-ophthalmology and MS Centre Amsterdam, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - S Anand Trip
- Department of Neuroinflammation, UCL Institute of Neurology, Queen Square, London, UK.,The National Hospital for Neurology and Neurosurgery, Queen Square, London, UK
| |
Collapse
|
27
|
Filippatou AG, Moniruzzaman M, Sotirchos ES, Fitzgerald KC, Kalaitzidis G, Lambe J, Vasileiou E, Saidha S, Prince JL, Haughey N, Calabresi PA, Bhargava P. Serum ceramide levels are altered in multiple sclerosis. Mult Scler 2020; 27:1506-1519. [PMID: 33307993 DOI: 10.1177/1352458520971816] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
BACKGROUND Sphingolipids are myelin components and inflammatory signaling intermediates. Sphingolipid metabolism may be altered in people with multiple sclerosis (PwMS), but existing studies are limited by small sample sizes. OBJECTIVES To compare the levels of serum ceramides between PwMS and healthy controls (HCs) and to determine whether ceramide levels correlate with disability status, as well as optical coherence tomography (OCT)-derived rates of retinal layer atrophy. METHODS We performed targeted lipidomics analyses for 45 ceramides in PwMS (n = 251) and HCs (n = 68). For a subset of PwMS, baseline and 5-year Expanded Disability Status Scale (EDSS) assessments (n = 185), or baseline and serial spectral-domain OCT (n = 180) were assessed. RESULTS Several ceramides, including hexosylceramides, lactosylceramides, and dihydroceramides, were altered in PwMS compared with HCs. Higher levels of Cer16:0 were associated with higher odds of EDSS worsening at 5 years in univariable (odds ratio (OR) = 3.84, 95% confidence interval (CI) = 1.41-10.43) and multivariable analyses accounting for age, sex, and race (OR = 2.97, 95% CI = 1.03-8.59). Each 1 ng/mL higher concentration of Hex-Cer22:0 and DH-HexCer22:0 was associated with accelerated rates (μm/year) of ganglion cell + inner plexiform layer (-0.138 ± 0.053, p = 0.01; -0.158 ± 0.053, p = 0.003, respectively) and peripapillary retinal nerve fiber layer thinning (-0.305 ± 0.107, p = 0.004; -0.358 ± 0.106, p = 0.001, respectively). CONCLUSION Ceramide levels are altered in PwMS and may be associated with retinal neurodegeneration and physical disability.
Collapse
Affiliation(s)
- Angeliki G Filippatou
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mohammed Moniruzzaman
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elias S Sotirchos
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kathryn C Fitzgerald
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Grigorios Kalaitzidis
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jeffrey Lambe
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Eleni Vasileiou
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Shiv Saidha
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jerry L Prince
- Department of Electrical and Computer Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Norman Haughey
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peter A Calabresi
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Pavan Bhargava
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
28
|
Alves MA, Lamichhane S, Dickens A, McGlinchey A, Ribeiro HC, Sen P, Wei F, Hyötyläinen T, Orešič M. Systems biology approaches to study lipidomes in health and disease. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1866:158857. [PMID: 33278596 DOI: 10.1016/j.bbalip.2020.158857] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/13/2020] [Accepted: 11/27/2020] [Indexed: 12/15/2022]
Abstract
Lipids have many important biological roles, such as energy storage sources, structural components of plasma membranes and as intermediates in metabolic and signaling pathways. Lipid metabolism is under tight homeostatic control, exhibiting spatial and dynamic complexity at multiple levels. Consequently, lipid-related disturbances play important roles in the pathogenesis of most of the common diseases. Lipidomics, defined as the study of lipidomes in biological systems, has emerged as a rapidly-growing field. Due to the chemical and functional diversity of lipids, the application of a systems biology approach is essential if one is to address lipid functionality at different physiological levels. In parallel with analytical advances to measure lipids in biological matrices, the field of computational lipidomics has been rapidly advancing, enabling modeling of lipidomes in their pathway, spatial and dynamic contexts. This review focuses on recent progress in systems biology approaches to study lipids in health and disease, with specific emphasis on methodological advances and biomedical applications.
Collapse
Affiliation(s)
- Marina Amaral Alves
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku 20520, Finland
| | - Santosh Lamichhane
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku 20520, Finland
| | - Alex Dickens
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku 20520, Finland
| | - Aidan McGlinchey
- School of Medical Sciences, Örebro University, 702 81 Örebro, Sweden
| | | | - Partho Sen
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku 20520, Finland; School of Medical Sciences, Örebro University, 702 81 Örebro, Sweden
| | - Fang Wei
- Oil Crops Research Institute, Chinese Academy of Agricultural Sciences, Wuhan, PR China
| | | | - Matej Orešič
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku 20520, Finland; School of Medical Sciences, Örebro University, 702 81 Örebro, Sweden.
| |
Collapse
|
29
|
Park SJ, Choi JW. Brain energy metabolism and multiple sclerosis: progress and prospects. Arch Pharm Res 2020; 43:1017-1030. [PMID: 33119885 DOI: 10.1007/s12272-020-01278-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Accepted: 10/21/2020] [Indexed: 02/07/2023]
Abstract
Multiple sclerosis (MS) is an autoimmune disease accompanied with nerve pain and paralysis. Although various pathogenic causes of MS have been suggested, including genetic and environmental factors, how MS occurs remains unclear. Moreover, MS should be diagnosed based on clinical experiences because of no disease-specific biomarker and currently available treatments for MS just can reduce relapsing frequency or severity with little effects on disease disability. Therefore, more efforts are required to identify pathophysiology of MS and diagnosis markers. Recent evidence indicates another aspect of MS pathogenesis, energy failure in the central nervous system (CNS). For instance, inflammation that is a characteristic MS symptom and occurs frequently in the CNS of MS patients can result into energy failure in mitochondria and cytosol. Indeed, metabolomics studies for MS have reported energy failure in oxidative phosphorylation and alteration of aerobic glycolysis. Therefore, studies on the metabolism in the CNS may provide another insight for understanding complexity of MS and pathogenesis, which would facilitate the discovery of promising strategies for developing therapeutics to treat MS. This review will provide an overview on recent progress of metabolomic studies for MS, with a focus on the fluctuation of energy metabolism in MS.
Collapse
Affiliation(s)
- Sung Jean Park
- College of Pharmacy and Gachon Institute of Pharmaceutical Sciences, Gachon University, 191 Hambakmoero, Yeonsu-gu, Incheon, 21936, Korea.
| | - Ji Woong Choi
- College of Pharmacy and Gachon Institute of Pharmaceutical Sciences, Gachon University, 191 Hambakmoero, Yeonsu-gu, Incheon, 21936, Korea.
| |
Collapse
|
30
|
Amatruda M, Petracca M, Wentling M, Inbar B, Castro K, Chen EY, Kiebish MA, Edwards K, Inglese M, Casaccia P. Retrospective unbiased plasma lipidomic of progressive multiple sclerosis patients-identifies lipids discriminating those with faster clinical deterioration. Sci Rep 2020; 10:15644. [PMID: 32973249 PMCID: PMC7515876 DOI: 10.1038/s41598-020-72654-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 08/28/2020] [Indexed: 11/09/2022] Open
Abstract
The disease course of patients with a confirmed diagnosis of primary progressive multiple sclerosis (PPMS) is uncertain. In an attempt to identify potential signaling pathways involved in the evolution of the disease, we conducted an exploratory unbiased lipidomic analysis of plasma from non-diseased controls (n = 8) and patients with primary progressive MS (PPMS, n = 19) and either a rapid (PPMS-P, n = 9) or slow (PPMS-NP, n = 10) disease course based on worsening disability and/or MRI-visible appearance of new T2 lesions over a one-year-assessment. Partial least squares-discriminant analysis of the MS/MSALL lipidomic dataset, identified lipids driving the clustering of the groups. Among these lipids, sphingomyelin-d18:1/14:0 and mono-hexosylceramide-d18:1/20:0 were differentially abundant in the plasma of PPMS patients compared to controls and their levels correlated with MRI signs of disease progression. Lyso-phosphatidic acid-18:2 (LPA-18:2) was the only lipid with significantly lower abundance in PPMS patients with a rapidly deteriorating disease course, and its levels inversely correlated with the severity of the neurological deficit. Decreased levels of LPA-18:2 were detected in patients with more rapid disease progression, regardless of therapy and these findings were validated in an independent cohort of secondary progressive (SPMS) patients, but not in a third cohorts of relapsing–remitting (RRMS) patients. Collectively, our analysis suggests that sphingomyelin-d18:1/14:0, mono-hexosylceramide-d18:1/20:0, and LPA-18:2 may represent important targets for future studies aimed at understanding disease progression in MS.
Collapse
Affiliation(s)
- Mario Amatruda
- Advanced Science Research Center at the Graduate Center of the City University of New York, 85 Saint Nicholas Terrace, 4th Fl, New York, NY, 10031, USA. .,Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Maria Petracca
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Neurosciences, Reproductive and Odontostomatological Sciences, Federico II University, Naples, Italy
| | - Maureen Wentling
- Advanced Science Research Center at the Graduate Center of the City University of New York, 85 Saint Nicholas Terrace, 4th Fl, New York, NY, 10031, USA
| | - Benjamin Inbar
- Advanced Science Research Center at the Graduate Center of the City University of New York, 85 Saint Nicholas Terrace, 4th Fl, New York, NY, 10031, USA
| | - Kamilah Castro
- Advanced Science Research Center at the Graduate Center of the City University of New York, 85 Saint Nicholas Terrace, 4th Fl, New York, NY, 10031, USA.,Department of Neuroscience, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | | | | - Matilde Inglese
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DiNOGMI) and Center of Excellence for Biomedical Research (CEBR), Neurologic Clinic, University of Genoa, Genoa, Italy
| | - Patrizia Casaccia
- Advanced Science Research Center at the Graduate Center of the City University of New York, 85 Saint Nicholas Terrace, 4th Fl, New York, NY, 10031, USA. .,Department of Neuroscience, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
31
|
Zeng Y, Li Z, Zhu H, Gu Z, Zhang H, Luo K. Recent Advances in Nanomedicines for Multiple Sclerosis Therapy. ACS APPLIED BIO MATERIALS 2020; 3:6571-6597. [PMID: 35019387 DOI: 10.1021/acsabm.0c00953] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Yujun Zeng
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhiqian Li
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hongyan Zhu
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhongwei Gu
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hu Zhang
- Amgen Bioprocessing Centre, Keck Graduate Institute, Claremont, California 91711, United States
| | - Kui Luo
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
32
|
Signatures of cell stress and altered bioenergetics in skin fibroblasts from patients with multiple sclerosis. Aging (Albany NY) 2020; 12:15134-15156. [PMID: 32640422 PMCID: PMC7425440 DOI: 10.18632/aging.103612] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 06/05/2020] [Indexed: 12/14/2022]
Abstract
Multiple sclerosis (MS) is a central nervous system inflammatory demyelinating disease and the most common cause of non-traumatic disability in young adults. Despite progress in the treatment of the active relapsing disease, therapeutic options targeting irreversible progressive decline remain limited. Studies using skin fibroblasts derived from patients with neurodegenerative disorders demonstrate that cell stress pathways and bioenergetics are altered when compared to healthy individuals. However, findings in MS skin fibroblasts are limited. Here, we collected skin fibroblasts from 24 healthy control individuals, 30 patients with MS, and ten with amyotrophic lateral sclerosis (ALS) to investigate altered cell stress profiles. We observed endoplasmic reticulum swelling in MS skin fibroblasts, and increased gene expression of cell stress markers including BIP, ATF4, CHOP, GRP94, P53, and P21. When challenged against hydrogen peroxide, MS skin fibroblasts had reduced resiliency compared to ALS and controls. Mitochondrial and glycolytic functions were perturbed in MS skin fibroblasts while exhibiting a significant increase in lactate production over ALS and controls. Our results suggest that MS skin fibroblasts have an underlying stress phenotype, which may be disease specific. Interrogating MS skin fibroblasts may provide patient specific molecular insights and aid in prognosis, diagnosis, and therapeutic testing enhancing individualized medicine.
Collapse
|
33
|
Vanherle S, Haidar M, Irobi J, Bogie JF, Hendriks JJ. Extracellular vesicle-associated lipids in central nervous system disorders. Adv Drug Deliv Rev 2020; 159:322-331. [PMID: 32360577 DOI: 10.1016/j.addr.2020.04.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/03/2020] [Accepted: 04/26/2020] [Indexed: 12/19/2022]
Abstract
Increasing evidence indicates that lipid metabolism is disturbed in central nervous system (CNS) disorders, such as multiple sclerosis, Alzheimer's, and Parkinson's disease. Extracellular vesicles (EVs), including exosomes and microvesicles, are nanosized particles that play an essential role in intercellular communication and tissue homeostasis by transporting diverse biologically active molecules, including a large variety of lipid species. In the last decade, studies defined that changes in the EV lipidome closely correlate with disease-progression and -remission in CNS disorders. In this review, we summarize and discuss these changes in the EV lipidome and elaborate on the impact of different EV-associated lipids on pathological processes in CNS disorders. We conclude that EV-associated lipids are closely associated with neuroinflammation, CNS repair, and pathological protein aggregation in CNS disorders, and that modulation of the EV lipidome represents a promising therapeutic strategy to halt disease progression in multiple sclerosis, Alzheimer's, and Parkinson's disease. Moreover, we predict that disease-stage specific EV-associated lipid signatures can be invaluable markers for the diagnosis and early detection of CNS disorders in the future.
Collapse
|
34
|
Jakimovski D, Bergsland N, Dwyer MG, Hagemeier J, Ramasamy DP, Szigeti K, Guttuso T, Lichter D, Hojnacki D, Weinstock-Guttman B, Benedict RHB, Zivadinov R. Long-standing multiple sclerosis neurodegeneration: volumetric magnetic resonance imaging comparison to Parkinson's disease, mild cognitive impairment, Alzheimer's disease, and elderly healthy controls. Neurobiol Aging 2020; 90:84-92. [PMID: 32147244 DOI: 10.1016/j.neurobiolaging.2020.02.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 01/30/2020] [Accepted: 02/03/2020] [Indexed: 12/16/2022]
Abstract
Multiple sclerosis (MS) exhibits neurodegeneration driven disability progression. We compared the extent of neurodegeneration among 112 long-standing MS patients, 37 Parkinson's disease (PD) patients, 34 amnestic mild cognitive impairment (aMCI) patients, 37 Alzheimer's disease (AD) patients, and 184 healthy controls. 3T MRI volumes of whole brain (WBV), white matter (WMV), gray matter (GMV), cortical (CV), deep gray matter (DGM), and nuclei-specific volumes of thalamus, caudate, putamen, globus pallidus, and hippocampus were derived with SIENAX and FIRST software. Аge and sex-adjusted analysis of covariance was used. WBV was not significantly different between diseases. MS had significantly lower WMV compared to other disease groups (p < 0.021). Only AD had smaller GMV and CV when compared to MS (both p < 0.001). MS had smaller DGM volume than PD and aMCI (p < 0.001 and p = 0.026, respectively) and lower thalamic volume when compared to all other neurodegenerative diseases (p < 0.008). Long-standing MS exhibits comparable global atrophy with lower WMV and thalamic volume when compared to other classical neurodegenerative diseases.
Collapse
Affiliation(s)
- Dejan Jakimovski
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Niels Bergsland
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA; IRCCS, Fondazione Don Carlo Gnocchi, Milan, Italy
| | - Michael G Dwyer
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Jesper Hagemeier
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Deepa P Ramasamy
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Kinga Szigeti
- Jacobs Multiple Sclerosis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, USA
| | - Thomas Guttuso
- Jacobs Multiple Sclerosis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, USA
| | - David Lichter
- Jacobs Multiple Sclerosis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, USA
| | - David Hojnacki
- Jacobs Multiple Sclerosis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, USA; Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, USA
| | - Bianca Weinstock-Guttman
- Jacobs Multiple Sclerosis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, USA; Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, USA
| | - Ralph H B Benedict
- Jacobs Multiple Sclerosis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, USA; Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, USA
| | - Robert Zivadinov
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA; Jacobs Multiple Sclerosis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, USA; Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, USA; Center for Biomedical Imaging at Clinical Translational Science Institute, University at Buffalo, State University of New York, Buffalo, NY, USA.
| |
Collapse
|
35
|
Casaccia P. Emerging concepts in neuroscience research: 2019 highlights. Lancet Neurol 2020; 19:21-22. [PMID: 31839244 DOI: 10.1016/s1474-4422(19)30452-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 11/25/2019] [Indexed: 10/25/2022]
Affiliation(s)
- Patrizia Casaccia
- Advanced Science Research Center, Graduate Center of the City University of New York, New York, NY 10031, USA; Icahn School of Medicine, Mount Sinai Hospital, New York, NY, USA.
| |
Collapse
|
36
|
Abstract
Emerging data point to important contributions of both autoimmune inflammation and progressive degeneration in the pathophysiology of multiple sclerosis (MS). Unfortunately, after decades of intensive investigation, the fundamental cause remains unknown. A large body of research on the immunobiology of MS has resulted in a variety of anti-inflammatory therapies that are highly effective at reducing brain inflammation and clinical/radiological relapses. However, despite potent suppression of inflammation, benefit in the more important and disabling progressive phase is extremely limited; thus, progressive MS has emerged as the greatest challenge for the MS research and clinical communities. Data obtained over the years point to a complex interplay between environment (e.g., the near-absolute requirement of Epstein-Barr virus exposure), immunogenetics (strong associations with a large number of immune genes), and an ever more convincing role of an underlying degenerative process resulting in demyelination (in both white and grey matter regions), axonal and neuro-synaptic injury, and a persistent innate inflammatory response with a seemingly diminishing role of T cell-mediated autoimmunity as the disease progresses. Together, these observations point toward a primary degenerative process, one whose cause remains unknown but one that entrains a nearly ubiquitous secondary autoimmune response, as a likely sequence of events underpinning this disease. Here, we briefly review what is known about the potential pathophysiological mechanisms, focus on progressive MS, and discuss the two main hypotheses of MS pathogenesis that are the topic of vigorous debate in the field: whether primary autoimmunity or degeneration lies at the foundation. Unravelling this controversy will be critically important for developing effective new therapies for the most disabling later phases of this disease.
Collapse
Affiliation(s)
- Peter K. Stys
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Medicine University of Calgary, Calgary, Alberta, Canada
| | - Shigeki Tsutsui
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Medicine University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
37
|
Multiple Sclerosis: Melatonin, Orexin, and Ceramide Interact with Platelet Activation Coagulation Factors and Gut-Microbiome-Derived Butyrate in the Circadian Dysregulation of Mitochondria in Glia and Immune Cells. Int J Mol Sci 2019; 20:ijms20215500. [PMID: 31694154 PMCID: PMC6862663 DOI: 10.3390/ijms20215500] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 10/30/2019] [Accepted: 11/04/2019] [Indexed: 12/24/2022] Open
Abstract
Recent data highlight the important roles of the gut microbiome, gut permeability, and alterations in mitochondria functioning in the pathophysiology of multiple sclerosis (MS). This article reviews such data, indicating two important aspects of alterations in the gut in the modulation of mitochondria: (1) Gut permeability increases toll-like receptor (TLR) activators, viz circulating lipopolysaccharide (LPS), and exosomal high-mobility group box (HMGB)1. LPS and HMGB1 increase inducible nitric oxide synthase and superoxide, leading to peroxynitrite-driven acidic sphingomyelinase and ceramide. Ceramide is a major driver of MS pathophysiology via its impacts on glia mitochondria functioning; (2) Gut dysbiosis lowers production of the short-chain fatty acid, butyrate. Butyrate is a significant positive regulator of mitochondrial function, as well as suppressing the levels and effects of ceramide. Ceramide acts to suppress the circadian optimizers of mitochondria functioning, viz daytime orexin and night-time melatonin. Orexin, melatonin, and butyrate increase mitochondria oxidative phosphorylation partly via the disinhibition of the pyruvate dehydrogenase complex, leading to an increase in acetyl-coenzyme A (CoA). Acetyl-CoA is a necessary co-substrate for activation of the mitochondria melatonergic pathway, allowing melatonin to optimize mitochondrial function. Data would indicate that gut-driven alterations in ceramide and mitochondrial function, particularly in glia and immune cells, underpin MS pathophysiology. Aryl hydrocarbon receptor (AhR) activators, such as stress-induced kynurenine and air pollutants, may interact with the mitochondrial melatonergic pathway via AhR-induced cytochrome P450 (CYP)1b1, which backward converts melatonin to N-acetylserotonin (NAS). The loss of mitochnodria melatonin coupled with increased NAS has implications for altered mitochondrial function in many cell types that are relevant to MS pathophysiology. NAS is increased in secondary progressive MS, indicating a role for changes in the mitochondria melatonergic pathway in the progression of MS symptomatology. This provides a framework for the integration of diverse bodies of data on MS pathophysiology, with a number of readily applicable treatment interventions, including the utilization of sodium butyrate.
Collapse
|