1
|
Sabaie H, Taghavi Rad A, Shabestari M, Habibi D, Saadattalab T, Seddiq S, Saeidian AH, Zahedi AS, Sanoie M, Vahidnezhad H, Zarkesh M, Foroutani L, Hakonarson H, Azizi F, Hedayati M, Daneshpour MS, Akbarzadeh M. Mitochondrial DNA Copy Number as a Hidden Player in the Progression of Multiple Sclerosis: A Bidirectional Two-Sample Mendelian Randomization Study. Mol Neurobiol 2025:10.1007/s12035-025-04980-9. [PMID: 40307428 DOI: 10.1007/s12035-025-04980-9] [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: 10/23/2024] [Accepted: 04/18/2025] [Indexed: 05/02/2025]
Abstract
The relationship between mitochondrial DNA copy number (mtDNA-CN) and multiple sclerosis (MS) progression remains unclear, as previous observational studies have reported conflicting results. This study aimed to clarify the association between mtDNA-CN and MS progression using a bidirectional two-sample Mendelian randomization (MR) approach. MR analyses were conducted using the latest summary statistics from genome-wide association studies (GWAS) on mtDNA-CN and MS progression. Single-nucleotide polymorphisms (SNPs) associated with mtDNA-CN were extracted from 383,476 participants of European ancestry in the UK Biobank, while SNPs associated with MS severity were obtained from the International Multiple Sclerosis Genetics Consortium (IMSGC), comprising 12,584 cases of European ancestry. The inverse variance weighted (IVW) method was used as the primary analysis. Potential heterogeneity and pleiotropy were evaluated, and sensitivity analyses were performed to ensure the robustness of the results. The forward MR analysis using the IVW method revealed no significant association between mtDNA-CN and MS progression (P = 0.487). However, reverse MR analysis identified a causal association between MS progression and mtDNA-CN (β = - 0.010, 95% CI = - 0.019 to - 0.001, P = 0.036). No evidence of heterogeneity or horizontal pleiotropy was found in the analyses. Sensitivity analyses yielded consistent results. Our findings suggest that MS progression may causally influence mtDNA-CN, highlighting the crucial role of mitochondria in the pathophysiology of MS. However, further research is needed to confirm mtDNA-CN as a reliable biomarker and a deeper understanding of the molecular mechanisms is necessary to develop targeted therapeutic interventions.
Collapse
Affiliation(s)
- Hani Sabaie
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Molecular Biology, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Taghavi Rad
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Molecular Biology, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Motahareh Shabestari
- Yazd Cardiovascular Research Center, Non-Communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Danial Habibi
- Department of Epidemiology and Biostatistics, School of Public Health, Babol University of Medical Sciences, Babol, Iran
| | - Toktam Saadattalab
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Sahar Seddiq
- Osteoporosis Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Hossein Saeidian
- Center for Applied Genomics (CAG), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Asiyeh Sadat Zahedi
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Molecular Biology, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Sanoie
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Molecular Biology, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hassan Vahidnezhad
- Center for Applied Genomics (CAG), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Maryam Zarkesh
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Molecular Biology, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Laleh Foroutani
- Department of Surgery, University of California, San Francisco, CA, USA
| | - Hakon Hakonarson
- Center for Applied Genomics (CAG), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Pulmonary Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Fereidoun Azizi
- Endocrine Research Center, Research Institute for Endocrine Disorders, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Hedayati
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Molecular Biology, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Sadat Daneshpour
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Molecular Biology, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mahdi Akbarzadeh
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Molecular Biology, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Campetella L, Smolik K, Farina A, Joubert B, Muñiz-Castrillo S, Desestret V, Honnorat J. Neurodegeneration and the immune system: lessons from autoimmune encephalitis. J Neurol 2025; 272:359. [PMID: 40274643 PMCID: PMC12021719 DOI: 10.1007/s00415-025-13094-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/06/2025] [Accepted: 04/10/2025] [Indexed: 04/26/2025]
Abstract
The spectrum of autoimmune encephalitis (AE) is expanding to atypical clinical presentations that can mimic neurodegenerative disorders. Among the autoantibodies most frequently associated with manifestations mimicking neurodegenerative disorders-such as dementia, parkinsonism, ataxia and motor neuron disease-IgLON5-, LGI1- and CASPR2-antibodies, predominantly of the IgG4 subclass and associated with specific HLA haplotypes, are the most common. Since these forms of autoimmune encephalitis often lack inflammatory findings in cerebrospinal fluid or magnetic resonance imaging, recognizing clinical 'red flags' suggestive of an autoimmune etiology is crucial for accurate diagnosis and timely initiation of immunotherapy. Interestingly, in these forms of autoimmune encephalitis, both inflammatory and neurodegenerative disease mechanisms may be involved. The neurodegenerative component may result directly from antibody effects (e.g., tau deposition in IgLON5-antibody disease) or arise through other mechanisms (e.g., seizures or exacerbation of pre-existing pathology). Moreover, neuroinflammation has recently emerged as a key contributor to primary neurodegenerative disorders. For instance, microglial activation promotes tau pathology propagation, as observed in Alzheimer's disease and other primary neurodegenerative disorders. While the precise mechanisms linking inflammation and neurodegeneration remain to be fully understood, further research into the interplay between autoimmunity and neurodegeneration may enhance our understanding of disease mechanisms and expand therapeutic opportunities in both autoimmune and neurodegenerative neurological disorders.
Collapse
Affiliation(s)
- Lucia Campetella
- French Reference Center for Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis, Hospices Civils de Lyon, 59 Boulevard Pinel, Bron Cedex, 69677, Lyon, France
- MeLiS - UCBL-CNRS UMR 5284 - INSERM U1314, Université Claude Bernard Lyon 1, Lyon, France
| | - Krzysztof Smolik
- French Reference Center for Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis, Hospices Civils de Lyon, 59 Boulevard Pinel, Bron Cedex, 69677, Lyon, France
- MeLiS - UCBL-CNRS UMR 5284 - INSERM U1314, Université Claude Bernard Lyon 1, Lyon, France
- Department of Biomedical, Metabolic and Neurosciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Antonio Farina
- French Reference Center for Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis, Hospices Civils de Lyon, 59 Boulevard Pinel, Bron Cedex, 69677, Lyon, France
- MeLiS - UCBL-CNRS UMR 5284 - INSERM U1314, Université Claude Bernard Lyon 1, Lyon, France
- Department of Neuroscience, Psychology, Pharmacology and Child Health, University of Florence, Florence, Italy
| | - Bastien Joubert
- French Reference Center for Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis, Hospices Civils de Lyon, 59 Boulevard Pinel, Bron Cedex, 69677, Lyon, France
- MeLiS - UCBL-CNRS UMR 5284 - INSERM U1314, Université Claude Bernard Lyon 1, Lyon, France
- Neurology Department, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, 69495, Oullins-Pierre-Bénite, France
| | - Sergio Muñiz-Castrillo
- French Reference Center for Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis, Hospices Civils de Lyon, 59 Boulevard Pinel, Bron Cedex, 69677, Lyon, France
- MeLiS - UCBL-CNRS UMR 5284 - INSERM U1314, Université Claude Bernard Lyon 1, Lyon, France
- Neurology Department, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041, Madrid, Spain
| | - Virginie Desestret
- French Reference Center for Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis, Hospices Civils de Lyon, 59 Boulevard Pinel, Bron Cedex, 69677, Lyon, France
- MeLiS - UCBL-CNRS UMR 5284 - INSERM U1314, Université Claude Bernard Lyon 1, Lyon, France
- Neurocognition and Neuro-Ophthalmology Department, Hôpital Pierre Wertheimer, Hospices Civils de Lyon, Lyon, France
| | - Jérôme Honnorat
- French Reference Center for Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis, Hospices Civils de Lyon, 59 Boulevard Pinel, Bron Cedex, 69677, Lyon, France.
- MeLiS - UCBL-CNRS UMR 5284 - INSERM U1314, Université Claude Bernard Lyon 1, Lyon, France.
| |
Collapse
|
3
|
Belen S, Patt N, Kupjetz M, Ueland PM, McCann A, Gonzenbach R, Bansi J, Zimmer P. Vitamin B 6 status is related to disease severity and modulated by endurance exercise in individuals with multiple sclerosis: a secondary analysis of a randomized controlled trial. Am J Clin Nutr 2025:S0002-9165(25)00200-X. [PMID: 40252731 DOI: 10.1016/j.ajcnut.2025.04.014] [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: 11/19/2024] [Revised: 04/02/2025] [Accepted: 04/16/2025] [Indexed: 04/21/2025] Open
Abstract
BACKGROUND Low circulating concentrations of B vitamins are linked to various chronic and neurodegenerative diseases. Notably, pyridoxal 5'-phosphate (vitamin B6) deficiency is linked to altered inflammatory responses and cellular immune function, both critical in multiple sclerosis (MS). Nevertheless, most MS research has focused on folate (vitamin B9) and vitamin B12, leaving other B vitamins understudied. OBJECTIVES This secondary analysis investigated B-vitamin serum concentrations and related metabolites across MS phenotypes (primary progressive MS, relapsing-remitting MS, and secondary progressive MS) and disease severity levels. Additionally, the impact of endurance exercise on B-vitamin concentrations was investigated. METHODS In total, 106 individuals with MS participated in a randomized controlled trial, including different endurance exercise conditions. Serum B-vitamin concentrations were analyzed in 99 participants before and after 3 wk of intervention. Before analysis, participants were dichotomized to 1 of the 2 disability groups based on their expanded disability status scale (EDSS) score: EDSS≥4.5 (n = 47, EDSS: 5.86 ± 0.56) and EDSS<4 (n = 52, EDSS: 3.59 ± 0.83). RESULTS Higher EDSS scores were associated with lower pyridoxal 5'-phosphate (vitamin B-6) concentrations (rs: -0.32; 95% CI: -0.49, -0.12; P = 0.011), with the EDSS≥4.5 group also showing lower baseline pyridoxal 5'-phosphate (vitamin B6) concentrations (β: -0.18; 95% CI: -0.30, -0.07; P = 0.007) than the EDSS<4 group. Significant time × EDSS group interactions were evident for pyridoxal 5'-phosphate (vitamin B6; β: 0.05; 95% CI: 0.02, 0.08; P = 0.011), pyridoxal (vitamin B6; β: 0.05; 95% CI: 0.02, 0.09; P = 0.005), and riboflavin (vitamin B2; β: 0.06; 95% CI: 0.02, 0.09; P = 0.008), showing increases in these vitamers in the EDSS≥4.5 group postexercise. N1-Methylnicotinamide (vitamin B3; β: -0.11; 95% CI: -0.15, -0.06; P < 0.001) decreased in both groups over time. CONCLUSIONS Disease severity is associated with distinct B-vitamin profiles in individuals with MS, although endurance exercise appears to modify specific B-vitamin concentrations. This trial was registered at clinicaltrials.gov as NCT04356248.
Collapse
Affiliation(s)
- Sergen Belen
- Department of Sports Medicine, Institute of Sport and Sport Science, TU Dortmund University, Dortmund, Germany; Department of Molecular and Cellular Sports Medicine, Institute of Cardiology and Sports Medicine, German Sports University Cologne, Cologne, Germany
| | - Nadine Patt
- Department of Neurology, Rehabilitation Centre Valens, Clinics of Valens, Valens, Switzerland
| | - Marie Kupjetz
- Department of Sports Medicine, Institute of Sport and Sport Science, TU Dortmund University, Dortmund, Germany
| | | | | | - Roman Gonzenbach
- Department of Neurology, Rehabilitation Centre Valens, Clinics of Valens, Valens, Switzerland
| | - Jens Bansi
- Department of Neurology, Rehabilitation Centre Valens, Clinics of Valens, Valens, Switzerland; Department of Health, Physiotherapy, OST-Eastern Switzerland University of Applied Science, Sankt Gallen, Switzerland
| | - Philipp Zimmer
- Department of Sports Medicine, Institute of Sport and Sport Science, TU Dortmund University, Dortmund, Germany.
| |
Collapse
|
4
|
De Meo E, Prados Carrasco F, Brown JWL, Coles AJ, Cunniffe NG, Jolly AE, Kanber B, Samson R, Barkhof F, Chard D. An MRI assessment of mechanisms underlying lesion growth and shrinkage in multiple sclerosis. Ann Clin Transl Neurol 2025; 12:686-700. [PMID: 39869436 PMCID: PMC12040513 DOI: 10.1002/acn3.52308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 12/18/2024] [Accepted: 01/09/2025] [Indexed: 01/29/2025] Open
Abstract
OBJECTIVE To assess the pathological mechanisms contributing to white matter (WM) lesion expansion or contraction and remyelination in multiple sclerosis (MS). METHODS We assessed 1,613 lesions in 49 people with relapsing-remitting MS in the CCMR-One bexarotene trial (EudraCT 2014-003145-99). We measured lesion orientation relative to WM tracts, surface-in gradients and veins. Jacobian deformation was used to assess lesion expansion over 6 months, while magnetization transfer ratio (MTR) imaging was used to assess remyelination. RESULTS At baseline, 33% of lesions were aligned with veins, 2% along WM tracts, 0% with surface-in gradients, and 4% orthogonal to veins. No significant differences were observed in lesion shape, while lesions aligned with surface-in gradients and with veins had lower volume compared to all remaining orientations. At follow-up, 13% of lesions expanded and 7% contracted. The directions for both expansion and contraction were 18% and 8%, respectively, along WM tracts, 20% and 15% parallel to veins, 22% and 23% orthogonal to veins and 0% and 1% along surface-in gradients. Bexarotene had no effect on lesion expansion or contraction, but MTR significantly increased in lesions aligned with surface-in gradients and veins. INTERPRETATION Lesion expansion and shrinkage are affected by venous and WM tract factors, but these do not influence bexarotene's capacity to promote remyelination. This, instead, appears to be affected by surface-in factors. To limit lesion expansion and maximize tissue repair, multiple processes may need to be targeted.
Collapse
Affiliation(s)
- Ermelinda De Meo
- NMR Research Unit, Queen Square Multiple Sclerosis Centre, UCL Queen Square Institute of NeurologyUniversity College LondonLondonUK
| | - Ferran Prados Carrasco
- NMR Research Unit, Queen Square Multiple Sclerosis Centre, UCL Queen Square Institute of NeurologyUniversity College LondonLondonUK
- Universitat Oberta de CatalunyaBarcelonaSpain
- Department of Medical Physics and Biomedical Engineering, Centre for Medical Image Computing (CMIC)University College LondonLondonUK
| | | | - Alasdair J. Coles
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
| | - Nick G. Cunniffe
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
| | - Amy E. Jolly
- NMR Research Unit, Queen Square Multiple Sclerosis Centre, UCL Queen Square Institute of NeurologyUniversity College LondonLondonUK
| | - Baris Kanber
- NMR Research Unit, Queen Square Multiple Sclerosis Centre, UCL Queen Square Institute of NeurologyUniversity College LondonLondonUK
- Department of Medical Physics and Biomedical Engineering, Centre for Medical Image Computing (CMIC)University College LondonLondonUK
| | - Rebecca Samson
- NMR Research Unit, Queen Square Multiple Sclerosis Centre, UCL Queen Square Institute of NeurologyUniversity College LondonLondonUK
| | - Frederik Barkhof
- NMR Research Unit, Queen Square Multiple Sclerosis Centre, UCL Queen Square Institute of NeurologyUniversity College LondonLondonUK
- National Institute for Health Research (NIHR) University College London Hospitals (UCLH) Biomedical Research CentreLondonUK
- Radiology and Nuclear MedicineAmsterdam UMCAmsterdamthe Netherlands
| | - Declan Chard
- NMR Research Unit, Queen Square Multiple Sclerosis Centre, UCL Queen Square Institute of NeurologyUniversity College LondonLondonUK
- National Institute for Health Research (NIHR) University College London Hospitals (UCLH) Biomedical Research CentreLondonUK
| |
Collapse
|
5
|
Schubert C, Schulz K, Sonner JK, Hadjilaou A, Seemann AL, Gierke J, Vieira V, Meurs N, Woo MS, Lohr C, Morellini F, Hirnet D, Friese MA. Neuroinflammation causes mitral cell dysfunction and olfactory impairment in a multiple sclerosis model. J Neuroinflammation 2025; 22:71. [PMID: 40057769 PMCID: PMC11889885 DOI: 10.1186/s12974-025-03388-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Accepted: 02/19/2025] [Indexed: 05/13/2025] Open
Abstract
BACKGROUND Olfactory dysfunction is an underestimated symptom in multiple sclerosis (MS). Here, we examined the pathogenic mechanisms underlying inflammation-induced dysfunction of the olfactory bulb using the animal model of MS, experimental autoimmune encephalomyelitis (EAE). RESULTS Reduced olfactory function in EAE was associated with the degeneration of short-axon neurons, immature neurons, and both mitral and tufted cells, along with their synaptic interactions and axonal repertoire. To dissect the mechanisms underlying the susceptibility of mitral cells, the main projection neurons of the olfactory bulb, we profiled their responses to neuroinflammation by single-nucleus RNA sequencing followed by functional validation. Neuroinflammation resulted in the induction of potassium channel transcripts in mitral cells, which was reflected in increased halothane-induced outward currents of these cells, likely contributing to the impaired olfaction in EAE animals. CONCLUSION This study reveals the crucial role of mitral cells and their potassium channel activity in the olfactory bulb during EAE, thereby enhancing our understanding of neuroinflammation-induced neurodegeneration in MS.
Collapse
Affiliation(s)
- Charlotte Schubert
- Institute of Neuroimmunology and Multiple Sclerosis (INIMS), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kristina Schulz
- Institute of Cell and Systems Biology of Animals, University of Hamburg, Hamburg, Germany
| | - Jana K Sonner
- Institute of Neuroimmunology and Multiple Sclerosis (INIMS), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alexandros Hadjilaou
- Institute of Neuroimmunology and Multiple Sclerosis (INIMS), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Protozoa Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Anna-Lena Seemann
- Institute of Neuroimmunology and Multiple Sclerosis (INIMS), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute of Cell and Systems Biology of Animals, University of Hamburg, Hamburg, Germany
| | - Janine Gierke
- Institute of Cell and Systems Biology of Animals, University of Hamburg, Hamburg, Germany
| | - Vanessa Vieira
- Institute of Neuroimmunology and Multiple Sclerosis (INIMS), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nina Meurs
- Institute of Neuroimmunology and Multiple Sclerosis (INIMS), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marcel S Woo
- Institute of Neuroimmunology and Multiple Sclerosis (INIMS), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian Lohr
- Institute of Cell and Systems Biology of Animals, University of Hamburg, Hamburg, Germany
| | - Fabio Morellini
- Research Group Behavioral Biology, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Daniela Hirnet
- Institute of Cell and Systems Biology of Animals, University of Hamburg, Hamburg, Germany
| | - Manuel A Friese
- Institute of Neuroimmunology and Multiple Sclerosis (INIMS), University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
6
|
Pfeffer LK, Fischbach F, Heesen C, Friese MA. Current state and perspectives of CAR T cell therapy in central nervous system diseases. Brain 2025; 148:723-736. [PMID: 39530593 DOI: 10.1093/brain/awae362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 10/03/2024] [Accepted: 10/20/2024] [Indexed: 11/16/2024] Open
Abstract
B cell-directed CAR T cell therapy has fundamentally changed the treatment of haematological malignancies, and its scope of application is rapidly expanding to include other diseases such as solid tumours or autoimmune disorders. Therapy-refractoriness remains an important challenge in various inflammatory and non-inflammatory disorders of the CNS. The reasons for therapy failure are diverse and include the limited access current therapies have to the CNS, as well as enormous inter- and intra-individual disease heterogeneity. The tissue-penetrating properties of CAR T cells make them a promising option for overcoming this problem and tackling pathologies directly within the CNS. First application of B cell-directed CAR T cells in neuromyelitis optica spectrum disorder and multiple sclerosis patients has recently revealed promising outcomes, expanding the potential of CAR T cell therapy to encompass CNS diseases. Additionally, the optimization of CAR T cells for the therapy of gliomas is a growing field. As a further prospect, preclinical data reveal the potential benefits of CAR T cell therapy in the treatment of primary neurodegenerative diseases such as Alzheimer's disease. Considering the biotechnological optimizations in the field of T cell engineering, such as extension to target different antigens or variation of the modified T cell subtype, new and promising fields of CAR T cell application are rapidly opening up. These innovations offer the potential to address the complex pathophysiological properties of CNS diseases. To use CAR T cell therapy optimally to treat CNS diseases in the future while minimizing therapy risks, further mechanistic research and prospective controlled trials are needed to assess seriously the disease and patient-specific risk-benefit ratio.
Collapse
Affiliation(s)
- Lena Kristina Pfeffer
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
- Department of Neurology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Felix Fischbach
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
- Department of Neurology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Christoph Heesen
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
- Department of Neurology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Manuel A Friese
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
- Department of Neurology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| |
Collapse
|
7
|
Belbasis L, Morris S, van Duijn C, Bennett D, Walters R. Mendelian randomization identifies proteins involved in neurodegenerative diseases. Brain 2025:awaf018. [PMID: 40037332 DOI: 10.1093/brain/awaf018] [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: 02/24/2024] [Revised: 10/26/2024] [Accepted: 12/20/2024] [Indexed: 03/06/2025] Open
Abstract
Proteins are involved in multiple biological functions. High-throughput technologies have allowed the measurement of thousands of proteins in population biobanks. In this study, we aimed to identify proteins related to Alzheimer's disease, Parkinson's disease, multiple sclerosis and amyotrophic lateral sclerosis by leveraging large-scale genetic and proteomic data. We performed a two-sample cis Mendelian randomization study by selecting instrumental variables for the abundance of >2700 proteins measured by either Olink or SomaScan platforms in plasma from the UK Biobank and the deCODE Health Study. We also used the latest publicly available genome-wide association studies for the neurodegenerative diseases of interest. The potentially causal effect of proteins on neurodegenerative diseases was estimated based on the Wald ratio. We tested 13 377 protein-disease associations, identifying 169 associations that were statistically significant (5% false discovery rate). Evidence of co-localization between plasma protein abundance and disease risk (posterior probability > 0.80) was identified for 61 protein-disease pairs, leading to 50 unique protein-disease associations. Notably, 23 of 50 protein-disease associations corresponded to genetic loci not previously reported by genome-wide association studies. The two-sample Mendelian randomization and co-localization analysis also showed that APOE abundance in plasma was associated with three subcortical volumes (hippocampus, amygdala and nucleus accumbens) and white matter hyper-intensities, whereas PILRA and PILRB abundance in plasma was associated with caudate nucleus volume. Our study provided a comprehensive assessment of the effect of the human proteome that is currently measurable through two different platforms on neurodegenerative diseases. The newly associated proteins indicated the involvement of complement (C1S and C1R), microglia (SIRPA, SIGLEC9 and PRSS8) and lysosomes (CLN5) in Alzheimer's disease; the interleukin-6 pathway (CTF1) in Parkinson's disease; lysosomes (TPP1), blood-brain barrier integrity (MFAP2) and astrocytes (TNFSF13) in amyotrophic lateral sclerosis; and blood-brain barrier integrity (VEGFB), oligodendrocytes (PARP1), node of Ranvier and dorsal root ganglion (NCS1, FLRT3 and CDH15) and the innate immune system (CR1, AHSG and WARS) in multiple sclerosis. Our study demonstrates how harnessing large-scale genomic and proteomic data can yield new insights into the role of the plasma proteome in the pathogenesis of neurodegenerative diseases.
Collapse
Affiliation(s)
- Lazaros Belbasis
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Sam Morris
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Cornelia van Duijn
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Derrick Bennett
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Robin Walters
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| |
Collapse
|
8
|
Rahimi Darehbagh R, Khanmohammadi S, Rezaei N. The role of mitochondrial DNA variants and dysfunction in the pathogenesis and progression of multiple sclerosis. Mitochondrion 2025; 81:102002. [PMID: 39732186 DOI: 10.1016/j.mito.2024.102002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 08/10/2024] [Accepted: 12/24/2024] [Indexed: 12/30/2024]
Abstract
Multiple sclerosis (MS) is a chronic autoimmune disease that affects the central nervous system (CNS). The etiology of MS remains elusive, with a complex interplay of genetic and environmental factors contributing to its pathogenesis. Recent studies showed mitochondrial DNA (mtDNA) as a potential player in the development and progression of MS. These studies encompassed mtDNA variants, copy number variations, and haplogroups. This narrative review aims to synthesize the current understanding of the role of mtDNA's in MS. The findings of this review suggest that mtDNA may indeed play a role in the development and progression of MS. Several studies have reported an association between mtDNA variants and increased susceptibility to MS, while others have found a link between mtDNA copy number variations and disease severity. Furthermore, specific mtDNA haplogroups have been demonstrated to confer protection against MS. MtDNA alterations may make neurons and oligodendrocytes more susceptible to inflammatory and oxidative stress, causing demyelination and axonal degeneration in MS patients. In conclusion, this review underscores the potential significance of mtDNA in the pathogenesis of MS and highlights the need for further research to fully elucidate its role. A deeper understanding of mtDNA's involvement in MS may pave the way for the development of novel therapeutic strategies to combat this debilitating disease.
Collapse
Affiliation(s)
- Ramyar Rahimi Darehbagh
- Student Research Committee, Kurdistan University of Medical Sciences, Sanandaj, Iran; Nanoclub Elites Association, Tehran, Iran; Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran; Universal Scientific Education and Research Network (USERN), Sanandaj, Kurdistan, Iran
| | - Shaghayegh Khanmohammadi
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran; School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
9
|
Zone-Abid I, Maaloul K, Hamza N, Hdiji O, Mhiri C, Trigui A. Optical coherence tomography in multiple sclerosis: A Tunisian tertiary center study. J Fr Ophtalmol 2025; 48:104371. [PMID: 39662309 DOI: 10.1016/j.jfo.2024.104371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 07/12/2024] [Accepted: 09/17/2024] [Indexed: 12/13/2024]
Abstract
PURPOSE To study retinal layers on OCT in patients with multiple sclerosis (MS) and look for correlations with clinical and electrophysiological characteristics. METHODS We conducted a cross-sectional study including MS patients aged between 18 and 60 years and a reference group of healthy, age- and gender-matched, control participants. A neurological examination with assessment of disability by the Expanded Disability Status Scale (EDSS), an ophthalmological examination, a spectral-domain OCT, and visual evoked potentials (VEP) were performed. RESULTS Fifty-one patients with MS and 30 control subjects were included in the study. The mean age of our patients was 38 years, and the sex ratio (male/female) was 0.49. Mean total thickness of the peripapillary retinal nerve fiber layer (pRNFL) and mean thicknesses in the individual quadrants were significantly lower than those of control subjects (P<0.001). All mean thicknesses of the various retinal layers were reduced compared to those of control eyes, but the difference was statistically significant only for the inner plexiform layer (IPL), the inner nuclear layer (INL) and the outer plexiform layer (OPL). We found a significant relationship between pRNFL atrophy as well as ganglion cell inner plexiform layer (GCIPL) atrophy and history of MS-ON (multiple sclerosis-optic neuritis) (P<0.001). pRNFL was preserved in the primary progressive form of MS, while it was atrophied in relapsing-remitting and secondary progressive forms. There was no significant change in inner retinal layer thicknesses according to duration of MS progression. We found a significant correlation between pRNFL atrophy in the superior (R=-0.22, P=0.03), inferior (R=-0.28, P=0.005) and temporal (R=-0.21, P=0.03) quadrants and the EDSS score. The difference in the thickness of the other retinal layers was significant for the GCIPL in patients with a high EDSS score (>3). There was no significant difference in the thickness of the various retinal layers between eyes of patients on first- or a second-line treatment. We found a correlation between visual acuity and pRNFL (R=0.446, P<0.001) and GCIPL thickness (R=0.343, P=0.001). There was a correlation between the increase of P100 wave latency in VEP and pRNFL atrophy (R=-0.32, P=0.01). A correlation between pRNFL atrophy and the decrease in the amplitude of the P100 wave was only seen in MS-ON eyes (R=0.41, P=0.03). CONCLUSIONS Correlations between pRNFL and GCIPL atrophy and clinical and electrophysiological parameters of MS suggest that OCT is an important tool to quantify neurodegeneration and to monitor disease progression in MS patients.
Collapse
Affiliation(s)
- I Zone-Abid
- Department of Ophtalmology, Habib Bourguiba University Hospital, Faculty of Medicine of Sfax, University of Sfax, Sfax, Tunisia
| | - K Maaloul
- Department of Ophtalmology, Habib Bourguiba University Hospital, Faculty of Medicine of Sfax, University of Sfax, Sfax, Tunisia.
| | - N Hamza
- Department of Neurology, Habib Bourguiba University Hospital, Faculty of Medicine of Sfax, University of Sfax, Sfax, Tunisia
| | - O Hdiji
- Department of Neurology, Habib Bourguiba University Hospital, Faculty of Medicine of Sfax, University of Sfax, Sfax, Tunisia
| | - C Mhiri
- Department of Neurology, Habib Bourguiba University Hospital, Faculty of Medicine of Sfax, University of Sfax, Sfax, Tunisia
| | - A Trigui
- Department of Ophtalmology, Habib Bourguiba University Hospital, Faculty of Medicine of Sfax, University of Sfax, Sfax, Tunisia
| |
Collapse
|
10
|
Yang Z, Shi L, Wang Y, Zhou D, Zhang C, Lin Y. Unveiling the Potential of Tetrahedral DNA Frameworks in Clinical Medicine: Mechanisms, Advances, and Future Perspectives. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2410162. [PMID: 39707665 DOI: 10.1002/smll.202410162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/24/2024] [Indexed: 12/23/2024]
Abstract
As deoxyribonucleic acis (DNA) nanotechnology advances, DNA, a fundamental biological macromolecule, has been employed to treat various clinical diseases. Among the advancements in this field, tetrahedral frameworks nucleic acids (tFNAs) have gained significant attention due to their straightforward design, structural simplicity, low cost, and high yield since their introduction by Turberfield in the early 2000s. Due to its stable spatial structure, tFNAs can resist the impact of innate immune responses on DNA and nuclease activity. Meanwhile, structural programmability of tFNAs allows for the development of static tFNA-based nanomaterials through the engineering of functional oligonucleotides or therapeutic molecules and dynamic tFNAs through the attachment of stimuli-responsive DNA apparatuses. This review first summarizes the key merits of tFNAs, including natural biocompatibility, biodegradability, structural stability, unparalleled programmability, functional diversity, and efficient cellular internalization. Based on these strengths, this review comprehensively analyzes applications of tFNAs in different clinical settings, including orthopedics, stomatology, urinary system diseases, liver-related diseases, tumors, infection, neural system diseases, ophthalmic diseases, and immunoprophylaxis. We also discuss the limitations of tFNAs and the challenges encountered in preclinical studies. This review provides new perspectives for future research and valuable guidance for researchers working in this field.
Collapse
Affiliation(s)
- Zhengyang Yang
- Department of General Surgery, State Key Lab of Digestive Health, National Clinical Research Center for Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Lin Shi
- Department of Orthopedics, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Yun Wang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Dongfang Zhou
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Chao Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
- Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, 610041, China
| |
Collapse
|
11
|
Dagnew B, Honan CA, Laslett LL, Taylor BV, Campbell J, Blizzard L, van der Mei I. Impact of sleep quality on health-related quality of life domains and the mediating effects of symptoms in people with multiple sclerosis. Qual Life Res 2025; 34:563-575. [PMID: 39537977 DOI: 10.1007/s11136-024-03836-4] [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] [Accepted: 11/02/2024] [Indexed: 11/16/2024]
Abstract
PURPOSE Many people living with multiple sclerosis (PwMS) experience poor sleep, which is associated with diminished overall health-related quality of life (HRQoL). We quantified associations between sleep quality and HRQoL domains and examined the extent to which other MS symptoms could account for these associations. METHODS In this cross-sectional survey of 1,717 Australians with MS, we used Assessment of Quality of Life (AQoL) 8D and Pittsburgh Sleep Quality Index (PSQI) to assess HRQoL and sleep quality, respectively. Total, direct, and indirect effects of sleep quality on HRQoL domains were determined using mediation regression analysis. RESULTS Poor sleep quality was significantly associated with all domains of HRQoL, with strongest associations seen for mental health (β=-0.08) and pain (β=-0.11), and weaker associations for independent living (β=-0.05) and senses (β=-0.03). Poor sleep quality had the largest direct effect on mental health (60.8%), happiness (48.7%), and pain (49.7%). MS symptom clusters mostly contributing to indirect effects were "feelings of anxiety and depression" for psychosocial, and "pain and sensory symptoms" for physical HRQoL super dimensions. CONCLUSION Improving sleep could lead to substantial improvements in all HRQoL domains and the improvement in HRQoL could be partially achieved through indirect improvements in sleep on MS symptoms.
Collapse
Affiliation(s)
- Baye Dagnew
- Menzies Institute for Medical Research, University of Tasmania, Private Bag 23, Hobart, TAS, 7001, Australia
- College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Cynthia A Honan
- School of Psychological Sciences, University of Tasmania, Launceston, Australia
| | - Laura L Laslett
- Menzies Institute for Medical Research, University of Tasmania, Private Bag 23, Hobart, TAS, 7001, Australia
| | - Bruce V Taylor
- Menzies Institute for Medical Research, University of Tasmania, Private Bag 23, Hobart, TAS, 7001, Australia
| | - Julie Campbell
- Menzies Institute for Medical Research, University of Tasmania, Private Bag 23, Hobart, TAS, 7001, Australia
| | - Leigh Blizzard
- Menzies Institute for Medical Research, University of Tasmania, Private Bag 23, Hobart, TAS, 7001, Australia
| | - Ingrid van der Mei
- Menzies Institute for Medical Research, University of Tasmania, Private Bag 23, Hobart, TAS, 7001, Australia.
| |
Collapse
|
12
|
Ibrahim DR, Schwarz K, Suiwal S, Maragkou S, Schmitz F. Early Synapse-Specific Alterations of Photoreceptor Mitochondria in the EAE Mouse Model of Multiple Sclerosis. Cells 2025; 14:206. [PMID: 39936997 PMCID: PMC11816939 DOI: 10.3390/cells14030206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/26/2025] [Accepted: 01/28/2025] [Indexed: 02/13/2025] Open
Abstract
Multiple sclerosis (MS) is an inflammatory autoimmune disease of the central nervous system (CNS) linked to many neurological disabilities. The visual system is frequently impaired in MS. In previous studies, we observed early malfunctions of rod photoreceptor ribbon synapses in the EAE mouse model of MS that included alterations in synaptic vesicle cycling and disturbances of presynaptic Ca2+ homeostasis. Since these presynaptic events are highly energy-demanding, we analyzed whether synaptic mitochondria, which play a major role in synaptic energy metabolism, might be involved at that early stage. Rod photoreceptor presynaptic terminals contain a single large mitochondrion next to the synaptic ribbon. In the present study, we analyzed the expression of functionally relevant mitochondrial proteins (MIC60, ATP5B, COX1, PINK1, DRP1) by high-resolution qualitative and quantitative immunofluorescence microscopy, immunogold electron microscopy and quantitative Western blot experiments. We observed a decreased expression of many functionally relevant proteins in the synaptic mitochondria of EAE photoreceptors at an early stage, suggesting that early mitochondrial dysfunctions play an important role in the early synapse pathology. Interestingly, mitochondria in presynaptic photoreceptor terminals were strongly compromised in early EAE, whereas extra-synaptic mitochondria in photoreceptor inner segments remained unchanged, demonstrating a functional heterogeneity of photoreceptor mitochondria.
Collapse
Affiliation(s)
- Dalia R. Ibrahim
- Institute of Anatomy, Department of Neuroanatomy, Medical School Homburg, Saarland University, 66421 Homburg, Germany; (K.S.); (S.S.); (S.M.)
| | | | | | | | - Frank Schmitz
- Institute of Anatomy, Department of Neuroanatomy, Medical School Homburg, Saarland University, 66421 Homburg, Germany; (K.S.); (S.S.); (S.M.)
| |
Collapse
|
13
|
Mohapatra A, Amarjeet, Pancholi B, Babu R, Abate SK, Garabadu D. Lysophosphatidylcholine induces ceramide synthase-2 expression in primary culture model of astrocytopathy: potential therapeutic target in multiple sclerosis. Mol Biol Rep 2025; 52:166. [PMID: 39870973 DOI: 10.1007/s11033-025-10252-5] [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: 10/07/2024] [Accepted: 01/13/2025] [Indexed: 01/30/2025]
Abstract
BACKGROUND Multiple sclerosis (MS) is a chronic autoimmune condition that damages the myelin sheath of neurons in the central nervous system, resulting in compromised nerve transmission and motor impairment. The astrocytopathy is considered one of the prominent etiological factor in the pathophysiology of demyelination in MS. The expression level of ceramide synthase-2 (CS-2) is yet to be established in the pathophysiology of astrocytopathy although the derailed ceramide biosynthetic pathways is well demonstrated in the pathophysiology of demyelination. Therefore, in the present study, the expression level of CS-2 has been evaluated in lysophosphatidylcholine (LPC)-challenged primary astrocytes in the presence of anti-demyelinating agents such as Fingolimod, a clinically approved anti-demyelinating drug, and Ursolic Acid (UA), an experimentally accepted anti-demyelinating agent, in MS. METHODS AND RESULTS LPC (150 µg/ml) caused astrocytopathy evident by decrease in percentage of viable astrocytes, increase in percentage of apoptotic astrocytes, increase in the level of reactive oxygen species (ROS), and increase in the level of activated astrocytes. Interestingly, LPC significantly increased the expression level of CS-2 in the primary culture of astrocytes where as fingolimod and UA (1, 10, and 100 µM) were able to attenuate the extent of LPC-induced astrocytopathy in the primary culture model of MS. Further, both the drugs drastically reduced the LPC-induced increase in the level of expression of CS-2 in the astrocytes. CONCLUSIONS These observations indicate the fact that CS-2 could be a potential target in the management of astrocytopathy and astrocytopathy-related neurological disorders including MS. In conclusion, CS-2-targeted drugs could be potential therapeutic options in the management of MS.
Collapse
Affiliation(s)
- Abhipsa Mohapatra
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, 151401, India
| | - Amarjeet
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, 151401, India
| | - Bhaskaranand Pancholi
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, 151401, India
| | - Raja Babu
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, 151401, India
| | - Selamu Kebamo Abate
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, 151401, India
| | - Debapriya Garabadu
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, 151401, India.
| |
Collapse
|
14
|
Clarke JPWE, Messmer ML, Pilon J, Reding J, Thibault PA, Salapa HE, Levin MC. Dysfunctional RNA binding protein induced neurodegeneration is attenuated by inhibition of the integrated stress response. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167562. [PMID: 39521193 DOI: 10.1016/j.bbadis.2024.167562] [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: 10/14/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
Dysfunction of the RNA binding protein heterogeneous nuclear ribonucleoprotein A1 (hnRNP A1) contributes to neurodegeneration, the primary cause of permanent disability in multiple sclerosis (MS). To better understand the role of hnRNP A1 dysfunction in the pathogenesis of neurodegeneration, we utilized optogenetics-driven hnRNP A1 clustering to model its dysfunction in neuron-like differentiated Neuro-2A cells. hnRNP A1 clustering activates the integrated stress response (ISR) and results in a neurodegenerative phenotype marked by decreased neuronal protein translation and neurite loss. Small molecule inhibition of the ISR with either PERKi (GSK2606414) or ISRIB (integrated stress response inhibitor) attenuated both the decrease in neuronal translation and neurite loss, without affecting hnRNP A1 clustering. We then confirmed a strong association between hnRNP A1 clustering and ISR activation in neurons from MS brains. These data illustrate that hnRNP A1 dysfunction promotes neurodegeneration by activation of the ISR in vitro and in vivo, thus revealing a novel therapeutic target to reduce neurodegeneration and subsequent disability in MS.
Collapse
Affiliation(s)
- Joseph-Patrick W E Clarke
- Office of the Saskatchewan Multiple Sclerosis Clinical Research Chair, University of Saskatchewan, Saskatoon, SK S7K-0M7, Canada; Cameco MS Neuroscience Research Centre, College of Medicine, University of Saskatchewan, Saskatoon, SK S7K 0M7, Canada; Department of Medicine, Neurology Division, University of Saskatchewan, Saskatoon, SK S7N-5E5, Canada.
| | - Miranda L Messmer
- Office of the Saskatchewan Multiple Sclerosis Clinical Research Chair, University of Saskatchewan, Saskatoon, SK S7K-0M7, Canada; Cameco MS Neuroscience Research Centre, College of Medicine, University of Saskatchewan, Saskatoon, SK S7K 0M7, Canada; Department of Anatomy, Physiology and Pharmacology, University of Saskatchewan, Saskatoon, SK S7N-5E5, Canada
| | - Jacob Pilon
- Office of the Saskatchewan Multiple Sclerosis Clinical Research Chair, University of Saskatchewan, Saskatoon, SK S7K-0M7, Canada; Cameco MS Neuroscience Research Centre, College of Medicine, University of Saskatchewan, Saskatoon, SK S7K 0M7, Canada; Department of Health Sciences, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N-5E5, Canada
| | - Jenna Reding
- Office of the Saskatchewan Multiple Sclerosis Clinical Research Chair, University of Saskatchewan, Saskatoon, SK S7K-0M7, Canada; Cameco MS Neuroscience Research Centre, College of Medicine, University of Saskatchewan, Saskatoon, SK S7K 0M7, Canada; Department of Anatomy, Physiology and Pharmacology, University of Saskatchewan, Saskatoon, SK S7N-5E5, Canada
| | - Patricia A Thibault
- Office of the Saskatchewan Multiple Sclerosis Clinical Research Chair, University of Saskatchewan, Saskatoon, SK S7K-0M7, Canada; Cameco MS Neuroscience Research Centre, College of Medicine, University of Saskatchewan, Saskatoon, SK S7K 0M7, Canada; Department of Medicine, Neurology Division, University of Saskatchewan, Saskatoon, SK S7N-5E5, Canada
| | - Hannah E Salapa
- Office of the Saskatchewan Multiple Sclerosis Clinical Research Chair, University of Saskatchewan, Saskatoon, SK S7K-0M7, Canada; Cameco MS Neuroscience Research Centre, College of Medicine, University of Saskatchewan, Saskatoon, SK S7K 0M7, Canada; Department of Medicine, Neurology Division, University of Saskatchewan, Saskatoon, SK S7N-5E5, Canada
| | - Michael C Levin
- Office of the Saskatchewan Multiple Sclerosis Clinical Research Chair, University of Saskatchewan, Saskatoon, SK S7K-0M7, Canada; Cameco MS Neuroscience Research Centre, College of Medicine, University of Saskatchewan, Saskatoon, SK S7K 0M7, Canada; Department of Medicine, Neurology Division, University of Saskatchewan, Saskatoon, SK S7N-5E5, Canada; Department of Health Sciences, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N-5E5, Canada; Department of Anatomy, Physiology and Pharmacology, University of Saskatchewan, Saskatoon, SK S7N-5E5, Canada.
| |
Collapse
|
15
|
Li Y, Sun J, Zhuo Z, Guo M, Duan Y, Xu X, Tian D, Li K, Zhou F, Li H, Zhang N, Han X, Shi F, Li Y, Zhang X, Liu Y. Imaging Transcriptomics of Brain Functional Alterations in MS and Neuromyelitis Optica Spectrum Disorder. AJNR Am J Neuroradiol 2024; 45:1901-1909. [PMID: 39510804 PMCID: PMC11630882 DOI: 10.3174/ajnr.a8480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 07/17/2024] [Indexed: 11/15/2024]
Abstract
BACKGROUND AND PURPOSE The underlying transcriptomic signatures driving brain functional alterations in MS and neuromyelitis optica spectrum disorder (NMOSD) are still unclear. MATERIALS AND METHODS Regional fractional amplitude of low-frequency fluctuation (fALFF) values were obtained and compared among 209 patients with MS, 90 patients with antiaquaporin-4 antibody (AQP4)+ NMOSD, 49 with AQP4- NMOSD, and 228 healthy controls from a discovery cohort. We used partial least squares (PLS) regression to identify the gene transcriptomic signatures associated with disease-related fALFF alterations. The biologic process and cell type-specific signature of the identified PLS genes were explored by enrichment analysis. The correlation between PLS genes and clinical variables was explored. A prospective independent cohort was used to validate the brain fALFF alterations and the repeatability of identified genes. RESULTS MS, AQP4+ NMOSD, and AQP4- NMOSD showed decreased fALFF in cognition-related regions and deep gray matter, while NMOSD (both AQP4+ and AQP4-) additionally demonstrated lower fALFF in the visual region. The overlapping PLS1- genes (indicating that the genes were overexpressed as regional fALFF decreased) were enriched in response to regulation of the immune response in all diseases, and the PLS1- genes were specifically enriched in the epigenetics profile in MS, membrane disruption and cell adhesion in AQP4+ NMOSD, and leukocyte activation in AQP4- NMOSD. For the cell type transcriptional signature, microglia and astrocytes accounted for the decreased fALFF. The fALFF-associated PLS1- genes directly correlated with Expanded Disability Status Scale of MS and disease duration across disorders. CONCLUSIONS We revealed the functional activity alterations and their underlying shared and specific gene transcriptional signatures in MS, AQP4+ NMOSD, and AQP4- NMOSD.
Collapse
Affiliation(s)
- Yuna Li
- From the Department of Radiology (Yuna Li, J.S., Z.Z., M.G., Y.D., X.X., Y. Liu), Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jun Sun
- From the Department of Radiology (Yuna Li, J.S., Z.Z., M.G., Y.D., X.X., Y. Liu), Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhizheng Zhuo
- From the Department of Radiology (Yuna Li, J.S., Z.Z., M.G., Y.D., X.X., Y. Liu), Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Min Guo
- From the Department of Radiology (Yuna Li, J.S., Z.Z., M.G., Y.D., X.X., Y. Liu), Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yunyun Duan
- From the Department of Radiology (Yuna Li, J.S., Z.Z., M.G., Y.D., X.X., Y. Liu), Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xiaolu Xu
- From the Department of Radiology (Yuna Li, J.S., Z.Z., M.G., Y.D., X.X., Y. Liu), Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Decai Tian
- Center for Neurology (D.T., X.Z.), Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Kuncheng Li
- Department of Radiology (K.L.), Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Fuqing Zhou
- Department of Radiology (F.Z.), The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi Province, China
| | - Haiqing Li
- Department of Radiology (H.L.), Huashan Hospital, Fudan University, Shanghai, China
| | - Ningnannan Zhang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging (N.Z.), Tianjin Medical University General Hospital, Tianjin, China
| | - Xuemei Han
- Department of Neurology (X.H.), China-Japan Union Hospital of Jilin University, Changchun, China
| | - Fudong Shi
- China National Clinical Research Center for Neurological Diseases (F.S.), Beijing, China
- Department of Neurology (F.S.), Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Yongmei Li
- Department of Radiology (Yongmei Li), The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xinghu Zhang
- Center for Neurology (D.T., X.Z.), Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yaou Liu
- From the Department of Radiology (Yuna Li, J.S., Z.Z., M.G., Y.D., X.X., Y. Liu), Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
16
|
Jalilian M, Elhaie M, Sharifi M, Abedi I. Assessment of axonal injury in multiple sclerosis: combined analysis of serum light-chain neurofilaments and diffusion tensor imaging. BMJ Neurol Open 2024; 6:e000788. [PMID: 39649079 PMCID: PMC11624819 DOI: 10.1136/bmjno-2024-000788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 11/18/2024] [Indexed: 12/10/2024] Open
Abstract
Background Multiple sclerosis (MS) is a chronic neuroinflammatory condition characterised by demyelination and axonal damage in the central nervous system. Diffusion tensor imaging (DTI) enables non-invasive investigation of microstructural white matter alterations, while serum neurofilament light chain (NFL) holds promise as a fluid biomarker of axonal injury. Objectives To use DTI and serum NFL measurements to evaluate white matter pathology in patients with MS and explore the relationship between in vivo imaging and biochemical indicators of axonal damage. Methods 41 patients with relapse-remitting MS and 41 age-matched healthy controls underwent brain MRI including DTI acquisition. Serum samples were analysed for NFL concentrations using ELISA. Region of interest analysis was conducted to derive DTI metrics including fractional anisotropy, mean diffusivity, axial diffusivity and radial diffusivity. Correlational analyses were used to explore the associations between the imaging and biochemical indices. Results Patients exhibited significantly elevated serum NFL levels and altered DTI metrics compared with controls, indicative of axonal/myelin pathology. DTI parameters were positively correlated with serum NFL concentration (p value<0.0001). Visual analogue scale scores demonstrated a significant positive relationship between DTI metrics and NFL, validating their potential as radiological and fluid-based markers of symptom severity. Conclusions Combined DTI and serum NFL measurements may enhance the evaluation of axonal injury in MS by providing complementary in vivo and biochemical perspectives. The corresponding changes observed between the modalities support their utility as non-invasive biomarkers reflecting pathophysiological processes and clinical status in MS. Larger validation cohorts are needed to determine the clinical applicability.
Collapse
Affiliation(s)
- Milad Jalilian
- Department of Neuroscience and Addiction Studies, Tehran University of Medical Sciences, Tehran, Iran (the Islamic Republic of)
| | - Mohammadreza Elhaie
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran (the Islamic Republic of)
| | - Mohammadreza Sharifi
- Department of Genetics and Molecular Biology, Isfahan University of Medical Sciences, Isfahan, Iran (the Islamic Republic of)
| | - Iraj Abedi
- Department of Medical Physics, Isfahan University of Medical Sciences, Isfahan, Iran (the Islamic Republic of)
| |
Collapse
|
17
|
Panda P, Mohapatra R. Herbal nanoparticles: a targeted approach for neurodegenerative disorder treatment. J Drug Target 2024; 32:1233-1246. [PMID: 39133517 DOI: 10.1080/1061186x.2024.2391913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 08/13/2024]
Abstract
Nanotechnology has significantly impacted human life, particularly in overcoming the limitations associated with neurodegenerative diseases (NDs). Various nanostructures and vehicle systems, such as polymer nanoparticles, carbon nanotubes (CNTs), nanoliposomes, nano-micelles, lipid nanoparticles, lactoferrin, polybutylcyanoacrylate, and poly lactic-co-glycolic acid, have been shown to enhance drug efficacy, reduce side effects, and improve pharmacokinetics. NDs affect millions worldwide and are challenging to treat due to the blood-brain barrier (BBB), which hinders drug delivery to the central nervous system (CNS). Research suggests that natural ingredients can be formulated into nanoparticles, offering a promising approach for ND treatment. This review examines the advantages and disadvantages of herbal-based nanoformulations, highlighting their potential effectiveness when used alone or in combination with other medications. Herbal nanoparticles provide benefits over synthetic ones due to their biocompatibility, reduced toxicity, and potential for synergistic effects. The study's findings can be applied to develop more efficient drug delivery systems, improving the treatment of NDs by enhancing drug penetration across the BBB and targeting affected CNS areas more precisely.
Collapse
Affiliation(s)
- Pratikeswar Panda
- Department of Pharmaceutics, School of pharmaceutical science, Siksha 'O' Anusandhan University, Bhubaneswar, Odisha, India
| | - Rajaram Mohapatra
- Department of Pharmaceutics, School of pharmaceutical science, Siksha 'O' Anusandhan University, Bhubaneswar, Odisha, India
| |
Collapse
|
18
|
Yu XH, Lu HM, Li J, Su MZ, Li XM, Jin Y. Association between 25(OH) vitamin D and multiple sclerosis: cohort, shared genetics, and Causality. Nutr J 2024; 23:151. [PMID: 39616386 PMCID: PMC11608472 DOI: 10.1186/s12937-024-01059-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 11/26/2024] [Indexed: 12/06/2024] Open
Abstract
BACKGROUND Multiple Sclerosis (MS), an autoimmune disorder causing demyelination and neurological damage, has been linked to 25-hydroxyvitamin D (25OHD) levels, suggesting its role in immune response and MS onset. This study used GWAS datasets to investigate genetic associations between 25OHD and MS. METHODS We utilized a large-scale prospective cohort to evaluate serum 25OHD levels and MS risk. Linkage Disequilibrium Score Regression (LDSC) assessed genetic correlations between 25OHD levels and MS. Cross-trait genome-wide pleiotropy analysis revealed shared genetic loci. MAGMA analysis identified pleiotropic genes, enriched tissues, and gene sets. Stratified LDSC estimated tissue-specific and cell-specific heritability enrichment, and multi-trait co-localization analysis identified shared immune cell subsets. Bidirectional Mendelian Randomization (MR) assessed the causal association between 25OHD and MS risk. RESULTS The observational study found a nonlinear relationship between 25OHD levels and MS risk, with the lowest quartile showing significant risk elevation. Our findings revealed shared genetic structure between 25OHD levels and MS, suggesting a common biological pathway involving immune function and CNS integrity. We found 24 independent loci shared between 25OHD levels and MS risk, enriched in brain tissues and involved in pathways like LDL, HDL, and TG metabolism. Four loci (6p24.3, 6p22.2, 12q14.1, and 19p13.2) had strong co-localization evidence, with mapped genes as potential drug targets. Bidirectional MR analysis supported a causal effect of 25OHD levels on MS risk, suggesting 25OHD supplementation could modulate MS risk. CONCLUSION This study reveals the complex relationship between 25OHD levels and MS, indicating that higher levels are not always advantageous and recommending moderation in supplementation. We identified SMARCA4 as a potential therapeutic target and detailed key pathways influencing this interaction.
Collapse
Affiliation(s)
- Xing-Hao Yu
- The Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, 213000, China
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215123, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, 215123, China
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Hui-Min Lu
- Department of Outpatient and Emergency, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Jun Li
- The Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, 213000, China
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
- Department of Pharmacy, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu, 213000, China
| | - Ming-Zhu Su
- The Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, 213000, China
- Department of Good Clinical Practice, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu, 213000, China
| | - Xiao-Min Li
- The Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, 213000, China.
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China.
- Department of Pharmacy, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu, 213000, China.
| | - Yi Jin
- The Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, 213000, China.
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China.
- Department of Pharmacy, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu, 213000, China.
| |
Collapse
|
19
|
Perdaens O, van Pesch V. Should We Consider Neurodegeneration by Itself or in a Triangulation with Neuroinflammation and Demyelination? The Example of Multiple Sclerosis and Beyond. Int J Mol Sci 2024; 25:12637. [PMID: 39684351 PMCID: PMC11641818 DOI: 10.3390/ijms252312637] [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: 10/31/2024] [Revised: 11/20/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Neurodegeneration is preeminent in many neurological diseases, and still a major burden we fail to manage in patient's care. Its pathogenesis is complicated, intricate, and far from being completely understood. Taking multiple sclerosis as an example, we propose that neurodegeneration is neither a cause nor a consequence by itself. Mitochondrial dysfunction, leading to energy deficiency and ion imbalance, plays a key role in neurodegeneration, and is partly caused by the oxidative stress generated by microglia and astrocytes. Nodal and paranodal disruption, with or without myelin alteration, is further involved. Myelin loss exposes the axons directly to the inflammatory and oxidative environment. Moreover, oligodendrocytes provide a singular metabolic and trophic support to axons, but do not emerge unscathed from the pathological events, by primary myelin defects and cell apoptosis or secondary to neuroinflammation or axonal damage. Hereby, trophic failure might be an overlooked contributor to neurodegeneration. Thus, a complex interplay between neuroinflammation, demyelination, and neurodegeneration, wherein each is primarily and secondarily involved, might offer a more comprehensive understanding of the pathogenesis and help establishing novel therapeutic strategies for many neurological diseases and beyond.
Collapse
Affiliation(s)
- Océane Perdaens
- Neurochemistry Group, Institute of NeuroScience, Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium;
| | - Vincent van Pesch
- Neurochemistry Group, Institute of NeuroScience, Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium;
- Department of Neurology, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| |
Collapse
|
20
|
Sandgren S, Novakova L, Nordin A, Sabir H, Axelsson M, Malmeström C, Zetterberg H, Lycke J. The effect of alemtuzumab on neurodegeneration in relapsing-remitting multiple sclerosis: A five-year prospective mono-center study. Mult Scler Relat Disord 2024; 91:105894. [PMID: 39293124 DOI: 10.1016/j.msard.2024.105894] [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: 06/29/2024] [Revised: 08/31/2024] [Accepted: 09/12/2024] [Indexed: 09/20/2024]
Abstract
BACKGROUND Relapsing-remitting multiple sclerosis (RRMS) is an inflammatory and neurodegenerative disease. After two or more short courses of alemtuzumab (ALZ), an immune reconstitution is achieved, which long-term results in reduced disease activity. We aimed to investigate the effect of ALZ on measures of neurodegeneration (i.e., brain atrophy, and retinal layer thinning). METHODS We designed an observational prospective mono-center study in RRMS patients initiating ALZ treatment. Patients were assessed at baseline (month 0) and thereafter annually for five years with clinical measures, synthetic magnetic resonance imaging (SyMRI) and optical coherence tomography (OCT), with a re-baseline SyMRI scan and an OCT exam 24 months after initiating ALZ. Persons with neurological symptoms but without evidence of neurological disease served as symptomatic controls (SCs, n = 27). RESULTS Forty-nine RRMS patients were included. Baseline median expanded disability status scale [2.0 (IQR 1.5)] was unchanged during follow-up, 71 % were progression-free, 33 % achieved no evidence of disease activity-3 (NEDA-3). Between baseline and month 60, SyMRI showed a reduction of brain parenchymal fraction (BPF) and grey matter (GM) volume in patients. The BPF reduction was greater in RRMS patients than in SCs (p < 0.05), and more pronounced in patients with high pre-baseline disease activity than in those without (p < 0.01). OCT showed significant thinning of macular ganglion cell and inner plexiform layers (mGCIPL) and in peripapillary retinal nerve fiber layer (pRNFL) in patients. In contrast, absolute values of white matter (WM) volume and myelin content (MyC) quantified by SyMRI, were stable or increased after re-baseline (month 24) and up to month 60, and this increase appeared limited to patients without high pre-baseline disease activity and to patients with NEDA-3 or disability worsening during follow-up. A strong positive correlation between WM volume and GM volume at baseline was lost after ALZ intervention for their delta values, i.e., change from re-baseline (month 24) to month 60. While the positive baseline correlation between WM volume and MyC increased for their delta values, the positive baseline correlation between GM volume and MyC changed to negative for their delta values. CONCLUSION We showed that neurodegeneration continued in RRMS patients under ALZ treatment, but it appeared to be limited to BPF and GM, and more pronounced in patients with disease activity. Our data suggest that patients who respond to ALZ treatment show signs of remyelination. OCT and SyMRI have potential to quantify measures of neurodegeneration that is affected by treatment intervention in RRMS.
Collapse
Affiliation(s)
- Sofia Sandgren
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Sahlgrenska, Department of Neurology, Region Västra Götaland, SE-413 45 Gothenburg, Sweden.
| | - Lenka Novakova
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Sahlgrenska, Department of Neurology, Region Västra Götaland, SE-413 45 Gothenburg, Sweden.
| | - Anna Nordin
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Sahlgrenska, Department of Neurology, Region Västra Götaland, SE-413 45 Gothenburg, Sweden.
| | - Hemin Sabir
- Department of Neurology and Ophthalmology outpatient clinics, Hallands Hospital Kungsbacka, SE-434 80 Kungsbacka, Sweden.
| | - Markus Axelsson
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Sahlgrenska, Department of Neurology, Region Västra Götaland, SE-413 45 Gothenburg, Sweden.
| | - Clas Malmeström
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Sahlgrenska, Department of Neurology, Region Västra Götaland, SE-413 45 Gothenburg, Sweden; Laboratory for Clinical Immunology, Sahlgrenska University Hospital, SE-413 46 Gothenburg, Sweden.
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology at Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, SE-431 80 Mölndal, Sweden; Department of Neurodegenerative Disease, University College London (UCL) Queen Square Institute of Neurology, London, United Kingdom; United Kingdom (UK) Dementia Research Institute at University College London (UCL), London, United Kingdom; Hong Kong Center for Neurodegenerative Diseases, Hong Kong, Hong Kong, China; Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States.
| | - Jan Lycke
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Sahlgrenska, Department of Neurology, Region Västra Götaland, SE-413 45 Gothenburg, Sweden.
| |
Collapse
|
21
|
Saridas F, Hojjati F, Alizada S, Lazrak S, Koc ER, Turan OF. Effects of disease-modifying therapies on remyelination in multiple sclerosis; evaluation via visual evoked potential test. Mult Scler Relat Disord 2024; 91:105850. [PMID: 39232395 DOI: 10.1016/j.msard.2024.105850] [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: 06/05/2024] [Revised: 08/12/2024] [Accepted: 08/26/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND Assessment of the visual pathway, which is frequently affected by MS, provides the opportunity to measure the remyelination of acute and chronic MS lesions in vivo and non-invasively. VEP can be used in this context. Amplitude is a parameter of axonal loss, whereas latency is an in vivo biomarker of myelin repair. This study aimed to evaluate DMT's neuroprotective and pro-remyelinating potential by evaluating VEP latency and amplitude in MS patients. MATERIALS AND METHODS A total of 74 patients with relapsing MS who had no evidence of optic neuritis were included in the study. Patient data were retrospectively analyzed and recorded. In the VEP test, latency above 118 ms and amplitude below 5.0 μV were considered abnormal. Classified according to DMTs (injectables, teriflunomide, dimethyl fumarate, fingolimod, cladribine, and alemtuzumab). Visual evoked potential tests, clinical features, and cerebrospinal fluid examinations were evaluated by three independent neurologists (one of whom was also a clinical neurophysiologist). RESULTS The mean age at diagnosis was 29.2 ± 9.01, and the mean age at first VEP was 34.97 ± 10.64. In women, latency was lower, and amplitude was higher. The mean differences in latency and amplitude were, respectively, latency prolonged by 0.7 ms on the right and 0.5 ms on the left, and amplitude increased by 0.6 μV on the right and 0.37 μV on the left. However, these changes were not statistically significant. Latency worsening was more prominent in those with longer disease duration (p = 0.011). Those with amplitude or latency worsening had higher EDSS (p = 0.016 and 0.013, respectively). DMTs did not affect these changes. CONCLUSION Prolonged latency is associated with a long disease duration. Deterioration in both amplitude and latency is evident in high EDSS. These results may be an indirect consequence of axonal degeneration dominating remyelination. DMTs do not ameliorate impaired remyelination and neurodegeneration but seem to be sufficient for short-term maintenance of the current state.
Collapse
Affiliation(s)
- Furkan Saridas
- Bursa Uludağ University Medicine Faculty, Department of Neurology, Türkiye.
| | - Farid Hojjati
- Bursa Uludağ University Medicine Faculty, Department of Neurology, Türkiye.
| | - Shanay Alizada
- Bursa Uludağ University Medicine Faculty, Department of Neurology, Türkiye.
| | - Sarra Lazrak
- Bursa Uludağ University Medicine Faculty, Department of Neurology, Türkiye.
| | - Emine Rabia Koc
- Bursa Uludağ University Medicine Faculty, Department of Neurology, Türkiye.
| | - Omer Faruk Turan
- Bursa Uludağ University Medicine Faculty, Department of Neurology, Türkiye.
| |
Collapse
|
22
|
Tisell A, Söderberg K, Link Y, Lundberg P, Mellergård J. Diffuse white matter pathology in multiple sclerosis during treatment with dimethyl fumarate-An observational study of changes in normal-appearing white matter using proton magnetic resonance spectroscopy. PLoS One 2024; 19:e0309547. [PMID: 39432495 PMCID: PMC11493296 DOI: 10.1371/journal.pone.0309547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 08/13/2024] [Indexed: 10/23/2024] Open
Abstract
BACKGROUND Multiple sclerosis (MS) is an inflammatory demyelinating disease with neurodegenerative features causing risk for neurologic irreversible disability over time. Examination of normal-appearing white matter (NAWM) changes in MS by proton magnetic resonance spectroscopy (1H-MRS), may detect diffuse white matter pathology that is associated with neurodegeneration. METHODS In this observational study of in total twenty-six patients with MS, starting treatment with dimethyl fumarate (DMF), we measured the absolute concentration of metabolites in periventricular NAWM using 1H-MRS at baseline and after one and three years of treatment. Metabolite concentrations were analyzed both cross-sectionally, in relation to 10 controls and longitudinally in relation to disease activity. RESULTS Patients with MS had higher concentrations of myo-inositol (mIns) in NAWM at baseline compared with controls (mean 5.98 ± 1.37 (SD) and 4.32 ± 1.16 (SD), p<0.01, independent samples t-test). The disease duration was inversely correlated with concentrations of total N-acetylaspartate and N-acetylaspartylglutamate (tNA) (r = -0.62, p<0.01) in NAWM as well as positively to the ratio of mIns and tNA (r = 0.51, p = 0.03). Metabolite concentrations during one-year (n = 19) and three-years (n = 11) follow-up were generally stable. The dropouts were caused by treatment switch after one year, mainly due to new MRI activity. Cross-sectional analyses showed that there was an inverse correlation between concentrations of tNA and mIns at both baseline and at 1 and 3-years follow-up (r = -0.44 to -0.65, p = 0.04 to 0.004). Metabolite concentrations were stable during 1-year follow-up independently of disease activity. CONCLUSIONS Higher concentrations of the astrogliosis marker mIns in MS compared to controls, the inverse relation between MS disease duration and the neuroaxonal integrity marker tNA, as well as the consistent inverse relation between these two metabolites during follow-up, showed that non-lesional white matter pathology is present in this cohort of MS patients in early disease stages. However, metabolite concentrations during follow-up were generally stable and did not reflect differences in disease activity among patients.
Collapse
Affiliation(s)
- Anders Tisell
- Department of Medical Radiation Physics in Linköping, and Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
- Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
| | - Kristina Söderberg
- Department of Radiology in Linköping, and Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Yumin Link
- Department of Neurology in Linköping, and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Peter Lundberg
- Department of Medical Radiation Physics in Linköping, and Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
- Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
| | - Johan Mellergård
- Department of Neurology in Linköping, and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
23
|
Huang Y, Yang D, Liao S, Guan X, Zhou F, Liu Y, Wang Y, Zhang Y. Ginsenoside Rg1 protects the blood-brain barrier and myelin sheath to prevent postoperative cognitive dysfunction in aged mice. Neuroreport 2024; 35:925-935. [PMID: 39166417 DOI: 10.1097/wnr.0000000000002083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2024]
Abstract
In this study, the postoperative cognitive dysfunction (POCD) mouse model was established to observe the changes in inflammation, blood-brain barrier permeability, and myelin sheath, and we explore the effect of ginsenoside Rg1 pretreatment on improving POCD syndrome. The POCD model of 15- to 18-month-old mice was carried out with internal fixation of tibial fractures under isoflurane anesthesia. Pretreatment was performed by continuous intraperitoneal injection of ginsenoside Rg1(40 mg/kg/day) for 14 days before surgery. The cognitive function was detected by the Morris water maze. The contents of interleukin-1β and tumor necrosis factor-α in the hippocampus, cortex, and serum were detected by ELISA. The permeability of blood-brain barrier was observed by Evans blue. The mRNA levels and protein expression levels of 2',3'-cyclic nucleotide 3'-phosphodiesterase (CNPase), myelin basic protein (MBP), beta-catenin, and cyclin D1 in the hippocampus were analyzed by quantitative PCR and western blotting. The protein expression levels of ZO-1 and Wnt1 in the hippocampus were analyzed by western blotting. Finally, the localizations of CNPase and MBP in the hippocampus were detected by immunofluorescence. Ginsenoside Rg1 can prevent POCD, peripheral and central inflammation, and blood-brain barrier leakage, and reverse the downregulation of ZO-1, CNPase, MBP, and Wnt pathway-related molecules in aged mice. Preclinical studies suggest that ginsenoside Rg1 improves postoperative cognitive function in aged mice by protecting the blood-brain barrier and myelin sheath, and its specific mechanism may be related to the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Yao Huang
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University
- Department of Anesthesiology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University
| | - Dianping Yang
- Department of Anesthesiology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University
| | - Sijing Liao
- Department of Anesthesiology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University
| | - Xilin Guan
- Department of Anesthesiology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University
| | - Feiran Zhou
- Department of Pain, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University
| | - Yan Liu
- Department of Anesthesiology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University
| | - Yong Wang
- Department of Anesthesiology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University
| | - Ying Zhang
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University
- Department of Anesthesiology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University
- Department of Anesthesiology, Heiiang Hospital of Traditional Chinese Medicine, Southwest Medical University, Luzhou, Sichuan Province, China
| |
Collapse
|
24
|
Mahapatra C, Thakkar R, Kumar R. Modulatory Impact of Oxidative Stress on Action Potentials in Pathophysiological States: A Comprehensive Review. Antioxidants (Basel) 2024; 13:1172. [PMID: 39456426 PMCID: PMC11504047 DOI: 10.3390/antiox13101172] [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: 08/06/2024] [Revised: 09/12/2024] [Accepted: 09/20/2024] [Indexed: 10/28/2024] Open
Abstract
Oxidative stress, characterized by an imbalance between the production of reactive oxygen species (ROS) and the body's antioxidant defenses, significantly affects cellular function and viability. It plays a pivotal role in modulating membrane potentials, particularly action potentials (APs), essential for properly functioning excitable cells such as neurons, smooth muscles, pancreatic beta cells, and myocytes. The interaction between oxidative stress and AP dynamics is crucial for understanding the pathophysiology of various conditions, including neurodegenerative diseases, cardiac arrhythmias, and ischemia-reperfusion injuries. This review explores how oxidative stress influences APs, focusing on alterations in ion channel biophysics, gap junction, calcium dynamics, mitochondria, and Interstitial Cells of Cajal functions. By integrating current research, we aim to elucidate how oxidative stress contributes to disease progression and discuss potential therapeutic interventions targeting this interaction.
Collapse
Affiliation(s)
- Chitaranjan Mahapatra
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA 94158, USA
| | - Ravindra Thakkar
- California Institute for Quantitative Biosciences, University of California, Berkeley, CA 94720, USA
| | - Ravinder Kumar
- Department of Pathology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| |
Collapse
|
25
|
Kennedy PGE, Fultz M, Phares J, Yu X. Immunoglobulin G and Complement as Major Players in the Neurodegeneration of Multiple Sclerosis. Biomolecules 2024; 14:1210. [PMID: 39456143 PMCID: PMC11506455 DOI: 10.3390/biom14101210] [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: 08/13/2024] [Revised: 09/22/2024] [Accepted: 09/24/2024] [Indexed: 10/28/2024] Open
Abstract
Multiple Sclerosis (MS) is an inflammatory, demyelinating, and neurodegenerative disease of the central nervous system (CNS) and is termed as one of the most common causes of neurological disability in young adults. Axonal loss and neuronal cell damage are the primary causes of disease progression and disability. Yet, little is known about the mechanism of neurodegeneration in the disease, a limitation that impairs the development of more effective treatments for progressive MS. MS is characterized by the presence of oligoclonal bands and raised levels of immunoglobulins in the CNS. The role of complement in the demyelinating process has been detected in both experimental animal models of MS and within the CNS of affected MS patients. Furthermore, both IgG antibodies and complement activation can be detected in the demyelinating plaques and cortical gray matter lesions. We propose here that both immunoglobulins and complement play an active role in the neurodegenerative process of MS. We hypothesize that the increased CNS IgG antibodies form IgG aggregates and bind complement C1q with high affinity, activating the classical complement pathway. This results in neuronal cell damage, which leads to neurodegeneration and demyelination in MS.
Collapse
Affiliation(s)
- Peter G. E. Kennedy
- Institute of Neuroscience and Psychology, University of Glasgow, Glasgow G61 1QH, UK;
| | - Matthew Fultz
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (M.F.); (J.P.)
| | - Jeremiah Phares
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (M.F.); (J.P.)
| | - Xiaoli Yu
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (M.F.); (J.P.)
| |
Collapse
|
26
|
Bose A, Pahan K. Build muscles and protect myelin. NEUROIMMUNE PHARMACOLOGY AND THERAPEUTICS 2024; 3:175-182. [PMID: 39741558 PMCID: PMC11683878 DOI: 10.1515/nipt-2024-0015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 10/02/2024] [Indexed: 01/03/2025]
Abstract
Multiple sclerosis (MS) is a chronic and debilitating autoimmune disease of the central nervous system (CNS) in which a CNS-driven immune response destroys myelin, leading to wide range of symptoms including numbness and tingling, vision problems, mobility impairment, etc. Oligodendrocytes are the myelinating cells in the CNS, which are generated from oligodendroglial progenitor cells (OPCs) via differentiation. However, for multiple reasons, OPCs fail to differentiate to oligodendrocytes in MS and as a result, stimulating the differentiation of OPCs to oligodendrocytes is considered beneficial for MS. The β-hydroxy β-methylbutyrate (HMB) is a widely-used muscle-building supplement in human and recently it has been shown that low-dose HMB is capable of stimulating the differentiation of cultured OPCs to oligodendrocytes for remyelination. Moreover, other causes of autoimmune demyelination are the decrease and/or suppression of Foxp3-expressing anti-autoimmune regulatory T cells (Tregs) and upregulation of autoimmune T-helper 1(Th1) and Th17 cells. Experimental autoimmune encephalomyelitis (EAE) is an animal model of MS in which the autoimmune demyelination is nicely visible. It has been reported that in EAE mice, oral HMB upregulates Tregs and decreases Th1 and Th17 responses, leading to remyelination in the CNS. Here, we analyze these newly-described features of HMB, highlighting the putative promyelinating nature of this supplement.
Collapse
Affiliation(s)
- Ahana Bose
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, USA
| | - Kalipada Pahan
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
27
|
Berndt C, Alborzinia H, Amen VS, Ayton S, Barayeu U, Bartelt A, Bayir H, Bebber CM, Birsoy K, Böttcher JP, Brabletz S, Brabletz T, Brown AR, Brüne B, Bulli G, Bruneau A, Chen Q, DeNicola GM, Dick TP, Distéfano A, Dixon SJ, Engler JB, Esser-von Bieren J, Fedorova M, Friedmann Angeli JP, Friese MA, Fuhrmann DC, García-Sáez AJ, Garbowicz K, Götz M, Gu W, Hammerich L, Hassannia B, Jiang X, Jeridi A, Kang YP, Kagan VE, Konrad DB, Kotschi S, Lei P, Le Tertre M, Lev S, Liang D, Linkermann A, Lohr C, Lorenz S, Luedde T, Methner A, Michalke B, Milton AV, Min J, Mishima E, Müller S, Motohashi H, Muckenthaler MU, Murakami S, Olzmann JA, Pagnussat G, Pan Z, Papagiannakopoulos T, Pedrera Puentes L, Pratt DA, Proneth B, Ramsauer L, Rodriguez R, Saito Y, Schmidt F, Schmitt C, Schulze A, Schwab A, Schwantes A, Soula M, Spitzlberger B, Stockwell BR, Thewes L, Thorn-Seshold O, Toyokuni S, Tonnus W, Trumpp A, Vandenabeele P, Vanden Berghe T, Venkataramani V, Vogel FCE, von Karstedt S, Wang F, Westermann F, Wientjens C, Wilhelm C, Wölk M, Wu K, Yang X, Yu F, Zou Y, Conrad M. Ferroptosis in health and disease. Redox Biol 2024; 75:103211. [PMID: 38908072 PMCID: PMC11253697 DOI: 10.1016/j.redox.2024.103211] [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: 03/21/2024] [Revised: 05/24/2024] [Accepted: 05/24/2024] [Indexed: 06/24/2024] Open
Abstract
Ferroptosis is a pervasive non-apoptotic form of cell death highly relevant in various degenerative diseases and malignancies. The hallmark of ferroptosis is uncontrolled and overwhelming peroxidation of polyunsaturated fatty acids contained in membrane phospholipids, which eventually leads to rupture of the plasma membrane. Ferroptosis is unique in that it is essentially a spontaneous, uncatalyzed chemical process based on perturbed iron and redox homeostasis contributing to the cell death process, but that it is nonetheless modulated by many metabolic nodes that impinge on the cells' susceptibility to ferroptosis. Among the various nodes affecting ferroptosis sensitivity, several have emerged as promising candidates for pharmacological intervention, rendering ferroptosis-related proteins attractive targets for the treatment of numerous currently incurable diseases. Herein, the current members of a Germany-wide research consortium focusing on ferroptosis research, as well as key external experts in ferroptosis who have made seminal contributions to this rapidly growing and exciting field of research, have gathered to provide a comprehensive, state-of-the-art review on ferroptosis. Specific topics include: basic mechanisms, in vivo relevance, specialized methodologies, chemical and pharmacological tools, and the potential contribution of ferroptosis to disease etiopathology and progression. We hope that this article will not only provide established scientists and newcomers to the field with an overview of the multiple facets of ferroptosis, but also encourage additional efforts to characterize further molecular pathways modulating ferroptosis, with the ultimate goal to develop novel pharmacotherapies to tackle the various diseases associated with - or caused by - ferroptosis.
Collapse
Affiliation(s)
- Carsten Berndt
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Hamed Alborzinia
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM GGmbH), Heidelberg, Germany; Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Vera Skafar Amen
- Rudolf Virchow Zentrum, Center for Integrative and Translational Bioimaging - University of Würzburg, Germany
| | - Scott Ayton
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Australia
| | - Uladzimir Barayeu
- Division of Redox Regulation, DKFZ-ZMBH Alliance, German Cancer Research Center (DKFZ) Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, 69120, Heidelberg, Germany; Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Alexander Bartelt
- Institute for Cardiovascular Prevention (IPEK), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany; Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, Neuherberg, Germany; German Center for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Hülya Bayir
- Department of Pediatrics, Columbia University, New York City, NY, USA
| | - Christina M Bebber
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Translational Genomics, Cologne, Germany; CECAD Cluster of Excellence, University of Cologne, Cologne, Germany
| | - Kivanc Birsoy
- Laboratory of Metabolic Regulation and Genetics, Rockefeller University, New York City, NY, USA
| | - Jan P Böttcher
- Institute of Molecular Immunology, School of Medicine, Technical University of Munich (TUM), Germany
| | - Simone Brabletz
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Germany
| | - Thomas Brabletz
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Germany
| | - Ashley R Brown
- Department of Biological Sciences, Columbia University, New York City, NY, USA
| | - Bernhard Brüne
- Institute of Biochemistry1-Pathobiochemistry, Goethe-Universität, Frankfurt Am Main, Germany
| | - Giorgia Bulli
- Department of Physiological Genomics, Ludwig-Maximilians-University, Munich, Germany
| | - Alix Bruneau
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Berlin, Germany
| | - Quan Chen
- College of Life Sciences, Nankai University, Tianjin, China
| | - Gina M DeNicola
- Department of Metabolism and Physiology, Moffitt Cancer Center, Tampa, FL, USA
| | - Tobias P Dick
- Division of Redox Regulation, DKFZ-ZMBH Alliance, German Cancer Research Center (DKFZ) Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, 69120, Heidelberg, Germany
| | - Ayelén Distéfano
- Instituto de Investigaciones Biológicas, CONICET, National University of Mar Del Plata, Argentina
| | - Scott J Dixon
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Jan B Engler
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Germany
| | | | - Maria Fedorova
- Center of Membrane Biochemistry and Lipid Research, University Hospital Carl Gustav Carus and Faculty of Medicine of TU Dresden, Germany
| | - José Pedro Friedmann Angeli
- Rudolf Virchow Zentrum, Center for Integrative and Translational Bioimaging - University of Würzburg, Germany
| | - Manuel A Friese
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Germany
| | - Dominic C Fuhrmann
- Institute of Biochemistry1-Pathobiochemistry, Goethe-Universität, Frankfurt Am Main, Germany
| | - Ana J García-Sáez
- Institute for Genetics, CECAD, University of Cologne, Germany; Max Planck Institute of Biophysics, Frankfurt/Main, Germany
| | | | - Magdalena Götz
- Department of Physiological Genomics, Ludwig-Maximilians-University, Munich, Germany; Institute of Stem Cell Research, Helmholtz Center Munich, Germany
| | - Wei Gu
- Institute for Cancer Genetics, And Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA; Department of Pathology and Cell Biology, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Linda Hammerich
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Berlin, Germany
| | | | - Xuejun Jiang
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Aicha Jeridi
- Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Comprehensive Pneumology Center (CPC-M), Germany, Member of the German Center for Lung Research (DZL)
| | - Yun Pyo Kang
- College of Pharmacy and Research Institute of Pharmaceutical Science, Seoul National University, Republic of Korea
| | | | - David B Konrad
- Department of Pharmacy, Ludwig-Maximilians-University, Munich, Germany
| | - Stefan Kotschi
- Institute for Cardiovascular Prevention (IPEK), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Peng Lei
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Marlène Le Tertre
- Center for Translational Biomedical Iron Research, Heidelberg University, Germany
| | - Sima Lev
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Deguang Liang
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Andreas Linkermann
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Germany; Division of Nephrology, Department of Medicine, Albert Einstein College of Medicine, New York, NY, USA
| | - Carolin Lohr
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Svenja Lorenz
- Institute of Metabolism and Cell Death, Helmholtz Center Munich, Germany
| | - Tom Luedde
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Axel Methner
- Institute of Molecular Medicine, Johannes Gutenberg-Universität Mainz, Germany
| | - Bernhard Michalke
- Research Unit Analytical Biogeochemistry, Helmholtz Center Munich, Germany
| | - Anna V Milton
- Department of Pharmacy, Ludwig-Maximilians-University, Munich, Germany
| | - Junxia Min
- School of Medicine, Zhejiang University, Hangzhou, China
| | - Eikan Mishima
- Institute of Metabolism and Cell Death, Helmholtz Center Munich, Germany
| | | | - Hozumi Motohashi
- Department of Gene Expression Regulation, Tohoku University, Sendai, Japan
| | | | - Shohei Murakami
- Department of Gene Expression Regulation, Tohoku University, Sendai, Japan
| | - James A Olzmann
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA; Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA, USA; Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Gabriela Pagnussat
- Instituto de Investigaciones Biológicas, CONICET, National University of Mar Del Plata, Argentina
| | - Zijan Pan
- School of Life Sciences, Westlake University, Hangzhou, China
| | | | | | - Derek A Pratt
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Canada
| | - Bettina Proneth
- Institute of Metabolism and Cell Death, Helmholtz Center Munich, Germany
| | - Lukas Ramsauer
- Institute of Molecular Immunology, School of Medicine, Technical University of Munich (TUM), Germany
| | | | - Yoshiro Saito
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Felix Schmidt
- Institute of Molecular Medicine, Johannes Gutenberg-Universität Mainz, Germany
| | - Carina Schmitt
- Department of Pharmacy, Ludwig-Maximilians-University, Munich, Germany
| | - Almut Schulze
- Division of Tumour Metabolism and Microenvironment, DKFZ Heidelberg and DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Annemarie Schwab
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Germany
| | - Anna Schwantes
- Institute of Biochemistry1-Pathobiochemistry, Goethe-Universität, Frankfurt Am Main, Germany
| | - Mariluz Soula
- Laboratory of Metabolic Regulation and Genetics, Rockefeller University, New York City, NY, USA
| | - Benedikt Spitzlberger
- Department of Immunobiology, Université de Lausanne, Switzerland; Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, Munich, Germany
| | - Brent R Stockwell
- Department of Biological Sciences, Columbia University, New York City, NY, USA; Department of Pathology and Cell Biology, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA; Department of Chemistry, Columbia University, New York, NY, USA
| | - Leonie Thewes
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | | | - Shinya Toyokuni
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, Japan; Center for Low-temperature Plasma Sciences, Nagoya University, Nagoya, Japan; Center for Integrated Sciences of Low-temperature Plasma Core Research (iPlasma Core), Tokai National Higher Education and Research System, Nagoya, Japan
| | - Wulf Tonnus
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Germany
| | - Andreas Trumpp
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM GGmbH), Heidelberg, Germany; Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Peter Vandenabeele
- VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Tom Vanden Berghe
- Department of Biomedical Sciences, University of Antwerp, Belgium; VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Vivek Venkataramani
- Comprehensive Cancer Center Mainfranken, University Hospital Würzburg, Germany
| | - Felix C E Vogel
- Division of Tumour Metabolism and Microenvironment, DKFZ Heidelberg and DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Silvia von Karstedt
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Translational Genomics, Cologne, Germany; CECAD Cluster of Excellence, University of Cologne, Cologne, Germany; University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Molecular Medicine Cologne, Germany
| | - Fudi Wang
- School of Medicine, Zhejiang University, Hangzhou, China
| | | | - Chantal Wientjens
- Immunopathology Unit, Institute of Clinical Chemistry and Clinical Pharmacology, Medical Faculty, University Hospital Bonn, University of Bonn, Germany
| | - Christoph Wilhelm
- Immunopathology Unit, Institute of Clinical Chemistry and Clinical Pharmacology, Medical Faculty, University Hospital Bonn, University of Bonn, Germany
| | - Michele Wölk
- Center of Membrane Biochemistry and Lipid Research, University Hospital Carl Gustav Carus and Faculty of Medicine of TU Dresden, Germany
| | - Katherine Wu
- Department of Pathology, Grossman School of Medicine, New York University, NY, USA
| | - Xin Yang
- Institute for Cancer Genetics, And Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Fan Yu
- College of Life Sciences, Nankai University, Tianjin, China
| | - Yilong Zou
- School of Life Sciences, Westlake University, Hangzhou, China; Westlake Four-Dimensional Dynamic Metabolomics (Meta4D) Laboratory, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
| | - Marcus Conrad
- Institute of Metabolism and Cell Death, Helmholtz Center Munich, Germany.
| |
Collapse
|
28
|
Lozinski BM, Ta K, Dong Y. Emerging role of galectin 3 in neuroinflammation and neurodegeneration. Neural Regen Res 2024; 19:2004-2009. [PMID: 38227529 DOI: 10.4103/1673-5374.391181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 11/15/2023] [Indexed: 01/17/2024] Open
Abstract
Neuroinflammation and neurodegeneration are key processes that mediate the development and progression of neurological diseases. However, the mechanisms modulating these processes in different diseases remain incompletely understood. Advances in single cell based multi-omic analyses have helped to identify distinct molecular signatures such as Lgals3 that is associated with neuroinflammation and neurodegeneration in the central nervous system (CNS). Lgals3 encodes galectin-3 (Gal3), a β-galactoside and glycan binding glycoprotein that is frequently upregulated by reactive microglia/macrophages in the CNS during various neurological diseases. While Gal3 has previously been associated with non-CNS inflammatory and fibrotic diseases, recent studies highlight Gal3 as a prominent regulator of inflammation and neuroaxonal damage in the CNS during diseases such as multiple sclerosis, Alzheimer's disease, and Parkinson's disease. In this review, we summarize the pleiotropic functions of Gal3 and discuss evidence that demonstrates its detrimental role in neuroinflammation and neurodegeneration during different neurological diseases. We also consider the challenges of translating preclinical observations into targeting Gal3 in the human CNS.
Collapse
Affiliation(s)
- Brian M Lozinski
- Department of Clinical Neuroscience, University of Calgary, Calgary, AB, Canada
| | - Khanh Ta
- Deparment of Biochemistry, Microbiology & Immunology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Yifei Dong
- Deparment of Biochemistry, Microbiology & Immunology, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
29
|
He Y, Xie H, Xu Z, Zhang L, Feng Y, Long Y, Wang S, He Y, Li J, Zou Y, Zheng W, Xiao L. Rapid and prolonged response of oligodendrocyte lineage cells in standard acute cuprizone demyelination model revealed by in situ hybridization. Neurosci Lett 2024; 836:137869. [PMID: 38852766 DOI: 10.1016/j.neulet.2024.137869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 05/29/2024] [Accepted: 06/06/2024] [Indexed: 06/11/2024]
Abstract
Dietary administration of a copper chelator, cuprizone (CPZ), has long been reported to induce intense and reproducible demyelination of several brain structures such as the corpus callosum. Despite the widespread use of CPZ as an animal model for demyelinating diseases such as multiple sclerosis (MS), the mechanism by which it induces demyelination and then allows robust remyelination is still unclear. An intensive mapping of the cell dynamics of oligodendrocyte (OL) lineage during the de- and remyelination course would be particularly important for a deeper understanding of this model. Here, using a panel of OL lineage cell markers as in situ hybridization (ISH) probes, including Pdgfra, Plp, Mbp, Mog, Enpp6, combined with immunofluorescence staining of CC1, SOX10, we provide a detailed dynamic profile of OL lineage cells during the entire course of the model from 1, 2, 3.5 days, 1, 2, 3, 4,5 weeks of CPZ treatment, as well as after 1, 2, 3, 4 weeks of recovery from CPZ treatment. The result showed an unexpected early death of mature OLs and response of OL progenitor cells (OPCs) in vivo upon CPZ challenge, and a prolonged upregulation of myelin-forming OLs compared to the intact control even 4 weeks after CPZ withdrawal. These data may serve as a basic reference system for future studies of the effects of any intervention on de- and remyelination using the CPZ model, and imply the need to optimize the timing windows for the introduction of pro-remyelination therapies in demyelinating diseases such as MS.
Collapse
Affiliation(s)
- Yuehua He
- Key Laboratory of Brain, Cognition and Education Sciences of Ministry of Education, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, and Center for Studies of Psychological Application, South China Normal University, Guangzhou 510631, China
| | - Hua Xie
- Key Laboratory of Brain, Cognition and Education Sciences of Ministry of Education, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, and Center for Studies of Psychological Application, South China Normal University, Guangzhou 510631, China
| | - ZhengTao Xu
- Key Laboratory of Brain, Cognition and Education Sciences of Ministry of Education, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, and Center for Studies of Psychological Application, South China Normal University, Guangzhou 510631, China
| | - Liuning Zhang
- Key Laboratory of Brain, Cognition and Education Sciences of Ministry of Education, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, and Center for Studies of Psychological Application, South China Normal University, Guangzhou 510631, China
| | - Yuanyu Feng
- Key Laboratory of Brain, Cognition and Education Sciences of Ministry of Education, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, and Center for Studies of Psychological Application, South China Normal University, Guangzhou 510631, China
| | - Yu Long
- Key Laboratory of Brain, Cognition and Education Sciences of Ministry of Education, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, and Center for Studies of Psychological Application, South China Normal University, Guangzhou 510631, China
| | - Shuming Wang
- Key Laboratory of Brain, Cognition and Education Sciences of Ministry of Education, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, and Center for Studies of Psychological Application, South China Normal University, Guangzhou 510631, China
| | - Yongxiang He
- Key Laboratory of Brain, Cognition and Education Sciences of Ministry of Education, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, and Center for Studies of Psychological Application, South China Normal University, Guangzhou 510631, China
| | - Jiong Li
- Key Laboratory of Brain, Cognition and Education Sciences of Ministry of Education, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, and Center for Studies of Psychological Application, South China Normal University, Guangzhou 510631, China
| | - Yanping Zou
- Key Laboratory of Brain, Cognition and Education Sciences of Ministry of Education, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, and Center for Studies of Psychological Application, South China Normal University, Guangzhou 510631, China
| | - Wei Zheng
- Key Laboratory of Brain, Cognition and Education Sciences of Ministry of Education, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, and Center for Studies of Psychological Application, South China Normal University, Guangzhou 510631, China
| | - Lin Xiao
- Key Laboratory of Brain, Cognition and Education Sciences of Ministry of Education, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, and Center for Studies of Psychological Application, South China Normal University, Guangzhou 510631, China.
| |
Collapse
|
30
|
Foong YC, Merlo D, Gresle M, Buzzard K, Zhong M, Yeh WZ, Jokubaitis V, Monif M, Skibina O, Ozakbas S, Patti F, Grammond P, Amato MP, Kalincik T, Horakova D, Kubala Havrdova E, Weinstock-Guttman B, Lechner Scott J, Boz C, Sa MJ, Butzkueven H, van der Walt A, Zhu C. Comparing ocrelizumab to interferon/glatiramer acetate in people with multiple sclerosis over age 60. J Neurol Neurosurg Psychiatry 2024; 95:767-774. [PMID: 38453478 DOI: 10.1136/jnnp-2023-332883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 02/11/2024] [Indexed: 03/09/2024]
Abstract
BACKGROUND Ongoing controversy exists regarding optimal management of disease modifying therapy (DMT) in older people with multiple sclerosis (pwMS). There is concern that the lower relapse rate, combined with a higher risk of DMT-related infections and side effects, may alter the risk-benefit balance in older pwMS. Given the lack of pwMS above age 60 in randomised controlled trials, the comparative efficacy of high-efficacy DMTs such as ocrelizumab has not been shown in older pwMS. We aimed to evaluate the comparative effectiveness of ocrelizumab, a high-efficacy DMT, versus interferon/glatiramer acetate (IFN/GA) in pwMS over the age of 60. METHODS Using data from MSBase registry, this multicentre cohort study included pwMS above 60 who switched to or started on ocrelizumab or IFN/GA. We analysed relapse and disability outcomes after balancing covariates using an inverse probability treatment weighting (IPTW) method. Propensity scores were obtained based on age, country, disease duration, sex, baseline Expanded Disability Status Scale, prior relapses (all-time, 12 months and 24 months) and prior DMT exposure (overall number and high-efficacy DMTs). After weighting, all covariates were balanced. Primary outcomes were time to first relapse and annualised relapse rate (ARR). Secondary outcomes were 6-month confirmed disability progression (CDP) and confirmed disability improvement (CDI). RESULTS A total of 248 participants received ocrelizumab, while 427 received IFN/GA. The IPTW-weighted ARR for ocrelizumab was 0.01 and 0.08 for IFN/GA. The IPTW-weighted ARR ratio was 0.15 (95% CI 0.06 to 0.33, p<0.001) for ocrelizumab compared with IFN/GA. On IPTW-weighted Cox regression models, HR for time to first relapse was 0.13 (95% CI 0.05 to 0.26, p<0.001). The hazard of first relapse was significantly reduced in ocrelizumab users after 5 months compared with IFN/GA users. However, the two groups did not differ in CDP or CDI over 3.57 years. CONCLUSION In older pwMS, ocrelizumab effectively reduced relapses compared with IFN/GA. Overall relapse activity was low. This study adds valuable real-world data for informed DMT decision making with older pwMS. Our study also confirms that there is a treatment benefit in older people with MS, given the existence of a clear differential treatment effect between ocrelizumab and IFN/GA in the over 60 age group.
Collapse
Affiliation(s)
- Yi Chao Foong
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Alfred Health, Melbourne, Victoria, Australia
- Department of Neurology, Tasmanian Health Service, Hobart, Tasmania, Australia
| | - Daniel Merlo
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Eastern Health, Box Hill, Victoria, Australia
| | - Melissa Gresle
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Alfred Health, Melbourne, Victoria, Australia
- Melbourne Health, Melbourne, Victoria, Australia
| | - Katherine Buzzard
- Eastern Health, Box Hill, Victoria, Australia
- Department of Neurology, The Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Michael Zhong
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Alfred Health, Melbourne, Victoria, Australia
| | - Wei Zhen Yeh
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Alfred Health, Melbourne, Victoria, Australia
| | - Vilija Jokubaitis
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Alfred Health, Melbourne, Victoria, Australia
| | - Mastura Monif
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Alfred Health, Melbourne, Victoria, Australia
| | - Olga Skibina
- Alfred Health, Melbourne, Victoria, Australia
- Eastern Health, Box Hill, Victoria, Australia
| | | | - Francesco Patti
- Neuroscience, University of Catania Department of Surgical and Medical Sciences and Advanced Technologies 'G.F. Ingrassia', Catania, Italy
- University of Catania, Catania, Italy
| | | | - Maria Pia Amato
- Department of Neurological Siences, University of Florence, Florence, Italy
| | - Tomas Kalincik
- Department of Neurology, The Royal Melbourne Hospital, Parkville, Victoria, Australia
- CORe, Department of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| | - Dana Horakova
- Department of Neurology and Center of Clinical Neuroscience, Charles University in Prague, 1st Faculty of Medicine and General University Hospital in Prague, Prague, Czech Republic
| | - Eva Kubala Havrdova
- Department of Neurology and Center of Clinical Neuroscience, Charles University in Prague, 1st Faculty of Medicine and General University Hospital in Prague, Prague, Czech Republic
| | | | - Jeanette Lechner Scott
- Hunter New England Health, New Lambton, New South Wales, Australia
- The University of Newcastle, Newcastle, New South Wales, Australia
| | - Cavit Boz
- Karadeniz Technical University, Trabzon, Turkey
| | - Maria Jose Sa
- Neurology, Centro Hospitalar de São João, Porto, Portugal
- Faculty of Health Sciences University Fernando Pessoa, Porto, Portugal
| | - Helmut Butzkueven
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Alfred Health, Melbourne, Victoria, Australia
| | - Anneke van der Walt
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Alfred Health, Melbourne, Victoria, Australia
| | - Chao Zhu
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
31
|
Jakimovski D, Zivadinov R, Weinstock Z, Burnham A, Wicks TR, Suchan C, Sciortino T, Schweser F, Bergsland N, Dwyer MG, Eckert SP, Young-Hong D, Weinstock-Guttman B, Benedict RHB. Cognitive function in severe progressive multiple sclerosis. Brain Commun 2024; 6:fcae226. [PMID: 39015768 PMCID: PMC11250210 DOI: 10.1093/braincomms/fcae226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 05/13/2024] [Accepted: 07/01/2024] [Indexed: 07/18/2024] Open
Abstract
Cognitive impairment is common in multiple sclerosis and negatively impacts quality of life. Cognitive status has yet to be described in people with severe progressive multiple sclerosis, in whom conventional neuropsychological testing is exceptionally difficult. The objective for the study was to characterize cognitive performance in severe progressive multiple sclerosis and compare them with age-, sex- and disease duration-matched less disabled people with multiple sclerosis using a specifically developed auditory, non-motor test of attention/cognitive processing speed-Auditory Test of Processing Speed. Also, we aimed to determine the relationship between cognitive performance and MRI-based outcomes in these matched cohorts. The Comprehensive Assessment of Severely Affected Multiple Sclerosis study was carried out at the University at Buffalo and the Boston Home, a skilled nursing facility in Dorchester, MA. Inclusion criteria were age 30-80 years and expanded disability status scale 3.0-6.5 for community-dwelling and 7.0-9.5 for skilled nursing facility people with multiple sclerosis. The cognitive assessment was performed using the Brief International Cognitive Assessment for Multiple Sclerosis consisting of Symbol Digit Modalities Test, Brief Visuospatial Memory Test-Revised, California Verbal Learning Test-2nd edition along with Auditory Test of Processing Speed, Paced Auditory Serial Addition Test-3 second and Controlled Oral Word Association Test. MRI scans were retrospectively collected and analysed for lesion and volumetric brain measurements. The rate of completion and performance of the cognitive tests was compared between the groups, and the relationship with MRI measures was determined using sex, age and years of education-adjusted linear regression models. Significantly greater percentage of the severe multiple sclerosis group completed Auditory Test of Processing Speed when compared with the current gold standard of Symbol Digit Modalities Test (93.2% versus 65.9%). Severe progressive multiple sclerosis had worse cognitive performance in all cognitive domains with greatest differences for cognitive processing speed (Symbol Digit Modalities Test > Paced Auditory Serial Addition Test-3 second > Auditory Test of Processing Speed, Cohen's d < 2.13, P < 0.001), learning and memory (Cohen's d < 1.1, P < 0.001) and language (Controlled Oral Word Association Test with Cohen's d = 0.97, P < 0.001). Multiple cognitive domains were significantly associated with lower thalamic (standardized β < 0.419, P < 0.006) and cortical (standardized β < 0.26, P < 0.031) volumes. Specially designed (auditory) cognitive processing speed tests may provide more sensitive screening of cognitive function in severe progressive multiple sclerosis. The cognitive profile of severe multiple sclerosis is proportional to their physical outcomes and best explained by decreased grey matter volume.
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 14203, 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 14203, USA
- Center for Biomedical Imaging at the Clinical Translational Science Institute, University at Buffalo, State University of New York, Buffalo, NY 14203, USA
| | - Zachary Weinstock
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14203, USA
| | | | - Taylor R Wicks
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14203, USA
| | - Christopher Suchan
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14203, USA
| | - Tommaso Sciortino
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14203, USA
| | - Ferdinand Schweser
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14203, USA
- Center for Biomedical Imaging at the Clinical Translational Science Institute, University at Buffalo, State University of New York, Buffalo, NY 14203, 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 14203, USA
| | - 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 14203, USA
| | - Svetlana P Eckert
- Department of Neurology, Jacobs Comprehensive MS Treatment and Research Center, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14203 USA
| | | | - Bianca Weinstock-Guttman
- Department of Neurology, Jacobs Comprehensive MS Treatment and Research Center, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14203 USA
| | - Ralph H B Benedict
- Department of Neurology, Jacobs Comprehensive MS Treatment and Research Center, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14203 USA
| |
Collapse
|
32
|
Rajalingam A, Ganjiwale A. Identification of common genetic factors and immune-related pathways associating more than two autoimmune disorders: implications on risk, diagnosis, and treatment. Genomics Inform 2024; 22:10. [PMID: 38956704 PMCID: PMC11221123 DOI: 10.1186/s44342-024-00004-5] [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: 09/21/2023] [Accepted: 12/22/2023] [Indexed: 07/04/2024] Open
Abstract
Autoimmune disorders (ADs) are chronic conditions resulting from failure or breakdown of immunological tolerance, resulting in the host immune system attacking its cells or tissues. Recent studies report shared effects, mechanisms, and evolutionary origins among ADs; however, the possible factors connecting them are unknown. This study attempts to identify gene signatures commonly shared between different autoimmune disorders and elucidate their molecular pathways linking the pathogenesis of these ADs using an integrated gene expression approach. We employed differential gene expression analysis across 19 datasets of whole blood/peripheral blood cell samples with five different autoimmune disorders (rheumatoid arthritis, multiple sclerosis, systemic lupus erythematosus, Crohn's disease, and type 1 diabetes) to get nine key genes-EGR1, RUNX3, SMAD7, NAMPT, S100A9, S100A8, CYBB, GATA2, and MCEMP1 that were primarily involved in cell and leukocyte activation, leukocyte mediated immunity, IL-17, AGE-RAGE signaling in diabetic complications, prion disease, and NOD-like receptor signaling confirming its role in immune-related pathways. Combined with biological interpretations such as gene ontology (GO), pathway enrichment, and protein-protein interaction (PPI) network, our current study sheds light on the in-depth research on early detection, diagnosis, and prognosis of different ADs.
Collapse
Affiliation(s)
- Aruna Rajalingam
- Department of Life Science, Bangalore University, Bangalore, Karnataka, 560056, India
| | - Anjali Ganjiwale
- Department of Life Science, Bangalore University, Bangalore, Karnataka, 560056, India.
| |
Collapse
|
33
|
Jana M, Prieto S, Gorai S, Dasarathy S, Kundu M, Pahan K. Muscle-building supplement β-hydroxy β-methylbutyrate stimulates the maturation of oligodendroglial progenitor cells to oligodendrocytes. J Neurochem 2024; 168:1340-1358. [PMID: 38419348 PMCID: PMC11260247 DOI: 10.1111/jnc.16084] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 01/10/2024] [Accepted: 02/06/2024] [Indexed: 03/02/2024]
Abstract
Oligodendrocytes are the myelinating cells in the CNS and multiple sclerosis (MS) is a demyelinating disorder that is characterized by progressive loss of myelin. Although oligodendroglial progenitor cells (OPCs) should be differentiated into oligodendrocytes, for multiple reasons, OPCs fail to differentiate into oligodendrocytes in MS. Therefore, increasing the maturation of OPCs to oligodendrocytes may be of therapeutic benefit for MS. The β-hydroxy β-methylbutyrate (HMB) is a muscle-building supplement in humans and this study underlines the importance of HMB in stimulating the maturation of OPCs to oligodendrocytes. HMB treatment upregulated the expression of different maturation markers including PLP, MBP, and MOG in cultured OPCs. Double-label immunofluorescence followed by immunoblot analyses confirmed the upregulation of OPC maturation by HMB. While investigating mechanisms, we found that HMB increased the maturation of OPCs isolated from peroxisome proliferator-activated receptor β-/- (PPARβ-/-) mice, but not PPARα-/- mice. Similarly, GW6471 (an antagonist of PPARα), but not GSK0660 (an antagonist of PPARβ), inhibited HMB-induced maturation of OPCs. GW9662, a specific inhibitor of PPARγ, also could not inhibit HMB-mediated stimulation of OPC maturation. Furthermore, PPARα agonist GW7647, but neither PPARβ agonist GW0742 nor PPARγ agonist GW1929, alone increased the maturation of OPCs. Finally, HMB treatment of OPCs led to the recruitment of PPARα, but neither PPARβ nor PPARγ, to the PLP gene promoter. These results suggest that HMB stimulates the maturation of OPCs via PPARα and that HMB may have therapeutic prospects in remyelination.
Collapse
Affiliation(s)
- Malabendu Jana
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, USA
| | - Shelby Prieto
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Sukhamoy Gorai
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Sridevi Dasarathy
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Madhuchhanda Kundu
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Kalipada Pahan
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, USA
| |
Collapse
|
34
|
Woo MS, Engler JB, Friese MA. The neuropathobiology of multiple sclerosis. Nat Rev Neurosci 2024; 25:493-513. [PMID: 38789516 DOI: 10.1038/s41583-024-00823-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/30/2024] [Indexed: 05/26/2024]
Abstract
Chronic low-grade inflammation and neuronal deregulation are two components of a smoldering disease activity that drives the progression of disability in people with multiple sclerosis (MS). Although several therapies exist to dampen the acute inflammation that drives MS relapses, therapeutic options to halt chronic disability progression are a major unmet clinical need. The development of such therapies is hindered by our limited understanding of the neuron-intrinsic determinants of resilience or vulnerability to inflammation. In this Review, we provide a neuron-centric overview of recent advances in deciphering neuronal response patterns that drive the pathology of MS. We describe the inflammatory CNS environment that initiates neurotoxicity by imposing ion imbalance, excitotoxicity and oxidative stress, and by direct neuro-immune interactions, which collectively lead to mitochondrial dysfunction and epigenetic dysregulation. The neuronal demise is further amplified by breakdown of neuronal transport, accumulation of cytosolic proteins and activation of cell death pathways. Continuous neuronal damage perpetuates CNS inflammation by activating surrounding glia cells and by directly exerting toxicity on neighbouring neurons. Further, we explore strategies to overcome neuronal deregulation in MS and compile a selection of neuronal actuators shown to impact neurodegeneration in preclinical studies. We conclude by discussing the therapeutic potential of targeting such neuronal actuators in MS, including some that have already been tested in interventional clinical trials.
Collapse
Affiliation(s)
- Marcel S Woo
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Jan Broder Engler
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Manuel A Friese
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
35
|
Cujbă L, Banc A, Stan C, Drugan T, Nicula C. Macular OCT's Proficiency in Identifying Retrochiasmal Visual Pathway Lesions in Multiple Sclerosis-A Pilot Study. Diagnostics (Basel) 2024; 14:1221. [PMID: 38928637 PMCID: PMC11202879 DOI: 10.3390/diagnostics14121221] [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: 04/29/2024] [Revised: 06/01/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Optical coherence tomography (OCT) is a non-invasive imaging technique based on the principle of low-coherence interferometry that captures detailed images of ocular structures. Multiple sclerosis (MS) is a neurodegenerative disease that can lead to damage of the optic nerve and retina, which can be depicted by OCT. The purpose of this pilot study is to determine whether macular OCT can be used as a biomarker in the detection of retrochiasmal lesions of the visual pathway in MS patients. We conducted a prospective study in which we included 52 MS patients and 27 healthy controls. All participants underwent brain MRI, visual field testing, and OCT evaluation of the thicknesses of the peripapillary retinal nerve fiber layer (pRNFL), macular ganglion cell layer (GCL), and macular inner plexiform layer (IPL). OCT measurements were adjusted for optic neuritis (ON). VF demonstrated poor capability to depict a retrochiasmal lesion identified by brain MRI (PPV 0.50). In conclusion, the OCT analysis of the macula appears to excel in identifying retrochiasmal MS lesions compared to VF changes. The alterations in the GCL and IPL demonstrate the most accurate detection of retrochiasmal visual pathway changes in MS patients.
Collapse
Affiliation(s)
- Larisa Cujbă
- Medical Doctoral School, University of Oradea, 410087 Oradea, Romania;
| | - Ana Banc
- Department of Ophthalmology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
| | - Cristina Stan
- Department of Ophthalmology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
| | - Tudor Drugan
- Department of Medical Informatics and Biostatistics, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Cristina Nicula
- Department of Maxillo-Facial Surgery and Radiology, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
| |
Collapse
|
36
|
Gisevius B, Duscha A, Poschmann G, Stühler K, Motte J, Fisse AL, Augustyniak S, Rehm A, Renk P, Böse C, Hubert D, Peters K, Jagst M, Gömer A, Todt D, Bader V, Tokic M, Hirschberg S, Krogias C, Trampe N, Coutourier C, Winnesberg C, Steinmann E, Winklhofer K, Gold R, Haghikia A. Propionic acid promotes neurite recovery in damaged multiple sclerosis neurons. Brain Commun 2024; 6:fcae182. [PMID: 38894951 PMCID: PMC11184351 DOI: 10.1093/braincomms/fcae182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 03/21/2024] [Accepted: 05/31/2024] [Indexed: 06/21/2024] Open
Abstract
Neurodegeneration in the autoimmune disease multiple sclerosis still poses a major therapeutic challenge. Effective drugs that target the inflammation can only partially reduce accumulation of neurological deficits and conversion to progressive disease forms. Diet and the associated gut microbiome are currently being discussed as crucial environmental risk factors that determine disease onset and subsequent progression. In people with multiple sclerosis, supplementation of the short-chain fatty acid propionic acid, as a microbial metabolite derived from the fermentation of a high-fiber diet, has previously been shown to regulate inflammation accompanied by neuroprotective properties. We set out to determine whether the neuroprotective impact of propionic acid is a direct mode of action of short-chain fatty acids on CNS neurons. We analysed neurite recovery in the presence of the short-chain fatty acid propionic acid and butyric acid in a reverse-translational disease-in-a-dish model of human-induced primary neurons differentiated from people with multiple sclerosis-derived induced pluripotent stem cells. We found that recovery of damaged neurites is induced by propionic acid and butyric acid. We could also show that administration of butyric acid is able to enhance propionic acid-associated neurite recovery. Whole-cell proteome analysis of induced primary neurons following recovery in the presence of propionic acid revealed abundant changes of protein groups that are associated with the chromatin assembly, translational, and metabolic processes. We further present evidence that these alterations in the chromatin assembly were associated with inhibition of histone deacetylase class I/II following both propionic acid and butyric acid treatment, mediated by free fatty acid receptor signalling. While neurite recovery in the presence of propionic acid is promoted by activation of the anti-oxidative response, administration of butyric acid increases neuronal ATP synthesis in people with multiple sclerosis-specific induced primary neurons.
Collapse
Affiliation(s)
- Barbara Gisevius
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Alexander Duscha
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
- Department of Neurology, Otto-von-Guericke University, 39120 Magdeburg, Germany
| | - Gereon Poschmann
- Institute of Molecular Medicine, Proteome Research, Medical Faculty and University Hospital, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
- Molecular Proteomics Laboratory, BMFZ, Heinrich Heine University Düsseldorf, 40335 Düsseldorf, Germany
| | - Kai Stühler
- Institute of Molecular Medicine, Proteome Research, Medical Faculty and University Hospital, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
- Molecular Proteomics Laboratory, BMFZ, Heinrich Heine University Düsseldorf, 40335 Düsseldorf, Germany
| | - Jeremias Motte
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Anna Lena Fisse
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Sanja Augustyniak
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Adriana Rehm
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Pia Renk
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Celina Böse
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Diana Hubert
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Kathrin Peters
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Michelle Jagst
- Department for Molecular and Medical Virology, Ruhr-University Bochum, 44801 Bochum, Germany
- Institute of Virology, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - André Gömer
- Department for Molecular and Medical Virology, Ruhr-University Bochum, 44801 Bochum, Germany
| | - Daniel Todt
- Department for Molecular and Medical Virology, Ruhr-University Bochum, 44801 Bochum, Germany
- European Virus Bioinformatics Center (EVBC), 07743 Jena, Germany
| | - Verian Bader
- Department of Molecular Cell Biology, Institute of Biochemistry and Pathobiochemistry, Ruhr-University Bochum, 44801 Bochum, Germany
| | - Marianne Tokic
- Department of Medical Informatics, Biometry and Epidemiology, Ruhr University Bochum, 44780 Bochum, Germany
| | - Sarah Hirschberg
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Christos Krogias
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Nadine Trampe
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Charlotta Coutourier
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Carmen Winnesberg
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Eike Steinmann
- Department for Molecular and Medical Virology, Ruhr-University Bochum, 44801 Bochum, Germany
| | - Konstanze Winklhofer
- Department of Molecular Cell Biology, Institute of Biochemistry and Pathobiochemistry, Ruhr-University Bochum, 44801 Bochum, Germany
- Cluster of Excellence RESOLV, 44801 Bochum, Germany
| | - Ralf Gold
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Aiden Haghikia
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
- Department of Neurology, Otto-von-Guericke University, 39120 Magdeburg, Germany
| |
Collapse
|
37
|
Preziosa P, Storelli L, Tedone N, Margoni M, Mistri D, Azzimonti M, Filippi M, Rocca MA. Spatial correspondence among regional gene expressions and gray matter volume loss in multiple sclerosis. Mol Psychiatry 2024; 29:1833-1843. [PMID: 38326561 DOI: 10.1038/s41380-024-02452-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 01/22/2024] [Accepted: 01/23/2024] [Indexed: 02/09/2024]
Abstract
In multiple sclerosis (MS), a non-random and clinically relevant pattern of gray matter (GM) volume loss has been described. Whether differences in regional gene expression might underlay distinctive pathological processes contributing to this regional variability has not been explored yet. Two hundred eighty-six MS patients and 172 healthy controls (HC) underwent a brain 3T MRI, a complete neurological evaluation and a neuropsychological assessment. Using Allen Human Brain Atlas, voxel-based morphometry and MENGA platform, we integrated brain transcriptome and neuroimaging data to explore the spatial cross-correlations between regional GM volume loss and expressions of 2710 genes involved in MS (p < 0.05, family-wise error-corrected). Enrichment analyses were performed to evaluate overrepresented molecular functions, biological processes and cellular components involving genes significantly associated with voxel-based morphometry-derived GM maps (p < 0.05, Bonferroni-corrected). A diffuse GM volume loss was found in MS patients compared to HC and it was spatially correlated with 74 genes involved in GABA neurotransmission and mitochondrial oxidoreductase activity mainly expressed in neurons and astrocytes. A more severe GM volume loss was spatially associated, in more disabled MS patients, with 44 genes involved in mitochondrial integrity of all resident cells of the central nervous system (CNS) and, in cognitively impaired MS patients, with 64 genes involved in mitochondrial protein heterodimerization and oxidoreductase activities expressed also in microglia and endothelial cells. Specific differences in the expressions of genes involved in synaptic GABA receptor activities and mitochondrial functions in resident CNS cells may influence regional susceptibility to MS-related excitatory/inhibitory imbalance and oxidative stress, and subsequently, to GM volume loss.
Collapse
Affiliation(s)
- Paolo Preziosa
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Loredana Storelli
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Nicolò Tedone
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Monica Margoni
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurorehabilitation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Damiano Mistri
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Matteo Azzimonti
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Massimo Filippi
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
- Neurorehabilitation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurophysiology Service, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Maria A Rocca
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.
- Vita-Salute San Raffaele University, Milan, Italy.
| |
Collapse
|
38
|
Tian H, Huang D, Wang J, Li H, Gao J, Zhong Y, Xia L, Zhang A, Lin Z, Ke X. The role of the "gut microbiota-mitochondria" crosstalk in the pathogenesis of multiple sclerosis. Front Microbiol 2024; 15:1404995. [PMID: 38741740 PMCID: PMC11089144 DOI: 10.3389/fmicb.2024.1404995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 04/11/2024] [Indexed: 05/16/2024] Open
Abstract
Multiple Sclerosis (MS) is a neurologic autoimmune disease whose exact pathophysiologic mechanisms remain to be elucidated. Recent studies have shown that the onset and progression of MS are associated with dysbiosis of the gut microbiota. Similarly, a large body of evidence suggests that mitochondrial dysfunction may also have a significant impact on the development of MS. Endosymbiotic theory has found that human mitochondria are microbial in origin and share similar biological characteristics with the gut microbiota. Therefore, gut microbiota and mitochondrial function crosstalk are relevant in the development of MS. However, the relationship between gut microbiota and mitochondrial function in the development of MS is not fully understood. Therefore, by synthesizing previous relevant literature, this paper focuses on the changes in gut microbiota and metabolite composition in the development of MS and the possible mechanisms of the crosstalk between gut microbiota and mitochondrial function in the progression of MS, to provide new therapeutic approaches for the prevention or reduction of MS based on this crosstalk.
Collapse
Affiliation(s)
- Huan Tian
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dunbing Huang
- Department of Rehabilitation Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiaqi Wang
- Department of Rehabilitation Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Huaqiang Li
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiaxin Gao
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yue Zhong
- Department of Rehabilitation Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Libin Xia
- Department of Rehabilitation Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Anren Zhang
- Department of Rehabilitation Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhonghua Lin
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
- Rehabilitation Medicine Center, Fujian Provincial Hospital, Fuzhou, China
- Fujian Provincial Center for Geriatrics, Fujian Provincia Hospital, Fuzhou, China
| | - Xiaohua Ke
- Department of Rehabilitation Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
39
|
Sanabria-Castro A, Alape-Girón A, Flores-Díaz M, Echeverri-McCandless A, Parajeles-Vindas A. Oxidative stress involvement in the molecular pathogenesis and progression of multiple sclerosis: a literature review. Rev Neurosci 2024; 35:355-371. [PMID: 38163257 DOI: 10.1515/revneuro-2023-0091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 11/26/2023] [Indexed: 01/03/2024]
Abstract
Multiple sclerosis (MS) is an autoimmune debilitating disease of the central nervous system caused by a mosaic of interactions between genetic predisposition and environmental factors. The pathological hallmarks of MS are chronic inflammation, demyelination, and neurodegeneration. Oxidative stress, a state of imbalance between the production of reactive species and antioxidant defense mechanisms, is considered one of the key contributors in the pathophysiology of MS. This review is a comprehensive overview of the cellular and molecular mechanisms by which oxidant species contribute to the initiation and progression of MS including mitochondrial dysfunction, disruption of various signaling pathways, and autoimmune response activation. The detrimental effects of oxidative stress on neurons, oligodendrocytes, and astrocytes, as well as the role of oxidants in promoting and perpetuating inflammation, demyelination, and axonal damage, are discussed. Finally, this review also points out the therapeutic potential of various synthetic antioxidants that must be evaluated in clinical trials in patients with MS.
Collapse
Affiliation(s)
- Alfredo Sanabria-Castro
- Unidad de Investigación, Hospital San Juan de Dios, Caja Costarricense de Seguro Social, San José, 10103, Costa Rica
- Departamento de Farmacología, Toxicología y Farmacodependencia, Facultad de Farmacia, Universidad de Costa Rica, San Pedro de Montes de Oca, 11501, Costa Rica
| | - Alberto Alape-Girón
- Instituto Clodomiro Picado, Facultad de Microbiología, Universidad de Costa Rica, Dulce Nombre Vázquez de Coronado, 11103, Costa Rica
| | - Marietta Flores-Díaz
- Instituto Clodomiro Picado, Facultad de Microbiología, Universidad de Costa Rica, Dulce Nombre Vázquez de Coronado, 11103, Costa Rica
| | - Ann Echeverri-McCandless
- Unidad de Investigación, Hospital San Juan de Dios, Caja Costarricense de Seguro Social, San José, 10103, Costa Rica
| | - Alexander Parajeles-Vindas
- Servicio de Neurología, Hospital San Juan de Dios, Caja Costarricense de Seguro Social, San José, 10103, Costa Rica
- Servicio de Neurología, Hospital Clínica Bíblica, San José, 10104, Costa Rica
| |
Collapse
|
40
|
Mauker P, Beckmann D, Kitowski A, Heise C, Wientjens C, Davidson AJ, Wanderoy S, Fabre G, Harbauer AB, Wood W, Wilhelm C, Thorn-Seshold J, Misgeld T, Kerschensteiner M, Thorn-Seshold O. Fluorogenic Chemical Probes for Wash-free Imaging of Cell Membrane Damage in Ferroptosis, Necrosis, and Axon Injury. J Am Chem Soc 2024. [PMID: 38592946 DOI: 10.1021/jacs.3c07662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2024]
Abstract
Selectively labeling cells with damaged membranes is needed not only for identifying dead cells in culture, but also for imaging membrane barrier dysfunction in pathologies in vivo. Most membrane permeability stains are permanently colored or fluorescent dyes that need washing to remove their non-uptaken extracellular background and reach good image contrast. Others are DNA-binding environment-dependent fluorophores, which lack design modularity, have potential toxicity, and can only detect permeabilization of cell volumes containing a nucleus (i.e., cannot delineate damaged volumes in vivo nor image non-nucleated cell types or compartments). Here, we develop modular fluorogenic probes that reveal the whole cytosolic volume of damaged cells, with near-zero background fluorescence so that no washing is needed. We identify a specific disulfonated fluorogenic probe type that only enters cells with damaged membranes, then is enzymatically activated and marks them. The esterase probe MDG1 is a reliable tool to reveal live cells that have been permeabilized by biological, biochemical, or physical membrane damage, and it can be used in multicolor microscopy. We confirm the modularity of this approach by also adapting it for improved hydrolytic stability, as the redox probe MDG2. We conclude by showing the unique performance of MDG probes in revealing axonal membrane damage (which DNA fluorogens cannot achieve) and in discriminating damage on a cell-by-cell basis in embryos in vivo. The MDG design thus provides powerful modular tools for wash-free in vivo imaging of membrane damage, and indicates how designs may be adapted for selective delivery of drug cargoes to these damaged cells: offering an outlook from selective diagnosis toward therapy of membrane-compromised cells in disease.
Collapse
Affiliation(s)
- Philipp Mauker
- Department of Pharmacy, Ludwig-Maximilians University of Munich, Butenandtstr. 7, 81377 Munich, Germany
| | - Daniela Beckmann
- Institute of Clinical Neuroimmunology, LMU University Hospital, Ludwig-Maximilians University of Munich, Marchioninistr. 15, 81377 Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians University of Munich, Grosshaderner Strasse 9, 82152 Martinsried, Germany
| | - Annabel Kitowski
- Department of Pharmacy, Ludwig-Maximilians University of Munich, Butenandtstr. 7, 81377 Munich, Germany
| | - Constanze Heise
- Department of Pharmacy, Ludwig-Maximilians University of Munich, Butenandtstr. 7, 81377 Munich, Germany
| | - Chantal Wientjens
- Immunopathology Unit, Institute of Clinical Chemistry and Clinical Pharmacology, Medical Faculty, University Hospital Bonn, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Andrew J Davidson
- Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh BioQuarter, 5 Little France Drive, Edinburgh EH16 4UU, U.K
| | - Simone Wanderoy
- University Hospital, Technical University of Munich, Ismaninger Straße 22, 81675 Munich, Germany
- Max Planck Institute for Biological Intelligence, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Gabin Fabre
- Pharmacology & Transplantation, UMR 1248 INSERM, University of Limoges, 87000 Limoges, France
| | - Angelika B Harbauer
- Max Planck Institute for Biological Intelligence, Am Klopferspitz 18, 82152 Martinsried, Germany
- Munich Cluster for Systems Neurology (SyNergy), Feodor-Lynen-Strasse 17, 81377 Munich, Germany
- Institute of Neuronal Cell Biology, Technical University of Munich, Biedersteiner Straße 29, 80802 Munich, Germany
| | - Will Wood
- Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh BioQuarter, 5 Little France Drive, Edinburgh EH16 4UU, U.K
| | - Christoph Wilhelm
- Immunopathology Unit, Institute of Clinical Chemistry and Clinical Pharmacology, Medical Faculty, University Hospital Bonn, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Julia Thorn-Seshold
- Department of Pharmacy, Ludwig-Maximilians University of Munich, Butenandtstr. 7, 81377 Munich, Germany
| | - Thomas Misgeld
- Munich Cluster for Systems Neurology (SyNergy), Feodor-Lynen-Strasse 17, 81377 Munich, Germany
- Institute of Neuronal Cell Biology, Technical University of Munich, Biedersteiner Straße 29, 80802 Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen-Strasse 17, 81377 Munich, Germany
| | - Martin Kerschensteiner
- Institute of Clinical Neuroimmunology, LMU University Hospital, Ludwig-Maximilians University of Munich, Marchioninistr. 15, 81377 Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians University of Munich, Grosshaderner Strasse 9, 82152 Martinsried, Germany
- Munich Cluster for Systems Neurology (SyNergy), Feodor-Lynen-Strasse 17, 81377 Munich, Germany
| | - Oliver Thorn-Seshold
- Department of Pharmacy, Ludwig-Maximilians University of Munich, Butenandtstr. 7, 81377 Munich, Germany
| |
Collapse
|
41
|
Sitruk-Ware R, Sussman H, Brinton R, Schumacher M, Singer P, Kumar N, De Nicola AF, El-Etr M, Guennoun R, V Borlongan C. Nestorone (segesterone acetate) effects on neuroregeneration. Front Neuroendocrinol 2024; 73:101136. [PMID: 38670433 DOI: 10.1016/j.yfrne.2024.101136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 04/08/2024] [Accepted: 04/22/2024] [Indexed: 04/28/2024]
Abstract
Nestorone® (segesterone acetate) is a progestin with a chemical structure closely related to progesterone with high affinity and selectivity for the progesterone receptor without significant interaction with other steroid receptors. It has been developed for female and male contraception and is FDA-approved in a first long-acting contraceptive vaginal system for female contraception. Its safety has been extensively demonstrated in both preclinical and clinical studies for contraceptive indications. Nestorone was found to display neuroprotective and neuroregenerative activity in animal models of various central nervous system diseases, including multiple sclerosis, stroke, and amyotrophic lateral sclerosis. Reviewed herein are neuroprotective and myelin- regenerating properties of Nestorone in various animal models and its translational potential as a therapeutic agent for debilitating neurological diseases for which limited therapeutic options are available (Table 1).
Collapse
Affiliation(s)
| | | | - Roberta Brinton
- Center for Innovation in Brain Science, Tucson, AZ, United States
| | | | | | | | | | - Martine El-Etr
- U1195 Inserm and University Paris-Saclay Le Kremlin Bicêtre, France
| | - Rachida Guennoun
- U1195 Inserm and University Paris-Saclay Le Kremlin Bicêtre, France
| | - Cesar V Borlongan
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| |
Collapse
|
42
|
Hou B, Yin J, Liu S, Guo J, Zhang B, Zhang Z, Yang L, Tan X, Long Y, Feng S, Zhou J, Wu Y, Wang X, Han S, Wang Z, He X. Inhibiting the NLRP3 Inflammasome with MCC950 Alleviates Neurological Impairment in the Brain of EAE Mice. Mol Neurobiol 2024; 61:1318-1330. [PMID: 37702910 PMCID: PMC10896958 DOI: 10.1007/s12035-023-03618-y] [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: 06/14/2023] [Accepted: 08/27/2023] [Indexed: 09/14/2023]
Abstract
Multiple sclerosis (MS) is a chronic disease that is characterized by demyelination and neuronal damage. Experimental autoimmune encephalomyelitis (EAE) mice are used to model the disease progression of MS and mirror MS-like pathology. Previous researches have confirmed that inhibition of NLRP3 inflammasome significantly alleviated the severity of EAE mice and the demyelination of spinal cord, but its effect on neuronal damage and oligodendrocyte loss in the brain remains unclear. In this study, female C57BL/6 mice were immunized with MOG35-55 and PTX to establish experimental autoimmune encephalomyelitis (EAE) model. MCC950, a selective NLRP3 inflammasome inhibitor, was used to investigate the effect of NLRP3 inflammasome on the pathological changes and glial cell activation in the brain of EAE mice by immunohistochemistry. Our results demonstrated that MCC950 ameliorated the neuronal damage, demyelination, and oligodendrocyte loss in the brain of EAE mice. This protective effect of MCC950 may be attributed to its ability to suppress the activation of glial cells and prevents microglia polarization to M1 phenotype. Our work indicates that inhibition of NLRP3 inflammasome has the therapeutic effects of neuroprotection through immunomodulation and is a promising therapeutic strategy for MS.
Collapse
Affiliation(s)
- Baohua Hou
- College of Medicine, Henan Polytechnic University, Jiaozuo, 454000, China
- Department of Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430000, China
- Central Laboratory, The First Affiliated Hospital of Henan Polytechnic University (Jiaozuo Second People's Hospital), Jiaozuo, China
| | - Jun Yin
- Department of Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430000, China
| | - Shuyan Liu
- Department of Endocrinology, The First Affiliated Hospital of Henan Polytechnic University (Jiaozuo Second People's Hospital), Jiaozuo, 454000, China
| | - Jincheng Guo
- Department of Thoracic Surgery, The First Affiliated Hospital of Henan Polytechnic University (Jiaozuo Second People's Hospital), Jiaozuo, 454000, China
| | - Baobao Zhang
- College of Medicine, Henan Polytechnic University, Jiaozuo, 454000, China
| | - Zhenzhen Zhang
- College of Medicine, Henan Polytechnic University, Jiaozuo, 454000, China
| | - Lanping Yang
- College of Medicine, Henan Polytechnic University, Jiaozuo, 454000, China
| | - Xiying Tan
- College of Medicine, Henan Polytechnic University, Jiaozuo, 454000, China
| | - Yijiao Long
- College of Medicine, Henan Polytechnic University, Jiaozuo, 454000, China
| | - Sijie Feng
- College of Medicine, Henan Polytechnic University, Jiaozuo, 454000, China
| | - Jingchun Zhou
- Beijing Bencaoyuan Pharmaceutical Co, Ltd, Beijing, 102629, China
| | - Yifan Wu
- Department of Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430000, China
| | - Xueyang Wang
- Department of Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430000, China
| | - Song Han
- Department of Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430000, China
| | - Zhenhui Wang
- College of Medicine, Henan Polytechnic University, Jiaozuo, 454000, China.
| | - Xiaohua He
- Department of Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430000, China.
| |
Collapse
|
43
|
Dolcetti E, Buttari F, Bruno A, Azzolini F, Gilio L, Di Caprio V, Lauritano G, Borrelli A, Galifi G, Furlan R, Finardi A, Musella A, Guadalupi L, Mandolesi G, Rovella V, Centonze D, Stampanoni Bassi M. Low-contrast visual acuity test is associated with central inflammation and predicts disability development in newly diagnosed multiple sclerosis patients. Front Neurol 2024; 15:1326506. [PMID: 38585351 PMCID: PMC10995923 DOI: 10.3389/fneur.2024.1326506] [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: 10/23/2023] [Accepted: 02/14/2024] [Indexed: 04/09/2024] Open
Abstract
Introduction The visual system is a prominent site of damage in MS since the earliest phases of the disease. Altered low-contrast visual acuity (LCVA) test has been associated with visual impairment and retinal degeneration, predicting medium- and long-term disability. However, it is unclear whether LCVA may also represent a reliable measure of neuroinflammation and a predictor of disease evolution in the very early stages of MS. Methods We explored in a group of 76 consecutive newly diagnosed relapsing-remitting MS (RR-MS) patients without visual impairment or altered visual evoked potentials, the association between LCVA scores at 2.5% and 1.25% and clinical characteristics, including prospective disability evaluated after 1- and 2 years of follow-up. Associations between LCVA and the CSF levels of IL-10 at diagnosis were also analyzed. Results A negative correlation was found between LCVA at 2.5% and Expanded Disability Status Scale (EDSS) evaluated at first (Spearman's Rho = -0.349, p = 0.005, n = 62) and second year (Spearman's Rho = -0.418, p < 0.001, n = 62) of follow-up, and negative correlations were found with Multiple Sclerosis Severity Score (MSSS) at first (Spearman's Rho = -0.359, p = 0.004, n = 62) and second year (Spearman's Rho = -0.472, p < 0.001, n = 62). All the data were confirmed by a mixed effect model, considering other clinical variables. A positive correlation was found between the CSF concentrations of IL-10 and LCVA at 2.5% (Spearman's Rho = 0.272, p = 0.020, n = 76), and 1.25% (Spearman's Rho, = 0.276, p = 0.018, n = 76), also evidenced in a linear regression. Discussion In MS patients at diagnosis, altered LCVA may be associated with CSF inflammation and represent a useful parameter to identify patients with worse disease course.
Collapse
Affiliation(s)
| | - Fabio Buttari
- Neurology Unit, IRCSS Neuromed, Pozzilli, Italy
- Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | | | | | - Luana Gilio
- Neurology Unit, IRCSS Neuromed, Pozzilli, Italy
- Faculty of Psychology, Uninettuno Telematic International University, Rome, Italy
| | | | | | | | | | - Roberto Furlan
- Clinical Neuroimmunology Unit, Institute of Experimental Neurology (INSpe), Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Annamaria Finardi
- Clinical Neuroimmunology Unit, Institute of Experimental Neurology (INSpe), Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Alessandra Musella
- Synaptic Immunopathology Lab, IRCCS San Raffaele Roma, Rome, Italy
- Department of Human Sciences and Quality of Life Promotion, University of Rome San Raffaele, Rome, Italy
| | - Livia Guadalupi
- Department of Systems Medicine, Tor Vergata University, Rome, Italy
- Synaptic Immunopathology Lab, IRCCS San Raffaele Roma, Rome, Italy
| | - Georgia Mandolesi
- Synaptic Immunopathology Lab, IRCCS San Raffaele Roma, Rome, Italy
- Department of Human Sciences and Quality of Life Promotion, University of Rome San Raffaele, Rome, Italy
| | | | - Diego Centonze
- Neurology Unit, IRCSS Neuromed, Pozzilli, Italy
- Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | | |
Collapse
|
44
|
Riboni-Verri G, Chen BS, McMurran CE, Halliwell GJ, Brown JWL, Coles AJ, Cunniffe NG. Visual outcome measures in clinical trials of remyelinating drugs. BMJ Neurol Open 2024; 6:e000560. [PMID: 38389586 PMCID: PMC10882304 DOI: 10.1136/bmjno-2023-000560] [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: 10/16/2023] [Accepted: 01/15/2024] [Indexed: 02/24/2024] Open
Abstract
One of the most promising approaches to delay, prevent or reverse disability progression in multiple sclerosis (MS) is to enhance endogenous remyelination and limit axonal degeneration. In clinical trials of remyelinating drugs, there is a need for reliable, sensitive and clinically relevant outcome measures. The visual pathway, which is frequently affected by MS, provides a unique model system to evaluate remyelination of acute and chronic MS lesions in vivo and non-invasively. In this review, we discuss the different measures that have been used and scrutinise visual outcome measure selection in current and future remyelination trials.
Collapse
Affiliation(s)
- Gioia Riboni-Verri
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Cambridge Clinical Vision Laboratory, University of Cambridge, Cambridge, UK
| | - Benson S Chen
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Cambridge Clinical Vision Laboratory, University of Cambridge, Cambridge, UK
| | - Christopher E McMurran
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Cambridge Clinical Vision Laboratory, University of Cambridge, Cambridge, UK
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Gregory J Halliwell
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - J William L Brown
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Clinical Outcomes Research Unit (CORe), University of Melbourne, Melborune, Melborune, Australia
| | - Alasdair J Coles
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Cambridge Clinical Vision Laboratory, University of Cambridge, Cambridge, UK
| | - Nick G Cunniffe
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Cambridge Clinical Vision Laboratory, University of Cambridge, Cambridge, UK
| |
Collapse
|
45
|
Shemirani F, Pingel WR, Titcomb TJ, Salari-Moghaddam A, Arsalandeh F, Saxby SM, Snetselaar LG, Wahls TL. The effect of dietary interventions on inflammatory biomarkers among people with multiple sclerosis: A protocol for systematic review and meta-analysis of randomized controlled trials. PLoS One 2024; 19:e0297510. [PMID: 38324543 PMCID: PMC10849228 DOI: 10.1371/journal.pone.0297510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 12/30/2023] [Indexed: 02/09/2024] Open
Abstract
BACKGROUND Multiple sclerosis (MS) is a chronic neuroinflammatory disease of the central nervous system, characterized by demyelination and neurodegeneration, which has a profound impact on the quality of life. Dysregulated inflammatory processes are a major driver of MS progression, with many areas of research being dedicated to modulating inflammation in people with MS. Several dietary patterns have been associated with improvements in inflammatory biomarkers; although, the findings have been inconsistent. Thus, this study aims to evaluate the effects of dietary interventions on inflammatory markers in adults with MS. METHODS Electronic databases, including PubMed/MEDLINE, Web of Science, Scopus, and Cochrane/Central, will be searched. Screening, selection, and extraction of data, along with quality assessment of included studies, will be done by two separate reviewers, and any potential conflicts will be settled through discussion. Two reviewers will independently assess the risk of bias in included studies using the Cochrane Risk of Bias Tool. If plausible, the results will be synthesized and pooled for meta-analysis. The overall quality of evidence of each study will be evaluated using the NutriGRADE tool, which is a modification to the Grading Recommendations Assessment, Development, and Evaluation (GRADE) developed specifically for nutrition research. DISCUSSION Studies have demonstrated conflicting results regarding the effects of dietary interventions on serum levels of inflammatory biomarkers among people with MS. Thus, it is expected that the planned systematic review and meta-analysis will yield robust evidence on the effects of diet on inflammatory profile in the setting of MS.
Collapse
Affiliation(s)
- Farnoosh Shemirani
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | - Wade R. Pingel
- University of Iowa Carver College of Medicine, Iowa City, Iowa, United States of America
| | - Tyler J. Titcomb
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | | | - Farshad Arsalandeh
- Department of Neuroscience, Iran University of Medical Sciences, Tehran, Iran
| | - Solange M. Saxby
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, United States of America
- Community Family Medicine, Dartmouth Health, Lebanon, New Hampshire, United States of America
| | - Linda G. Snetselaar
- Department of Epidemiology, University of Iowa, Iowa City, Iowa, United States of America
| | - Terry L. Wahls
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, United States of America
| |
Collapse
|
46
|
Hermann DM, Peruzzotti-Jametti L, Giebel B, Pluchino S. Extracellular vesicles set the stage for brain plasticity and recovery by multimodal signalling. Brain 2024; 147:372-389. [PMID: 37768167 PMCID: PMC10834259 DOI: 10.1093/brain/awad332] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/07/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Extracellular vesicles (EVs) are extremely versatile naturally occurring membrane particles that convey complex signals between cells. EVs of different cellular sources are capable of inducing striking therapeutic responses in neurological disease models. Differently from pharmacological compounds that act by modulating defined signalling pathways, EV-based therapeutics possess multiple abilities via a variety of effectors, thus allowing the modulation of complex disease processes that may have very potent effects on brain tissue recovery. When applied in vivo in experimental models of neurological diseases, EV-based therapeutics have revealed remarkable effects on immune responses, cell metabolism and neuronal plasticity. This multimodal modulation of neuroimmune networks by EVs profoundly influences disease processes in a highly synergistic and context-dependent way. Ultimately, the EV-mediated restoration of cellular functions helps to set the stage for neurological recovery. With this review we first outline the current understanding of the mechanisms of action of EVs, describing how EVs released from various cellular sources identify their cellular targets and convey signals to recipient cells. Then, mechanisms of action applicable to key neurological conditions such as stroke, multiple sclerosis and neurodegenerative diseases are presented. Pathways that deserve attention in specific disease contexts are discussed. We subsequently showcase considerations about EV biodistribution and delineate genetic engineering strategies aiming at enhancing brain uptake and signalling. By sketching a broad view of EV-orchestrated brain plasticity and recovery, we finally define possible future clinical EV applications and propose necessary information to be provided ahead of clinical trials. Our goal is to provide a steppingstone that can be used to critically discuss EVs as next generation therapeutics for brain diseases.
Collapse
Affiliation(s)
- Dirk M Hermann
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, D-45122 Essen, Germany
| | - Luca Peruzzotti-Jametti
- Department of Clinical Neurosciences and National Institute for Health Research (NIHR) Biomedical Research Centre, University of Cambridge, Cambridge CB2 0AH, UK
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London W12 0NN, UK
| | - Bernd Giebel
- Institute of Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, D-45147 Essen, Germany
| | - Stefano Pluchino
- Department of Clinical Neurosciences and National Institute for Health Research (NIHR) Biomedical Research Centre, University of Cambridge, Cambridge CB2 0AH, UK
| |
Collapse
|
47
|
Berglund R, Cheng Y, Piket E, Adzemovic MZ, Zeitelhofer M, Olsson T, Guerreiro-Cacais AO, Jagodic M. The aging mouse CNS is protected by an autophagy-dependent microglia population promoted by IL-34. Nat Commun 2024; 15:383. [PMID: 38195627 PMCID: PMC10776874 DOI: 10.1038/s41467-023-44556-6] [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: 02/08/2023] [Accepted: 12/15/2023] [Indexed: 01/11/2024] Open
Abstract
Microglia harness an unutilized health-promoting potential in age-related neurodegenerative and neuroinflammatory diseases, conditions like progressive multiple sclerosis (MS). Our research unveils an microglia population emerging in the cortical brain regions of aging mice, marked by ERK1/2, Akt, and AMPK phosphorylation patterns and a transcriptome indicative of activated autophagy - a process critical for cellular adaptability. By deleting the core autophagy gene Ulk1 in microglia, we reduce this population in the central nervous system of aged mice. Notably, this population is found dependent on IL-34, rather than CSF1, although both are ligands for CSF1R. When aging mice are exposed to autoimmune neuroinflammation, the loss of autophagy-dependent microglia leads to neural and glial cell death and increased mortality. Conversely, microglial expansion mediated by IL-34 exhibits a protective effect. These findings shed light on an autophagy-dependent neuroprotective microglia population as a potential target for treating age-related neuroinflammatory conditions, including progressive MS.
Collapse
Affiliation(s)
- Rasmus Berglund
- Department of Clinical Neuroscience, Division of Neuro, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 171 76, Stockholm, Sweden.
| | - Yufei Cheng
- Department of Clinical Neuroscience, Division of Neuro, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 171 76, Stockholm, Sweden
| | - Eliane Piket
- Department of Clinical Neuroscience, Division of Neuro, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 171 76, Stockholm, Sweden
| | - Milena Z Adzemovic
- Department of Clinical Neuroscience, Division of Neuro, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 171 76, Stockholm, Sweden
| | - Manuel Zeitelhofer
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institutet, 171 65, Solna, Sweden
| | - Tomas Olsson
- Department of Clinical Neuroscience, Division of Neuro, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 171 76, Stockholm, Sweden
| | - Andre Ortlieb Guerreiro-Cacais
- Department of Clinical Neuroscience, Division of Neuro, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 171 76, Stockholm, Sweden
| | - Maja Jagodic
- Department of Clinical Neuroscience, Division of Neuro, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 171 76, Stockholm, Sweden
| |
Collapse
|
48
|
Salapa HE, Thibault PA, Libner CD, Ding Y, Clarke JPWE, Denomy C, Hutchinson C, Abidullah HM, Austin Hammond S, Pastushok L, Vizeacoumar FS, Levin MC. hnRNP A1 dysfunction alters RNA splicing and drives neurodegeneration in multiple sclerosis (MS). Nat Commun 2024; 15:356. [PMID: 38191621 PMCID: PMC10774274 DOI: 10.1038/s41467-023-44658-1] [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: 06/23/2022] [Accepted: 12/22/2023] [Indexed: 01/10/2024] Open
Abstract
Neurodegeneration is the primary driver of disease progression in multiple sclerosis (MS) resulting in permanent disability, creating an urgent need to discover its underlying mechanisms. Herein, we establish that dysfunction of the RNA binding protein heterogeneous nuclear ribonucleoprotein A1 (hnRNP A1) results in differential of binding to RNA targets causing alternative RNA splicing, which contributes to neurodegeneration in MS and its models. Using RNAseq of MS brains, we discovered differential expression and aberrant splicing of hnRNP A1 target RNAs involved in neuronal function and RNA homeostasis. We confirmed this in vivo in experimental autoimmune encephalomyelitis employing CLIPseq specific for hnRNP A1, where hnRNP A1 differentially binds and regulates RNA, including aberrantly spliced targets identified in human samples. Additionally, dysfunctional hnRNP A1 expression in neurons caused neurite loss and identical changes in splicing, corroborating hnRNP A1 dysfunction as a cause of neurodegeneration. Collectively, these data indicate hnRNP A1 dysfunction causes altered neuronal RNA splicing, resulting in neurodegeneration in MS.
Collapse
Affiliation(s)
- Hannah E Salapa
- Office of the Saskatchewan Multiple Sclerosis Clinical Research Chair, University of Saskatchewan, Saskatoon, SK, S7K 0M7, Canada
- Cameco MS Neuroscience Research Centre, College of Medicine, University of Saskatchewan, Saskatoon, SK, S7K 0M7, Canada
- Neurology Division, Department of Medicine, University of Saskatchewan, Saskatoon, SK, S7N 0X8, Canada
| | - Patricia A Thibault
- Office of the Saskatchewan Multiple Sclerosis Clinical Research Chair, University of Saskatchewan, Saskatoon, SK, S7K 0M7, Canada
- Cameco MS Neuroscience Research Centre, College of Medicine, University of Saskatchewan, Saskatoon, SK, S7K 0M7, Canada
- Neurology Division, Department of Medicine, University of Saskatchewan, Saskatoon, SK, S7N 0X8, Canada
| | - Cole D Libner
- Office of the Saskatchewan Multiple Sclerosis Clinical Research Chair, University of Saskatchewan, Saskatoon, SK, S7K 0M7, Canada
- Cameco MS Neuroscience Research Centre, College of Medicine, University of Saskatchewan, Saskatoon, SK, S7K 0M7, Canada
- Department of Health Sciences, College of Medicine, University of Saskatchewan, Saskatoon, SK, S7N 5E5, Canada
| | - Yulian Ding
- Division of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, SK, S7N 5E5, Canada
- Division of Biomedical Engineering, College of Medicine, University of Saskatchewan, Saskatoon, SK, S7N 5A9, Canada
| | - Joseph-Patrick W E Clarke
- Office of the Saskatchewan Multiple Sclerosis Clinical Research Chair, University of Saskatchewan, Saskatoon, SK, S7K 0M7, Canada
- Cameco MS Neuroscience Research Centre, College of Medicine, University of Saskatchewan, Saskatoon, SK, S7K 0M7, Canada
- Neurology Division, Department of Medicine, University of Saskatchewan, Saskatoon, SK, S7N 0X8, Canada
| | - Connor Denomy
- Division of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, SK, S7N 5E5, Canada
| | - Catherine Hutchinson
- Office of the Saskatchewan Multiple Sclerosis Clinical Research Chair, University of Saskatchewan, Saskatoon, SK, S7K 0M7, Canada
- Cameco MS Neuroscience Research Centre, College of Medicine, University of Saskatchewan, Saskatoon, SK, S7K 0M7, Canada
- Neurology Division, Department of Medicine, University of Saskatchewan, Saskatoon, SK, S7N 0X8, Canada
| | - Hashim M Abidullah
- Office of the Saskatchewan Multiple Sclerosis Clinical Research Chair, University of Saskatchewan, Saskatoon, SK, S7K 0M7, Canada
- Cameco MS Neuroscience Research Centre, College of Medicine, University of Saskatchewan, Saskatoon, SK, S7K 0M7, Canada
- Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK, S7N 5E5, Canada
| | - S Austin Hammond
- Next-Generation Sequencing Facility, University of Saskatchewan, Saskatoon, SK, S7N 5E5, Canada
| | - Landon Pastushok
- Advanced Diagnostics Research Laboratory, Department of Pathology and Lab Medicine, College of Medicine, University of Saskatchewan, Saskatoon, SK, S7N 5E5, Canada
| | - Frederick S Vizeacoumar
- Department of Pathology and Laboratory Medicine, University of Saskatchewan, Saskatoon, SK, S7N 5E5, Canada
| | - Michael C Levin
- Office of the Saskatchewan Multiple Sclerosis Clinical Research Chair, University of Saskatchewan, Saskatoon, SK, S7K 0M7, Canada.
- Cameco MS Neuroscience Research Centre, College of Medicine, University of Saskatchewan, Saskatoon, SK, S7K 0M7, Canada.
- Neurology Division, Department of Medicine, University of Saskatchewan, Saskatoon, SK, S7N 0X8, Canada.
- Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK, S7N 5E5, Canada.
| |
Collapse
|
49
|
Kartau M, Kartau J, Pohja M, Verkkoniemi‐Ahola A. Plasma antioxidant potential measured by total radical trapping antioxidant parameter in a cohort of multiple sclerosis patients. Brain Behav 2024; 14:e3377. [PMID: 38376020 PMCID: PMC10794128 DOI: 10.1002/brb3.3377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 12/12/2023] [Accepted: 12/16/2023] [Indexed: 02/21/2024] Open
Abstract
BACKGROUND Oxidative injury has been implicated as a mediator of demyelination, axonal damage, and neurodegeneration in multiple sclerosis (MS). There is a high demand for oxidative injury biomarkers. The aim of the study was to evaluate MS patients' plasma antioxidant potential using the total radical trapping parameter (TRAP) assay and examine its usefulness as an MS disease biomarker. METHODS A total number of 112 MS patients underwent an analysis of TRAP. In addition, plasma uric acid (UA) levels were analyzed. The neurological and radiological data were collected from patient records from Helsinki University Hospital during 2012-2013 when first-line injectables of moderate-efficacy, natalizumab (NTZ), and fingolimod (FTY) of high efficacy disease modifying therapies and in some cases azathioprine (AZT) were used to treat MS. RESULTS TRAP values were negatively associated with expanded disability status scale (EDSS) score with p-value .052, β = -28. There was also a negative association in TRAP values between patients with no medication (n = 22, TRAP mean 1255 μmol/L (95% confidence interval [CI] 1136-1374)) and patients who received NTZ, p-value .020 (n = 19, TRAP mean was 991 μmol/L (95% CI 849-1133) or FTY treatment, p-value .030 (n = 5, TRAP mean 982 μmol/L (95% CI 55-1909). Due to a small sample size, these results were not significant after applying a false discovery rate correction at a 0.05 significance level but are worth highlighting. Men in the study had higher TRAP values, p-value = .001 (TRAP mean 1320 ± 293 μmol/L) than women (TRAP mean 1082 ± 288 μmol/L). UA was positively associated with TRAP values, p-value <.001 and UA levels in men (UA mean 334.5 ± 62.6 μmol/L) were higher compared to women (UA mean 240 ± 55.8 μmol/L), t-test p-value <.001. The significant difference in TRAP levels between genders, with men showing higher TRAP values than women, may be attributed to the variation in UA levels. CONCLUSION Our findings suggest that lower plasma antioxidant potential is linked to more severe disability measured by EDSS scores. Patients treated with NTZ and FTY had reduced antioxidant power, which might be influenced by the active MS disease rather than the treatments themselves. The study reveals a strong positive correlation between UA levels and TRAP, particularly among women. However, men on average had better antioxidant potential than women. Neither the disease type nor the duration influences TRAP levels. While serving as a marker of antioxidant potential, plasma TRAP in MS patients does not reliably reflect overall oxidative stress (OS) and should not be solely used as an indicator of OS.
Collapse
Affiliation(s)
- Marge Kartau
- Clinical Neurosciences, NeurologyHelsinki University Hospital and Helsinki UniversityHelsinkiFinland
| | - Joonas Kartau
- Department of Mathematics and StatisticsUniversity of HelsinkiHelsinkiFinland
| | - Marjatta Pohja
- Clinical Neurosciences, NeurologyHelsinki University HospitalHelsinkiFinland
| | | |
Collapse
|
50
|
Dejbakht M, Akhzari M, Jalili S, Faraji F, Barazesh M. Multiple Sclerosis: New Insights into Molecular Pathogenesis and Novel Platforms for Disease Treatment. Curr Drug Res Rev 2024; 16:175-197. [PMID: 37724675 DOI: 10.2174/2589977516666230915103730] [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: 01/08/2023] [Revised: 06/23/2023] [Accepted: 07/03/2023] [Indexed: 09/21/2023]
Abstract
BACKGROUND Multiple sclerosis (MS), a chronic inflammatory disorder, affects the central nervous system via myelin degradation. The cause of MS is not fully known, but during recent years, our knowledge has deepened significantly regarding the different aspects of MS, including etiology, molecular pathophysiology, diagnosis and therapeutic options. Myelin basic protein (MBP) is the main myelin protein that accounts for maintaining the stability of the myelin sheath. Recent evidence has revealed that MBP citrullination or deamination, which is catalyzed by Ca2+ dependent peptidyl arginine deiminase (PAD) enzyme leads to the reduction of positive charge, and subsequently proteolytic cleavage of MBP. The overexpression of PAD2 in the brains of MS patients plays an essential role in new epitope formation and progression of the autoimmune disorder. Some drugs have recently entered phase III clinical trials with promising efficacy and will probably obtain approval in the near future. As different therapeutic platforms develop, finding an optimal treatment for each individual patient will be more challenging. AIMS This review provides a comprehensive insight into MS with a focus on its pathogenesis and recent advances in diagnostic methods and its present and upcoming treatment modalities. CONCLUSION MS therapy alters quickly as research findings and therapeutic options surrounding MS expand. McDonald's guidelines have created different criteria for MS diagnosis. In recent years, ever-growing interest in the development of PAD inhibitors has led to the generation of many reversible and irreversible PAD inhibitors against the disease with satisfactory therapeutic outcomes.
Collapse
Affiliation(s)
- Majid Dejbakht
- Department of Cellular and Molecular Research Center, Gerash University of Medical Sciences, Gerash, Iran
| | - Morteza Akhzari
- School of Nursing, Larestan University of Medical Sciences, Larestan, Iran
| | - Sajad Jalili
- Department of Orthopedics, School of Medicine, Ahvaz Jundishapour University of Medical Sciences, Ahvaz, Iran
| | - Fouziyeh Faraji
- Department of Cellular and Molecular Research Center, Gerash University of Medical Sciences, Gerash, Iran
| | - Mahdi Barazesh
- Department of Biotechnology, Cellular and Molecular Research Center, School of Paramedical, Gerash University of Medical Sciences, Gerash, Iran
| |
Collapse
|