1
|
Jallouli S, Ghroubi S, Damak M, Sakka S, Elleuch MH, Mhiri C, Yahia A, Driss T, de Marco G, Hammouda O. 12-week melatonin supplementation improved dynamic postural stability and walking performance in persons living with multiple sclerosis: A randomized controlled trial. Behav Brain Res 2025; 476:115191. [PMID: 39122092 DOI: 10.1016/j.bbr.2024.115191] [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: 06/01/2024] [Revised: 07/27/2024] [Accepted: 08/06/2024] [Indexed: 08/12/2024]
Abstract
BACKGROUND Persons with multiple sclerosis (PwMS) suffer from sleep disturbances, fatigue and pain, which can be due, at least in part, to decreased levels of endogenous melatonin. These alterations could exacerbate postural instability, gait disorders and fall risk. Acute effects of exogenous melatonin on physical disorders have been studied in PwMS but its long-term effects on these parameters have not been explored yet in this population. This study aimed to determine the impact of chronic melatonin intake on dynamic postural stability, walking performance and fall risk in PwMS. METHODS This randomized placebo-controlled study included 27 PwMS who were assigned to either melatonin group (MG, n=15) or placebo group (PG, n=12) (3 mg/night for 12 weeks). Dynamic postural balance (force platform), walking performance (locometer) and fall risk (Four Square Step Test) were evaluated pre (T0)- and post (T1)-intervention. Sleep quality (Pittsburgh Sleep Quality Index (PSQI)), fatigue perception (Fatigue Severity Scale (FSS)), neuropathic pain (Neuropathic Pain Questionnaire 4 (DN4)) and quality of life (International Multiple Sclerosis (MS) Quality of Life Questionnaire) were also assessed at T0 and T1. RESULTS The center of pressure mean velocity decreased in MG compared with PG in the frontal plane (22.98 %, p=0.028). Stride length and walking speed increased in MG comparatively with PG (18.09 %, p=0.036; 9.65 %, p=0.025, respectively). The PSQI (55.89 %, p<0.001), FSS (32.38 %, p=0.003) and DN4 (32.41 %, p=0.035) scores decreased in MG compared with PG. CONCLUSION 12-week melatonin supplementation can be recommended for managing MS-related gait disorders and dynamic postural imbalance. This therapy may also be prescribed for PwMS due to its anti-fatigue and analgesic effects as well as its benefits on sleep quality. CLINICAL REGISTRATION This study was prospectively recorded in the Pan African Clinical Trial Registry database (PACTR202007465309582) (https://pactr.samrc.ac.za/.).
Collapse
Affiliation(s)
- Sonda Jallouli
- Research laboratory: Evaluation and Management of Musculoskeletal System Pathologies, LR20ES09, Faculty of Medicine, University of Sfax, Tunisia; High Institute of Sport and Physical Education of Sfax, University of Sfax, Sfax, Tunisia.
| | - Sameh Ghroubi
- Research laboratory: Evaluation and Management of Musculoskeletal System Pathologies, LR20ES09, Faculty of Medicine, University of Sfax, Tunisia
| | - Mariem Damak
- Department of Neurology, Habib Bourguiba University Hospital, Clinical Investigation Center, Faculty of Medicine, University of Sfax, Sfax, Tunisia; Laboratory of Neurogenetics, Parkinson's Disease and Cerebrovascular Disease, LR12SP19, Habib Bourguiba University Hospital, University of Sfax, Tunisia
| | - Salma Sakka
- Department of Neurology, Habib Bourguiba University Hospital, Clinical Investigation Center, Faculty of Medicine, University of Sfax, Sfax, Tunisia; Laboratory of Neurogenetics, Parkinson's Disease and Cerebrovascular Disease, LR12SP19, Habib Bourguiba University Hospital, University of Sfax, Tunisia
| | - Mohamed Habib Elleuch
- Research laboratory: Evaluation and Management of Musculoskeletal System Pathologies, LR20ES09, Faculty of Medicine, University of Sfax, Tunisia
| | - Chokri Mhiri
- Department of Neurology, Habib Bourguiba University Hospital, Clinical Investigation Center, Faculty of Medicine, University of Sfax, Sfax, Tunisia; Laboratory of Neurogenetics, Parkinson's Disease and Cerebrovascular Disease, LR12SP19, Habib Bourguiba University Hospital, University of Sfax, Tunisia
| | - Abdelmoneem Yahia
- Research laboratory: Evaluation and Management of Musculoskeletal System Pathologies, LR20ES09, Faculty of Medicine, University of Sfax, Tunisia
| | - Tarak Driss
- LINP2, UFR STAPS, University of Paris Nanterre, Nanterre, France
| | | | - Omar Hammouda
- LINP2, UFR STAPS, University of Paris Nanterre, Nanterre, France; Research Laboratory, Molecular bases of Human Pathology, LR19ES13, Faculty of medicine of Sfax, University of Sfax, Tunisia
| |
Collapse
|
2
|
Jingjing H, Tongqian W, Shirong Y, Lan M, Jing L, Shihui M, Haijian Y, Fang Y. S100A4 promotes experimental autoimmune encephalomyelitis by impacting microglial inflammation through TLR4/NF-κB signaling pathway. Int Immunopharmacol 2024; 142:112849. [PMID: 39241524 DOI: 10.1016/j.intimp.2024.112849] [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: 01/14/2024] [Revised: 06/30/2024] [Accepted: 07/29/2024] [Indexed: 09/09/2024]
Abstract
Multiple sclerosis (MS) is a neurodegenerating autoimmune disease with no clinical cure currently. The calcium-binding protein S100A4 has been demonstrated to exert regulatory roles in inflammatory disorders including MS. However, the precise mechanisms by which S100A4 regulates neuroinflammation in MS remains unknown. To investigate the regulatory effect of S100A4 on microglial inflammation and its impact on neuroinflammation, the mouse-derived microglia cell line BV2 cells were infected with lentivirus to knockout S100A4 for in vitro studies. Wild-type (WT) and S100A4-/- mice were induced to develop experimental autoimmune encephalomyelitis (EAE), an animal model of MS, for in vivo investigation. Results indicated that the frequencies of microglia in the spinal cord and brain and the expression of S100A4 in these tissues varied kinetically along with the progression of the disease in mice with EAE. S100A4-/- mice presented ameliorated clinical scores of EAE and exhibited less severe EAE signs, including inflammatory cell infiltration in the spinal cord and brain and demyelination of the spinal cord. Moreover, these mice demonstrated overall reduced levels of inflammatory cytokines in the spinal cord and brain. Compromised systematic inflammatory responses including circulating cytokines and frequencies of immune cells in the spleen were also observed in these mice. In addition, both exogenous and endogenous S100A4 could promote the microglial inflammation, affect the polarization of microglia and enhance inflamed microglia-mediated apoptosis of neuronal cells through TLR4/NF-κB signaling pathway. Thus, S100A4 may participate in the regulation of neuroinflammation at least partly through regulating the inflammation of microglia.
Collapse
Affiliation(s)
- He Jingjing
- Center for Clinical Laboratories, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China; Department of Clinical Laboratory, Guizhou Hospital, the First Affiliated Hospital of Sun Yat-sen University, Guiyang 550004, China
| | - Wu Tongqian
- Clinical Research Center, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Yan Shirong
- Center for Clinical Laboratories, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China; School for Laboratory Science, Guizhou Medical University, Guiyang 550004, China
| | - Ma Lan
- School for Laboratory Science, Guizhou Medical University, Guiyang 550004, China
| | - Li Jing
- Center for Clinical Laboratories, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China; School for Laboratory Science, Guizhou Medical University, Guiyang 550004, China
| | - Mo Shihui
- School for Laboratory Science, Guizhou Medical University, Guiyang 550004, China
| | - Yan Haijian
- Department of Emergency, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Yu Fang
- Center for Clinical Laboratories, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China; School for Laboratory Science, Guizhou Medical University, Guiyang 550004, China.
| |
Collapse
|
3
|
Carnero Contentti E, Alonso R, Silva B, Burgos M, Tavolini D, Lopez P, Cristiano E, Patrucco L, Tkachuk V, Mainella C, Zanga G, Leguizamón F, Luetic G, Silva E, Tizio S, Alonso Serena M, Rojas JI. Clinical effectiveness of dimethyl fumarate in multiple sclerosis patients from Argentina. Neurol Sci 2024; 45:5833-5840. [PMID: 39080156 DOI: 10.1007/s10072-024-07712-4] [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] [Received: 02/17/2024] [Accepted: 07/22/2024] [Indexed: 11/12/2024]
Abstract
BACKGROUND We assessed the effectiveness, safety and patient-reported outcomes (PROs) of dimethyl fumarate (DMF) in real-world clinical practice in patients with multiple sclerosis (PwMS) from Argentina. METHODS We conducted a multicenter ambispective cohort study in Argentina between September 2020 and March 2023. Changes in annualized relapse rate (ARR), Expanded Disability Status Scale (EDSS) score, magnetic resonance imaging (MRI), no evidence of disease activity (NEDA), PROs (depression, anxiety, fatigue, burden of treatment and quality of life), and safety data were collected at clinical visits performed every 6 months for at least 24 months. RESULTS We included 161 PwMS (64% female). DMF treatment was associated with a significant reduction in ARR from baseline after 24 months of treatment (from 0.87 to 0.23, p < 0.001). Disability progression was observed in 27.9% vs. 9.3% pre- and post-DMF, and disability improvement was found in 13% of patients from baseline to month 24. MRI activity was significantly reduced compared with baseline. Fatigue, depression, and quality of life scores were significantly improved from baseline to 24 months. Flushing was the most frequent adverse event reported in 19.2%. No significant reduction was observed in the hospitalization rate pre- and post-DMF (19.8% vs. 5.6%, p = 0.32). During follow-up, 135 (83%) patients were relapse-free, 110 (68.3%) were MRI free activity (Gad + lesion) and 108 (67%) reached NEDA. CONCLUSIONS DMF significantly reduced disease activity in PwMS from Argentina with a good safety profile in real-world settings. A significant impact on the quality of life during follow-up was found.
Collapse
Affiliation(s)
- Edgar Carnero Contentti
- Department of Neurosciences, Neuroimmunology Unit, Hospital Aleman, Buenos Aires, Argentina.
- CenRos, Neuroimmunology Clinic, INECO Neurociencias Oroño, Rosario, Santa Fe, Argentina.
| | - Ricardo Alonso
- Sanatorio Güemes, Buenos Aires, Argentina
- Hospital Ramos Mejía, Centro Universitario de Esclerosis Múltiple, Buenos Aires, Argentina
| | - Berenice Silva
- Hospital Ramos Mejía, Centro Universitario de Esclerosis Múltiple, Buenos Aires, Argentina
- Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Marcos Burgos
- Servicio de Neurología, Hospital San Bernardo, Salta, Argentina
| | - Darío Tavolini
- CenRos, Neuroimmunology Clinic, INECO Neurociencias Oroño, Rosario, Santa Fe, Argentina
| | - Pablo Lopez
- Department of Neurosciences, Neuroimmunology Unit, Hospital Aleman, Buenos Aires, Argentina
| | - Edgardo Cristiano
- Centro de esclerosis múltiple de Buenos Aires, Buenos Aires, Argentina
| | - Liliana Patrucco
- Centro de esclerosis múltiple de Buenos Aires, Buenos Aires, Argentina
| | - Verónica Tkachuk
- Sección de Neuroinmunología y Enfermedades Desmielinizantes, Servicio de Neurología, Hospital de Clínicas José de San Martín, Buenos Aires, Argentina
| | - Carolina Mainella
- CenRos, Neuroimmunology Clinic, INECO Neurociencias Oroño, Rosario, Santa Fe, Argentina
- Hospital Español de Rosario, Rosario, Santa Fe, Argentina
| | | | | | - Geraldine Luetic
- Instituto de Neurociencias de Rosario, San Lorenzo, Rosario, Santa Fé, Argentina
| | - Emanuel Silva
- Neurología Clínica, Posadas, Misiones. Predigma, Argentina
| | - Santiago Tizio
- Hospital Español de La Plata, La Plata, Buenos Aires, Argentina
| | | | | |
Collapse
|
4
|
Silva BA, Heriz A, Ayerbe J, Lázaro L, Casas M, López P, Tkachuk V, Balbuena ME, Nadur D, Liwacki S, Luetic G, Burgos M, Casales F, Piedrabuena A, Carnero Contentti E, Zárate A, Zanga G, Steinberg J, Mainella C, Tavolini D, Hryb J, Leguizamón F, Pagani Cassará F, José G, Carrizo P, Nofal P, Luis B, Pita C, Míguez J, Alonso R. Cladribine use trend in Latin America: the changes in patient profile impact in the drug effectiveness. Neurol Sci 2024; 45:5841-5848. [PMID: 39259243 DOI: 10.1007/s10072-024-07763-7] [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: 01/29/2024] [Accepted: 09/03/2024] [Indexed: 09/12/2024]
Abstract
INTRODUCTION Cladribine was approved for Multiple Sclerosis (MS) in our country in 2018. A previous study by our group showed that its use among high efficacy therapies options has been increasing along the years. OBJECTIVE to analyze the cladribine use trend across time since its approval. METHOD A retrospective cohort study was performed. People with MS (pwMS) treated with cladribine were included. Two periods were defined: P1 = 2018 - 2020 and P2 = 2021 - 2023. A comparative analysis was carry out between P1 and P2 to assess the trend of use, clinical/demographic characteristics, and effectiveness. RESULTS One hundred ninety- seven people with MS (pwMS) were included, mean EDSS: 2.2 ± 3.08, 72.6% female, mean age: 35.2 ± 9 years, mean disease duration: 6.6 ± 5.6 years, mean time lapse under cladribine: 26.1 ± 12.4 months. Regarding patient profile, we found significant differences between P1 and P2 in the MS evolution (p = 0.001) and EDSS ( p = 0.018) prior to initiation of cladribine. In the individualized analysis by year, we found a decrease in relapse number in the year prior to starting cladribine (p = 0.02). A higher proportion of No Evidence of Disease Activity (NEDA) was found in patients treated at P2 compared to those treated at P1 (p < 0.001). CONCLUSION An earlier use of cladribine achieved a significant increase in reaching NEDA. This learning curve in the use of cladribine allows a better identification of the candidate patient and influences the treatment effectiveness.
Collapse
Affiliation(s)
- Berenice A Silva
- Hospital Italiano de Buenos Aires, Sección Enfermedades Desmielinizantes, Buenos Aires, Argentina.
- Hospital Ramos Mejía, CABA, Hospital Italiano de Buenos Aires and Centro Universitario de Esclerosis Múltiple, Buenos Aires, Argentina.
| | - Alejandra Heriz
- Hospital Italiano de Buenos Aires, Sección Enfermedades Desmielinizantes, Buenos Aires, Argentina
| | - Jeremías Ayerbe
- Hospital Italiano de Buenos Aires, Sección Enfermedades Desmielinizantes, Buenos Aires, Argentina
| | - Luciana Lázaro
- Hospital Ramos Mejía, CABA, Hospital Italiano de Buenos Aires and Centro Universitario de Esclerosis Múltiple, Buenos Aires, Argentina
| | - Magdalena Casas
- Hospital Ramos Mejía, CABA, Hospital Italiano de Buenos Aires and Centro Universitario de Esclerosis Múltiple, Buenos Aires, Argentina
| | - Pablo López
- Hospital Alemán, Unidad de Neuroinmunología, Buenos Aires, Argentina
| | - Verónica Tkachuk
- Hospital de Clínicas José de San Martín, Clínica de Enfermedades Desmielinizantes, Buenos Aires, Argentina
| | - María Eugenia Balbuena
- Hospital de Clínicas José de San Martín, Clínica de Enfermedades Desmielinizantes, Buenos Aires, Argentina
| | - Débora Nadur
- Hospital de Clínicas José de San Martín, Clínica de Enfermedades Desmielinizantes, Buenos Aires, Argentina
| | - Susana Liwacki
- Hospital de Córdoba, Servicio de Neurología, Córdoba, Argentina
- Clínica Universitaria Reina Fabiola Servicio de Neurología, Córdoba, Argentina
| | | | | | - Federico Casales
- Sanatorio de Los Arcos, Servicio de Neurología, Buenos Aires, Argentina
| | | | | | | | | | - Judith Steinberg
- Hospital Británico, Servicio de Neurología, Buenos Aires, Argentina
| | | | | | - Javier Hryb
- Hospital Durand, Consultorio de Neuroinmunología Clínica, Buenos Aires, Argentina
| | - Felisa Leguizamón
- Hospital Álvarez, Buenos Aires, Argentina
- Hospital Austral, Buenos Aires, Argentina
| | | | | | | | - Pedro Nofal
- Hospital de Clínicas Nuestra Señora del Carmen, San Miguel de Tucumán, Argentina
| | - Belén Luis
- Hospital Güemes, Sección Enfermedades Desmielinizantes, Buenos Aires, Argentina
| | - Cecilia Pita
- Hospital Ramos Mejía, CABA, Hospital Italiano de Buenos Aires and Centro Universitario de Esclerosis Múltiple, Buenos Aires, Argentina
- INEBA, Buenos Aires, Argentina
| | - Jimena Míguez
- Hospital Italiano de Buenos Aires, Sección Enfermedades Desmielinizantes, Buenos Aires, Argentina
| | - Ricardo Alonso
- Hospital Ramos Mejía, CABA, Hospital Italiano de Buenos Aires and Centro Universitario de Esclerosis Múltiple, Buenos Aires, Argentina
- Hospital de Clínicas Nuestra Señora del Carmen, San Miguel de Tucumán, Argentina
| |
Collapse
|
5
|
Deng Y, Wang F, Wang T, Zhang X, Chen D, Wang Y, Chen C, Pan G. Research progress in the mechanisms and functions of specialized pro-resolving mediators in neurological diseases. Prostaglandins Other Lipid Mediat 2024; 175:106905. [PMID: 39265777 DOI: 10.1016/j.prostaglandins.2024.106905] [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: 07/19/2024] [Revised: 08/30/2024] [Accepted: 09/09/2024] [Indexed: 09/14/2024]
Abstract
The nervous system interacts with the immune system through a variety of cellular regulators, signaling pathways, and molecular mechanisms. Disruptions in these interactions lead to the development of multiple neurological diseases. Recent studies have identified that specialized pro-resolving mediators (SPMs) play a regulatory role in the neuroimmune system. This study reviews recent research on the function of SPMs in the inflammatory process and their association with the nervous system. The review aims to provide new perspectives for studying the pathogenesis of neurological diseases and identify novel targets for clinical therapy.
Collapse
Affiliation(s)
- Yu Deng
- Guangzhou Hospital of Integrated Chinese and Western Medicine Affiliated to Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510800, China
| | - Fei Wang
- Yancheng TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Yancheng, Jiangsu 224000, China; Yancheng TCM Hospital, Yancheng, Jiangsu 224000, China
| | - Tianle Wang
- Guangzhou Hospital of Integrated Chinese and Western Medicine Affiliated to Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510800, China
| | - Xu Zhang
- Guangzhou Hospital of Integrated Chinese and Western Medicine Affiliated to Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510800, China
| | - Du Chen
- Guangzhou Hospital of Integrated Chinese and Western Medicine Affiliated to Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510800, China
| | - Yuhan Wang
- Hubei University of Chinese Medicine, Wuhan, Hubei 430065, China
| | - Chaojun Chen
- Guangzhou Hospital of Integrated Chinese and Western Medicine Affiliated to Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510800, China.
| | - Guangtao Pan
- Yancheng TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Yancheng, Jiangsu 224000, China; Yancheng TCM Hospital, Yancheng, Jiangsu 224000, China.
| |
Collapse
|
6
|
Huang X, Zeng Q, Hu Y, Shi X. A mendelian randomization analysis of the associations between haptoglobin and multiple sclerosis. Neurol Sci 2024; 45:5823-5832. [PMID: 39400787 DOI: 10.1007/s10072-024-07786-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 09/25/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND Observational studies have suggested an association between plasma haptoglobin and multiple sclerosis (MS). Haptoglobin plays an important role in the pathogenesis of MS. However, whether it has a causal effect on MS remains unknown. METHODS We here used a two-sample bidirectional Mendelian randomization (MR) method to investigate the causality between haptoglobin and MS. Genetic variants associated with plasma haptoglobin from two independent genome wide association studies (GWASs) (used as the discovery and replication datasets, respectively) were applied as the exposure. Their causal effects on summary statistics of GWASs of MS and disease severity were evaluated using the inverse-variance weighted (IVW) approach as the main analysis component. RESULTS We found in both discovery and replication dataset that plasma haptoglobin was causally positively associated with the risk of MS (discovery: OR: 1.063, 95% CI: 1.022-1.106, P = 0.002; replication: OR: 1.041, 95% CI: 1.005-1.078, P = 0.026), but it was not associated with MS severity (discovery: OR: 1.017, 95% CI: 0.993-1.042, P = 0.168; replication: OR: 1.011, 95% CI: 0.987-1.036, P = 0.373). Besides, we did not detect any significant results in the reverse causation analysis. CONCLUSIONS Our study provides evidence for the causal effects of plasma haptoglobin on the risk of MS.
Collapse
Affiliation(s)
- Xingxiao Huang
- Geriatric Neuroscience Center, The Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou, China
- School of Mental Health, Guangzhou Medical University, Guangzhou, China
- Institute of Psychiatry and Psychology, Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province, Ministry of Education of China, Guangzhou Medical University, Guangzhou, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Qian Zeng
- School of Mental Health, Guangzhou Medical University, Guangzhou, China
- Institute of Psychiatry and Psychology, Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province, Ministry of Education of China, Guangzhou Medical University, Guangzhou, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
- Department of Neurology, The Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yachun Hu
- School of Mental Health, Guangzhou Medical University, Guangzhou, China
- Institute of Psychiatry and Psychology, Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province, Ministry of Education of China, Guangzhou Medical University, Guangzhou, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
- Department of Neurology, The Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xiaolei Shi
- Geriatric Neuroscience Center, The Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou, China.
- School of Mental Health, Guangzhou Medical University, Guangzhou, China.
- Institute of Psychiatry and Psychology, Guangzhou Medical University, Guangzhou, China.
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province, Ministry of Education of China, Guangzhou Medical University, Guangzhou, China.
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China.
| |
Collapse
|
7
|
Ionescu RB, Nicaise AM, Reisz JA, Williams EC, Prasad P, Willis CM, Simões-Abade MBC, Sbarro L, Dzieciatkowska M, Stephenson D, Suarez Cubero M, Rizzi S, Pirvan L, Peruzzotti-Jametti L, Fossati V, Edenhofer F, Leonardi T, Frezza C, Mohorianu I, D'Alessandro A, Pluchino S. Increased cholesterol synthesis drives neurotoxicity in patient stem cell-derived model of multiple sclerosis. Cell Stem Cell 2024; 31:1574-1590.e11. [PMID: 39437792 DOI: 10.1016/j.stem.2024.09.014] [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: 02/12/2024] [Revised: 08/01/2024] [Accepted: 09/18/2024] [Indexed: 10/25/2024]
Abstract
Senescent neural progenitor cells have been identified in brain lesions of people with progressive multiple sclerosis (PMS). However, their role in disease pathobiology and contribution to the lesion environment remains unclear. By establishing directly induced neural stem/progenitor cell (iNSC) lines from PMS patient fibroblasts, we studied their senescent phenotype in vitro. Senescence was strongly associated with inflammatory signaling, hypermetabolism, and the senescence-associated secretory phenotype (SASP). PMS-derived iNSCs displayed increased glucose-dependent fatty acid and cholesterol synthesis, which resulted in the accumulation of lipid droplets. A 3-hydroxy-3-methylglutaryl (HMG)-coenzyme A (CoA) reductase (HMGCR)-mediated lipogenic state was found to induce a SASP in PMS iNSCs via cholesterol-dependent transcription factors. SASP from PMS iNSC lines induced neurotoxicity in mature neurons, and treatment with the HMGCR inhibitor simvastatin altered the PMS iNSC SASP, promoting cytoprotective qualities and reducing neurotoxicity. Our findings suggest a disease-associated, cholesterol-related, hypermetabolic phenotype of PMS iNSCs that leads to neurotoxic signaling and is rescuable pharmacologically.
Collapse
Affiliation(s)
- Rosana-Bristena Ionescu
- Department of Clinical Neurosciences and NIHR Biomedical Research Centre, University of Cambridge, Cambridge CB2 0AH, UK
| | - Alexandra M Nicaise
- Department of Clinical Neurosciences and NIHR Biomedical Research Centre, University of Cambridge, Cambridge CB2 0AH, UK
| | - Julie A Reisz
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Eleanor C Williams
- Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK
| | - Pranathi Prasad
- Department of Clinical Neurosciences and NIHR Biomedical Research Centre, University of Cambridge, Cambridge CB2 0AH, UK
| | - Cory M Willis
- Department of Clinical Neurosciences and NIHR Biomedical Research Centre, University of Cambridge, Cambridge CB2 0AH, UK
| | - Madalena B C Simões-Abade
- Department of Clinical Neurosciences and NIHR Biomedical Research Centre, University of Cambridge, Cambridge CB2 0AH, UK
| | - Linda Sbarro
- Department of Clinical Neurosciences and NIHR Biomedical Research Centre, University of Cambridge, Cambridge CB2 0AH, UK
| | - Monika Dzieciatkowska
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Daniel Stephenson
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Marta Suarez Cubero
- Genomics, Stem Cell Biology and Regenerative Medicine Group, Institute of Molecular Biology & CMBI, Leopold-Franzens-University Innsbruck, Innsbruck 6020, Austria
| | - Sandra Rizzi
- Genomics, Stem Cell Biology and Regenerative Medicine Group, Institute of Molecular Biology & CMBI, Leopold-Franzens-University Innsbruck, Innsbruck 6020, Austria
| | - Liviu Pirvan
- Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK
| | - Luca Peruzzotti-Jametti
- Department of Clinical Neurosciences and NIHR Biomedical Research Centre, University of Cambridge, Cambridge CB2 0AH, UK; Department of Metabolism, Digestion and Reproduction, Imperial College London, London SW7 2AZ, UK
| | - Valentina Fossati
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, USA
| | - Frank Edenhofer
- Genomics, Stem Cell Biology and Regenerative Medicine Group, Institute of Molecular Biology & CMBI, Leopold-Franzens-University Innsbruck, Innsbruck 6020, Austria
| | - Tommaso Leonardi
- Center for Genomic Science of IIT@SEMM, Instituto Italiano di Tecnologia (IIT), 20139 Milan, Italy
| | - Christian Frezza
- Institute for Metabolomics in Ageing, Cluster of Excellence Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne 50931, Germany; Institute of Genetics, Faculty of Mathematics and Natural Sciences, Faculty of Medicine, University of Cologne, Cologne 50674, Germany
| | - Irina Mohorianu
- Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, CO 80045, USA.
| | - Stefano Pluchino
- Department of Clinical Neurosciences and NIHR Biomedical Research Centre, University of Cambridge, Cambridge CB2 0AH, UK.
| |
Collapse
|
8
|
Lerma-Martin C, Badia-I-Mompel P, Ramirez Flores RO, Sekol P, Schäfer PSL, Riedl CJ, Hofmann A, Thäwel T, Wünnemann F, Ibarra-Arellano MA, Trobisch T, Eisele P, Schapiro D, Haeussler M, Hametner S, Saez-Rodriguez J, Schirmer L. Cell type mapping reveals tissue niches and interactions in subcortical multiple sclerosis lesions. Nat Neurosci 2024:10.1038/s41593-024-01796-z. [PMID: 39501036 DOI: 10.1038/s41593-024-01796-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 09/30/2024] [Indexed: 11/08/2024]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system. Inflammation is gradually compartmentalized and restricted to specific tissue niches such as the lesion rim. However, the precise cell type composition of such niches, their interactions and changes between chronic active and inactive stages are incompletely understood. We used single-nucleus and spatial transcriptomics from subcortical MS and corresponding control tissues to map cell types and associated pathways to lesion and nonlesion areas. We identified niches such as perivascular spaces, the inflamed lesion rim or the lesion core that are associated with the glial scar and a cilia-forming astrocyte subtype. Focusing on the inflamed rim of chronic active lesions, we uncovered cell-cell communication events between myeloid, endothelial and glial cell types. Our results provide insight into the cellular composition, multicellular programs and intercellular communication in tissue niches along the conversion from a homeostatic to a dysfunctional state underlying lesion progression in MS.
Collapse
Affiliation(s)
- Celia Lerma-Martin
- Department of Neurology, Division of Neuroimmunology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Pau Badia-I-Mompel
- Institute for Computational Biomedicine, Faculty of Medicine, Heidelberg University and Heidelberg University Hospital, Heidelberg, Germany
- GSK, Cellzome, Heidelberg, Germany
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Hinxton, UK
| | - Ricardo O Ramirez Flores
- Institute for Computational Biomedicine, Faculty of Medicine, Heidelberg University and Heidelberg University Hospital, Heidelberg, Germany
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Hinxton, UK
| | - Patricia Sekol
- Department of Neurology, Division of Neuroimmunology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Philipp S L Schäfer
- Institute for Computational Biomedicine, Faculty of Medicine, Heidelberg University and Heidelberg University Hospital, Heidelberg, Germany
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Hinxton, UK
| | - Christian J Riedl
- Department of Neurology, Division of Neuropathology and Neurochemistry, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Annika Hofmann
- Department of Neurology, Division of Neuroimmunology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Thomas Thäwel
- Department of Neurology, Division of Neuroimmunology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Florian Wünnemann
- Institute for Computational Biomedicine, Faculty of Medicine, Heidelberg University and Heidelberg University Hospital, Heidelberg, Germany
| | - Miguel A Ibarra-Arellano
- Institute for Computational Biomedicine, Faculty of Medicine, Heidelberg University and Heidelberg University Hospital, Heidelberg, Germany
| | - Tim Trobisch
- Department of Neurology, Division of Neuroimmunology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Philipp Eisele
- Department of Neurology, Division of Neuroimmunology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Mannheim Center for Translational Neuroscience, Medical Faculty, Mannheim Heidelberg University, Mannheim, Germany
| | - Denis Schapiro
- Institute for Computational Biomedicine, Faculty of Medicine, Heidelberg University and Heidelberg University Hospital, Heidelberg, Germany
- Institute of Pathology, Faculty of Medicine, Heidelberg University and Heidelberg University Hospital, Heidelberg, Germany
- Translational Spatial Profiling Center (TSPC), Heidelberg, Germany
| | | | - Simon Hametner
- Department of Neurology, Division of Neuropathology and Neurochemistry, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Julio Saez-Rodriguez
- Institute for Computational Biomedicine, Faculty of Medicine, Heidelberg University and Heidelberg University Hospital, Heidelberg, Germany.
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Hinxton, UK.
| | - Lucas Schirmer
- Department of Neurology, Division of Neuroimmunology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
- Mannheim Center for Translational Neuroscience, Medical Faculty, Mannheim Heidelberg University, Mannheim, Germany.
- Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany.
| |
Collapse
|
9
|
Filippi M, Preziosa P, Barkhof F, Ciccarelli O, Cossarizza A, De Stefano N, Gasperini C, Geraldes R, Granziera C, Haider L, Lassmann H, Margoni M, Pontillo G, Ropele S, Rovira À, Sastre-Garriga J, Yousry TA, Rocca MA. The ageing central nervous system in multiple sclerosis: the imaging perspective. Brain 2024; 147:3665-3680. [PMID: 39045667 PMCID: PMC11531849 DOI: 10.1093/brain/awae251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 06/10/2024] [Accepted: 06/23/2024] [Indexed: 07/25/2024] Open
Abstract
The interaction between ageing and multiple sclerosis is complex and carries significant implications for patient care. Managing multiple sclerosis effectively requires an understanding of how ageing and multiple sclerosis impact brain structure and function. Ageing inherently induces brain changes, including reduced plasticity, diminished grey matter volume, and ischaemic lesion accumulation. When combined with multiple sclerosis pathology, these age-related alterations may worsen clinical disability. Ageing may also influence the response of multiple sclerosis patients to therapies and/or their side effects, highlighting the importance of adjusted treatment considerations. MRI is highly sensitive to age- and multiple sclerosis-related processes. Accordingly, MRI can provide insights into the relationship between ageing and multiple sclerosis, enabling a better understanding of their pathophysiological interplay and informing treatment selection. This review summarizes current knowledge on the immunopathological and MRI aspects of ageing in the CNS in the context of multiple sclerosis. Starting from immunosenescence, ageing-related pathological mechanisms and specific features like enlarged Virchow-Robin spaces, this review then explores clinical aspects, including late-onset multiple sclerosis, the influence of age on diagnostic criteria, and comorbidity effects on imaging features. The role of MRI in understanding neurodegeneration, iron dynamics and myelin changes influenced by ageing and how MRI can contribute to defining treatment effects in ageing multiple sclerosis patients, are also discussed.
Collapse
Affiliation(s)
- Massimo Filippi
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Neurorehabilitation Unit, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Neurophysiology Service, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Paolo Preziosa
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit, 1081 HV Amsterdam, The Netherlands
- Queen Square Institute of Neurology and Centre for Medical Image Computing, University College London, London WC1N 3BG, UK
| | - Olga Ciccarelli
- Queen Square MS Centre, UCL Institute of Neurology, UCL, London WC1N 3BG, UK
- NIHR (National Institute for Health and Care Research) UCLH (University College London Hospitals) BRC (Biomedical Research Centre), London WC1N 3BG, UK
| | - Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, 42121 Modena, Italy
| | - Nicola De Stefano
- Department of Medicine, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy
| | - Claudio Gasperini
- Department of Neurosciences, S Camillo Forlanini Hospital Rome, 00152 Rome, Italy
| | - Ruth Geraldes
- Clinical Neurology, John Radcliffe Hospital, Oxford University Foundation Trust, Oxford OX3 9DU, UK
- Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK
| | - Cristina Granziera
- Department of Neurology, University Hospital Basel and University of Basel, 4031 Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, 4031 Basel, Switzerland
- Translational Imaging in Neurology (ThINk) Basel, Department of Biomedical Engineering, University Hospital Basel and University of Basel, 4031 Basel, Switzerland
| | - Lukas Haider
- Queen Square Institute of Neurology and Centre for Medical Image Computing, University College London, London WC1N 3BG, UK
- Department of Biomedical Imaging and Image Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
| | - Hans Lassmann
- Center for Brain Research, Medical University of Vienna, 1090 Vienna, Austria
| | - Monica Margoni
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Neurorehabilitation Unit, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Giuseppe Pontillo
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit, 1081 HV Amsterdam, The Netherlands
- Queen Square Institute of Neurology and Centre for Medical Image Computing, University College London, London WC1N 3BG, UK
- Department of Advanced Biomedical Sciences, University “Federico II”, 80138 Naples, Italy
| | - Stefan Ropele
- Department of Neurology, Medical University of Graz, 8010 Graz, Austria
| | - Àlex Rovira
- Neuroradiology Section, Department of Radiology, Hospital Universitari Vall d'Hebron, 08035 Barcelona, Spain
| | - Jaume Sastre-Garriga
- Neurology Department and Multiple Sclerosis Centre of Catalunya (Cemcat), Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - Tarek A Yousry
- Lysholm Department of Neuroradiology, UCLH National Hospital for Neurology and Neurosurgery, Neuroradiological Academic Unit, UCL Institute of Neurology, London WC1N 3BG, UK
| | - Maria A Rocca
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Vita-Salute San Raffaele University, 20132 Milan, Italy
| |
Collapse
|
10
|
Piscitelli D, Brichetto G, Geri T, Battista S, Testa M, Monti Bragadin M, Pellicciari L. Italian adaptation and psychometric validation of the Fatigue Impact Scale (FIS) and its modified versions in adults with multiple sclerosis: a Rasch analysis study. Disabil Rehabil 2024; 46:5366-5379. [PMID: 38236054 DOI: 10.1080/09638288.2024.2302878] [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: 05/25/2023] [Revised: 12/14/2023] [Accepted: 12/21/2023] [Indexed: 01/19/2024]
Abstract
PURPOSE Several outcome measures are available to assess the severity of fatigue in people with multiple sclerosis (MS). The aim of this study was to adapt the Italian version of the Fatigue Impact Scale (FIS-40) and its modified versions: a 21-item Modified scale (MFIS-21), its 5-item short version (MFIS-5), and an 8-item version for daily use (DFIS-8) and investigate their measurement properties through classical theory-test (CTT) and Rasch analysis (RA). METHODS 229 Italian-speaking adults with MS were included. Questionnaires were cross-culturally translated and subjected to CTT (i.e. internal consistency through Cronbach's alpha and unidimensionality through confirmatory factor analysis [CFA]) and RA. (i.e. internal construct validity, reliability, and targeting). RESULTS Internal consistency was high for all scales (>0.850). Final CFAs reported issues in the unidimensionality for all scales except for FIS-40. Baseline RA revealed a misfit for all scales. After adjusting for local dependency, FIS-40, MFIS-21, and MFIS-5 fitted the Rasch model (RM). MFIS-21 and D-FIS-8 required a structural modification, i.e. item deletions to satisfy the RM. CONCLUSION The FIS-40, MFIS-21, MFIS-5, and DFIS-8 achieved the fit to the RM after statistical and structural modifications. The fit to the RM allowed for providing ordinal-to-interval measurement conversion tables for all the questionnaires.
Collapse
Affiliation(s)
- Daniele Piscitelli
- Doctor of Physical Therapy Program, Department of Kinesiology, University of Connecticut, Storrs, CT, USA
- School of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Giampaolo Brichetto
- Scientific Research Area, Italian MS Foundation (FISM), Genoa, Italy
- AISM Rehabilitation Center, Italian MS Society (AISM), Genoa, Italy
| | | | - Simone Battista
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Campus of Savona, Italy
| | - Marco Testa
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Campus of Savona, Italy
| | - Margherita Monti Bragadin
- Scientific Research Area, Italian MS Foundation (FISM), Genoa, Italy
- AISM Rehabilitation Center, Italian MS Society (AISM), Genoa, Italy
| | | |
Collapse
|
11
|
Chen Y, Wang Y, Lu Q, Zhao Y, Cruz J, Ma J, Ding G, Qiao X, Cheng X. Demyelination in cuprizone mice is ameliorated by calycosin mediated through astrocyte Nrf2 signaling pathway. Eur J Pharmacol 2024; 985:177090. [PMID: 39489278 DOI: 10.1016/j.ejphar.2024.177090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/27/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024]
Abstract
Oxidative stress plays a pivotal role in multiple sclerosis (MS), triggering demyelination predominantly through excessive peroxide production and the depletion of antioxidants. The accumulation of oxidative damage can be caused by dysregulation of astrocytes, which are the brain's main regulators of oxidative homeostasis. Calycosin, an essential bioactive component extracted from Astragalus, is recognized for its neuroprotective properties. Although recent research has highlighted calycosin's neuroprotective capabilities, its role in demyelinating conditions like MS remains unclear. In this work, we examined the possible molecular mechanism of calycosin's neuroprotective effect on cuprizone (CPZ)-induced demylination in mice. According to our research, calycosin successfully reduced demyelination and behavioral dysfuction in CPZ mice. Calycosin also decreased the production of oxidative stress and enhanced the expression of antioxidants in CPZ mice and in astrocytes induced by hydrogen peroxide (H2O2). Furthermore, both in vivo and in vitro experiments demonstrated that calycosin promoted the nuclear translocation of nuclear factor erythroid 2-related factor 2 (Nrf2) along with the upregulation of heme oxygenase 1 (HO-1), NAD(P)H quinone dehydrogenase 1 (NQO1), and superoxide dismutase (SOD). Importantly, the application of all-trans retinoic acid (ATRA), a specific inhibitor of Nrf2, effectively reversed the myelin-protective and antioxidant effects conferred by calycosin. This study suggested that calycosin might exert neuroprotection by inhibiting oxidative stress and reducing demyelination via the activation of astrocyte Nrf2 signaling. These findings indicated that calycosin might be a potential candidate for treating MS.
Collapse
Affiliation(s)
- Yuxin Chen
- Institute of Clinical Immunology, Yue-yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Yuanhua Wang
- Institute of Clinical Immunology, Yue-yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Qijin Lu
- Institute of Clinical Immunology, Yue-yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Yan Zhao
- Institute of Clinical Immunology, Yue-yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Jennifer Cruz
- Institute of Clinical Immunology, Yue-yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China; Doctoral Program of Acupuncture & Oriental Medicine, The Atlantic Institute of Oriental Medicine, Fort Lauderdale, FL, USA
| | - Jinyun Ma
- Institute of Clinical Immunology, Yue-yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Guiqing Ding
- Institute of Clinical Immunology, Yue-yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Xi Qiao
- Institute of Clinical Immunology, Yue-yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Xiaodong Cheng
- Institute of Clinical Immunology, Yue-yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China.
| |
Collapse
|
12
|
Özbaş E, Balkan AF, Salcı Y. The effect of cognitive rehabilitation on motor function and balance in individuals with multiple sclerosis: a systematic review. Acta Neurol Belg 2024:10.1007/s13760-024-02673-z. [PMID: 39485626 DOI: 10.1007/s13760-024-02673-z] [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: 06/04/2024] [Accepted: 10/17/2024] [Indexed: 11/03/2024]
Abstract
BACKGROUND Balance and motor functions are associated with a complex sensory-cognitive-motor system that is not reliant on a single component. In people with Multiple Sclerosis (pwMS), deficits in cognitive domains may cause abnormal gait and balance disorders. Therefore, the impact of cognitive rehabilitation (CR) on motor and balance functions in pwMS was investigated. METHODS Following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, the review was registered in the Prospero system. A systematic search was conducted in PubMed, Web of Science, Scopus, and Cochrane Library until April 2024. Articles meeting the eligibility criteria were assessed and included in the review, those that evaluated the impact of CR on balance and motor functions. Bias in the studies was assessed by using the Risk of Bias-2 tool for randomized controlled trials. The articles' evidence level was evaluated using the Modified Bakker Scale. RESULTS After searching four databases, eight studies were included in the review, comprising a total of 668 participants. The bias risks of the studies were categorized as low in two studies, unclear in five studies, and high in one study. The evidence levels of the studies showed high levels of evidence for balance and motor skills. CONCLUSION This review suggests that CR may have a positive impact on motor and balance performance in pwMS, underscoring the relationship between cognition and motor function. Further research is required to enhance this evidence base, particularly given the limited number of studies in this area.
Collapse
Affiliation(s)
- Ezgi Özbaş
- Faculty of Physical Therapy and Rehabilitation, Hacettepe University, 06230, Ankara, Turkey.
- Institute of Health Sciences, Neurology Physiotherapy Doctorate Program, Hacettepe University, 06230, Samanpazari, Ankara, Turkey.
| | - Ayla Fil Balkan
- Faculty of Physical Therapy and Rehabilitation, Hacettepe University, 06230, Ankara, Turkey
| | - Yeliz Salcı
- Faculty of Physical Therapy and Rehabilitation, Hacettepe University, 06230, Ankara, Turkey
| |
Collapse
|
13
|
Konen FF, Gingele S, Hümmert MW, Möhn N, Streichert AL, Kretschmer JR, Grote-Levi L, Nay S, Seeliger T, Ratuszny D, Jendretzky KF, Tkachenko D, Jacobs R, Skripuletz T, Schwenkenbecher P. Rapid depletion of CD20 + B and T cells following ofatumumab therapy onset. Mult Scler Relat Disord 2024; 91:105886. [PMID: 39299183 DOI: 10.1016/j.msard.2024.105886] [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: 07/08/2024] [Revised: 08/24/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND The humanized monoclonal anti-CD20-antibody ofatumumab is highly effective in treating relapsing multiple sclerosis (MS). OBJECTIVE This study aimed to investigate the immanent effect of ofatumumab on the peripheral immune system, particularly targeting B and T cells expressing CD20. METHODS Blood samples of 53 MS patients receiving ofatumumab were collected prior to first application and after one week, two weeks and three months. Multicolor flow cytometry was used to phenotype peripheral blood mononuclear cells, and immunoglobulin (Ig) concentrations were measured by nephelometry. RESULTS Among CD20+ lymphocytes, 13 % co-expressed CD3 (identifying them as CD3+CD20+ T lymphocytes), with a noticeable shift in the CD4/CD8-ratio towards CD8+ T cells. One week after administering ofatumumab, a significant reduction of CD20+ lymphocytes with complete depletion of CD3+CD20+ T lymphocytes was observed, persisting during the investigation period. During the treatment, IgM levels showed a slight but significant decrease, whereas IgA and IgG levels remained stable. CONCLUSION Ofatumumab effectively depletes CD20+ lymphocytes already after the first administration. This depletion affects not only B cells, but also a small proportion of T cells (CD3+CD20+), affirming the hypothesis that the anti-inflammatory effects of CD20+ cell depletion might extend to the reduction of CD3+CD20+ T lymphocytes.
Collapse
Affiliation(s)
- Franz Felix Konen
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Stefan Gingele
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Martin W Hümmert
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Nora Möhn
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Anna Lena Streichert
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Julian Reza Kretschmer
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Lea Grote-Levi
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Sandra Nay
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Tabea Seeliger
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Dominica Ratuszny
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | | | - Daria Tkachenko
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Roland Jacobs
- Department of Rheumatology and Clinical Immunology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Thomas Skripuletz
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Philipp Schwenkenbecher
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany.
| |
Collapse
|
14
|
Sadeghnejad A, Pazoki A, Yazdanpanah E, Esmaeili SA, Yousefi B, Sadighi-Moghaddam B, Baharlou R, Haghmorad D. Exploring the role of mesenchymal stem cells in modulating immune responses via Treg and Th2 cell activation: insights from mouse model of multiple sclerosis. APMIS 2024; 132:888-899. [PMID: 39030955 DOI: 10.1111/apm.13456] [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: 03/15/2024] [Accepted: 07/03/2024] [Indexed: 07/22/2024]
Abstract
Multiple sclerosis is a demyelinating neurodegenerative disease, and its animal model, experimental autoimmune encephalomyelitis (EAE), exhibits immunological and clinical similarities. The study aimed to examine mechanisms underlying therapeutic effects of mesenchymal stem cell administration in EAE. C57BL/6 mice were separated into control and treatment groups (T1, T2, and T3); EAE was induced in all animals. Clinical examinations were conducted daily, and on 25th day, animals were sacrificed, and spinal cord was stained for histological analysis. Additionally, spleen cell proliferation assay, assessments of cytokine, and gene expression in both spinal cord and spleen cells were performed. The results indicated a significant reduction in clinical symptoms among treatment groups compared to control group. Histological analyses revealed decreased infiltration of lymphocytes into the spinal cord and reduced demyelinated areas in treatment groups compared to control group. Cytokine production of IL-10, TGF-β, and IL-4 were significantly enhanced and IFN-γ and TNF-α in treatment groups were decreased relative to control group. Also, gene expression of CTLA-4, PD-1, IL-27, and IL-33 indicated a significant increase in treatment groups. The administration of MSCs significantly improved clinical symptoms, attenuated inflammation, and reduced spinal cord demyelination in EAE, suggesting a potential protective effect on disease progression.
Collapse
Affiliation(s)
- Abdolvahid Sadeghnejad
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Alireza Pazoki
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Esmaeil Yazdanpanah
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Immunology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed-Alireza Esmaeili
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Immunology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Bahman Yousefi
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Bijan Sadighi-Moghaddam
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Rasoul Baharlou
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Dariush Haghmorad
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
15
|
Mrabet S, Sghaier I, Souissi A, Gharbi A, Abida Y, Kacem I, Gargouri-Berrechid A, Gouider R. Neurofilaments light chains as a diagnostic and predictive biomarker for Tunisian Multiple Sclerosis patients. Mult Scler Relat Disord 2024; 91:105901. [PMID: 39341199 DOI: 10.1016/j.msard.2024.105901] [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: 01/01/2024] [Revised: 09/18/2024] [Accepted: 09/20/2024] [Indexed: 09/30/2024]
Abstract
BACKGROUND Multiple Sclerosis (MS) course was shown to be more severe among North Africans compared to Caucasians. Validation of prognostic biomarkers of disease activity and severity is a priority in our practice. OBJECTIVE We aimed to investigate the association between baseline cerebrospinal fluid (CSF) and serum NfL (sNFL) levels and disease activity and disability accrual in a cohort of Tunisian patients with MS. METHODS A cross-sectional study was conducted, in the department of Neurology of Razi Hospital, including patients diagnosed with MS. Patient's data were retrieved from our local MS database. Blood and CSF sampling were performed at the first visit. sNFL levels were measured using the Enzyme-Linked Immuno-Sorbent Assay (ELISA) sandwich technique. RESULTS Three hundred MS patients were enrolled (sex-ratio= 3.05; mean age at MS onset=28.83 years+9.55, mean MS course = 10.21 years+8.96). MS phenotype was predominately relapsing (73%). CSF NfL levels were significantly correlated to the serum ones. NfL concentrations were significantly associated with MS activity (p = 0.012), disease progression (p = 0.001), and higher Multiple Sclerosis Severity Scores (MSSS) (p = 0.0017, r = 0.28). CONCLUSIONS These results support the value of NfL as a sensitive and clinically meaningful CSF and blood biomarker to evaluate MS activity and outcomes among Tunisian MS patients.
Collapse
Affiliation(s)
- Saloua Mrabet
- Neurology Department, LR18SP03, Razi University Hospital, 1 rue des orangers, Manouba, Tunis 2010, Tunisia; Faculty of Medicine of Tunis, University of Tunis El Manar, 15, Rue Djebel Lakhdhar, La Rabta, Tunis 1007, Tunisia; Clinical Investigation Center (CIC) "Neurosciences and Mental Health", Razi University Hospital, 1 rue des orangers, Manouba, Tunis 2010, Tunisia
| | - Ikram Sghaier
- Neurology Department, LR18SP03, Razi University Hospital, 1 rue des orangers, Manouba, Tunis 2010, Tunisia; Clinical Investigation Center (CIC) "Neurosciences and Mental Health", Razi University Hospital, 1 rue des orangers, Manouba, Tunis 2010, Tunisia
| | - Amira Souissi
- Neurology Department, LR18SP03, Razi University Hospital, 1 rue des orangers, Manouba, Tunis 2010, Tunisia; Faculty of Medicine of Tunis, University of Tunis El Manar, 15, Rue Djebel Lakhdhar, La Rabta, Tunis 1007, Tunisia; Clinical Investigation Center (CIC) "Neurosciences and Mental Health", Razi University Hospital, 1 rue des orangers, Manouba, Tunis 2010, Tunisia
| | - Alya Gharbi
- Neurology Department, LR18SP03, Razi University Hospital, 1 rue des orangers, Manouba, Tunis 2010, Tunisia; Faculty of Medicine of Tunis, University of Tunis El Manar, 15, Rue Djebel Lakhdhar, La Rabta, Tunis 1007, Tunisia; Clinical Investigation Center (CIC) "Neurosciences and Mental Health", Razi University Hospital, 1 rue des orangers, Manouba, Tunis 2010, Tunisia
| | - Youssef Abida
- Neurology Department, LR18SP03, Razi University Hospital, 1 rue des orangers, Manouba, Tunis 2010, Tunisia; Faculty of Medicine of Tunis, University of Tunis El Manar, 15, Rue Djebel Lakhdhar, La Rabta, Tunis 1007, Tunisia; Clinical Investigation Center (CIC) "Neurosciences and Mental Health", Razi University Hospital, 1 rue des orangers, Manouba, Tunis 2010, Tunisia
| | - Imen Kacem
- Neurology Department, LR18SP03, Razi University Hospital, 1 rue des orangers, Manouba, Tunis 2010, Tunisia; Faculty of Medicine of Tunis, University of Tunis El Manar, 15, Rue Djebel Lakhdhar, La Rabta, Tunis 1007, Tunisia; Clinical Investigation Center (CIC) "Neurosciences and Mental Health", Razi University Hospital, 1 rue des orangers, Manouba, Tunis 2010, Tunisia
| | - Amina Gargouri-Berrechid
- Neurology Department, LR18SP03, Razi University Hospital, 1 rue des orangers, Manouba, Tunis 2010, Tunisia; Faculty of Medicine of Tunis, University of Tunis El Manar, 15, Rue Djebel Lakhdhar, La Rabta, Tunis 1007, Tunisia; Clinical Investigation Center (CIC) "Neurosciences and Mental Health", Razi University Hospital, 1 rue des orangers, Manouba, Tunis 2010, Tunisia
| | - Riadh Gouider
- Neurology Department, LR18SP03, Razi University Hospital, 1 rue des orangers, Manouba, Tunis 2010, Tunisia; Faculty of Medicine of Tunis, University of Tunis El Manar, 15, Rue Djebel Lakhdhar, La Rabta, Tunis 1007, Tunisia; Clinical Investigation Center (CIC) "Neurosciences and Mental Health", Razi University Hospital, 1 rue des orangers, Manouba, Tunis 2010, Tunisia.
| |
Collapse
|
16
|
Chen L, Ren Z, Clark KA, Lou C, Liu F, Cao Q, Manning AR, Martin ML, Luskin E, O'Donnell CM, Azevedo CJ, Calabresi PA, Freeman L, Henry RG, Longbrake EE, Oh J, Papinutto N, Bilello M, Song JW, Kaisey M, Sicotte NL, Reich DS, Solomon AJ, Ontaneda D, Sati P, Absinta M, Schindler MK, Shinohara RT. Multicenter validation of automated detection of paramagnetic rim lesions on brain MRI in multiple sclerosis. J Neuroimaging 2024; 34:750-757. [PMID: 39410780 DOI: 10.1111/jon.13242] [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: 07/21/2024] [Revised: 09/15/2024] [Accepted: 09/24/2024] [Indexed: 11/12/2024] Open
Abstract
BACKGROUND AND PURPOSE Paramagnetic rim lesions (PRLs) are an MRI biomarker of chronic inflammation in people with multiple sclerosis (MS). PRLs may aid in the diagnosis and prognosis of MS. However, manual identification of PRLs is time-consuming and prone to poor interrater reliability. To address these challenges, the Automated Paramagnetic Rim Lesion (APRL) algorithm was developed to automate PRL detection. The primary objective of this study is to evaluate the accuracy of APRL for detecting PRLs in a multicenter setting. METHODS We applied APRL to a multicenter dataset, which included 3-Tesla MRI acquired in 92 participants (43 with MS, 14 with clinically isolated syndrome [CIS]/radiologically isolated syndrome [RIS], 35 without RIS/CIS/MS). Subsequently, we assessed APRL's performance by comparing its results with manual PRL assessments carried out by a team of trained raters. RESULTS Among the 92 participants, expert raters identified 5637 white matter lesions and 148 PRLs. The automated segmentation method successfully captured 115 (78%) of the manually identified PRLs. Within these 115 identified lesions, APRL differentiated between manually identified PRLs and non-PRLs with an area under the curve (AUC) of .73 (95% confidence interval [CI]: [.68, .78]). At the subject level, the count of APRL-identified PRLs predicted MS diagnosis with an AUC of .69 (95% CI: [.57, .81]). CONCLUSION Our study demonstrated APRL's capability to differentiate between PRLs and lesions without paramagnetic rims in a multicenter study. Automated identification of PRLs offers greater efficiency over manual identification and could facilitate large-scale assessments of PRLs in clinical trials.
Collapse
Affiliation(s)
- Luyun Chen
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- School of Medicine, Georgetown University, Washington, DC, USA
| | - Zheng Ren
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Kelly A Clark
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | | | - Fang Liu
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Quy Cao
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Abigail R Manning
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Melissa L Martin
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Elaina Luskin
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Carly M O'Donnell
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Christina J Azevedo
- Department of Neurology, University of Southern California, Los Angeles, California, USA
| | - Peter A Calabresi
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Leorah Freeman
- Department of Neurology, Dell Medical School, The University of Texas at Austin, Austin, Texas, USA
| | - Roland G Henry
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, California, USA
| | - Erin E Longbrake
- Department of Neurology, Yale University, New Haven, Connecticut, USA
| | - Jiwon Oh
- Division of Neurology, Department of Medicine, St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Nico Papinutto
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, California, USA
| | - Michel Bilello
- Department of Radiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Jae W Song
- Department of Radiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Marwa Kaisey
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Nancy L Sicotte
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Daniel S Reich
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Andrew J Solomon
- Department of Neurological Sciences, Larner College of Medicine at the University of Vermont, Burlington, Vermont, USA
| | - Daniel Ontaneda
- Mellen Center for Multiple Sclerosis, Cleveland Clinic, Cleveland, Ohio, USA
| | - Pascal Sati
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Martina Absinta
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Matthew K Schindler
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Russell T Shinohara
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Center for Biomedical Image Computing and Analytics, Department of Radiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
17
|
Hu C, Vasileiou ES, Salter A, Marrie RA, Kowalec K, Fitzgerald KC. Evidence of symptom specificity for depression in multiple sclerosis: A two sample Mendelian randomization study. Mult Scler Relat Disord 2024; 91:105866. [PMID: 39276599 PMCID: PMC11527576 DOI: 10.1016/j.msard.2024.105866] [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: 01/22/2024] [Revised: 08/07/2024] [Accepted: 09/01/2024] [Indexed: 09/17/2024]
Abstract
BACKGROUND Depression is common and phenotypically heterogenous in multiple sclerosis (MS). MS may increase risk of some but not all affective symptoms or certain symptoms may predispose individuals to higher MS risk. OBJECTIVE To assess the existence and direction of causality between distinct depressive symptoms and MS using two-sample Mendelian randomization (MR). METHODS Using summary data from genome-wide association studies, we selected genetic instrument variables (IV) for MS (n = 115,776) and IVs for depressive symptoms (average n = 117,713): anhedonia, altered appetite, concentration, depressed mood, fatigue, inadequacy, psychomotor changes, sleeping problems and suicidality. We performed two-sample MR in either direction using inverse-variance models. Sensitivity analyses included weighted-median and MR-Egger regression. Obesity is a known risk factor for MS and depression; we adjusted for body mass index in multivariable-MR. RESULTS Genetic liability to MS was associated with anhedonia (IVW estimate per 102: 0.69; 95 % CI: 0.24-1.13; p = 0.002), concentration difficulty (0.66; 0.19-1.13; p = 0.006) and psychomotor changes (0.37; 0.08-0.65; p = 0.01). Results were similar in sensitivity analyses. In the opposite direction, we found no evidence of a causal relationship for any affective symptom on MS risk. CONCLUSIONS Genetic susceptibility to MS was associated with anhedonia, concentration, and psychomotor-related symptoms, suggesting a specific phenotype of depression in MS.
Collapse
Affiliation(s)
- Chen Hu
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Eleni S Vasileiou
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Amber Salter
- Department of Neurology, University of Texas Southwestern, Dallas, TX, United States
| | - Ruth Ann Marrie
- Department of Internal Medicine, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada; Department of Community Health Sciences, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Kaarina Kowalec
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada; Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Solna, Sweden
| | - Kathryn C Fitzgerald
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
18
|
Borrelli S, Leclercq S, Pasi M, Maggi P. Cerebral small vessel disease and glymphatic system dysfunction in multiple sclerosis: A narrative review. Mult Scler Relat Disord 2024; 91:105878. [PMID: 39276600 DOI: 10.1016/j.msard.2024.105878] [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: 06/24/2024] [Revised: 09/03/2024] [Accepted: 09/04/2024] [Indexed: 09/17/2024]
Abstract
As the multiple sclerosis (MS) population ages, the prevalence of vascular comorbidities increases, potentially accelerating disease progression and brain atrophy. Recent studies highlight the prevalence of cerebral small vessel disease (CSVD) in MS, suggesting a potential link between vascular comorbidities and accelerated disability. CSVD affects the brain's small vessels, often leading to identifiable markers on MRI such as enlarged perivascular spaces (EPVS). EPVS are increasingly recognized also in MS and have been associated with vascular comorbidities, lower percentage of MS-specific perivenular lesions, brain atrophy and aging. The exact sequence of event leading to MRI visible EPVS is yet to be determined, but an impaired perivascular brain fluid drainage appears a possible physiopathological explanation for EPVS in both CSVD and MS. In this context, a dysfunction of the brain fluid clearance system - also known as "glymphatic system" - appears associated in MS to aging, neuroinflammation, and vascular dysfunction. Advanced imaging techniques show an impaired glymphatic function in both MS and CSVD. Additionally, lifestyle factors such as physical exercise, diet, and sleep quality appear to influence glymphatic function, potentially revealing novel therapeutic strategies to mitigate microangiopathy and neuroinflammation in MS. This review underscores the potential role of glymphatic dysfunction in the complex and not-yet elucidated interplay between neuroinflammation and CSVD in MS.
Collapse
Affiliation(s)
- Serena Borrelli
- Neuroinflammation Imaging Lab (NIL), Institute of NeuroScience, Université catholique de Louvain, Brussels, Belgium; Department of Neurology, Hôpital Erasme, Hôpital Universitaire de Bruxelles, Université Libre de Brussels, Brussels, Belgium.
| | - Sophie Leclercq
- Laboratory of Nutritional Psychiatry, Institute of NeuroScience, Université catholique de Louvain, Brussels, Belgium
| | - Marco Pasi
- Stroke Unit, Department of Neurology, CIC-IT 1415, CHRU de Tours, INSERM 1253 iBrain, Tours, France
| | - Pietro Maggi
- Neuroinflammation Imaging Lab (NIL), Institute of NeuroScience, Université catholique de Louvain, Brussels, Belgium; Department of Neurology, Cliniques Universitaires Saint-Luc, Université catholique de Louvain, Av. Hippocrate 10, Brussels 1200, Belgium.
| |
Collapse
|
19
|
Stavrogianni K, Kitsos DK, Giannopapas V, Stefanou MI, Christouli N, Smyrni V, Chasiotis AK, Akrivaki A, Dimitriadou E, Chondrogianni M, Tsivgoulis G, Giannopoulos S. Impact of Siponimod on Clinical and Radiological Parameters of Secondary Progressive Multiple Sclerosis: A Real-World Prospective Study. J Clin Neurol 2024; 20:591-598. [PMID: 39505311 PMCID: PMC11543385 DOI: 10.3988/jcn.2024.0149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/11/2024] [Accepted: 09/05/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND AND PURPOSE Secondary progressive multiple sclerosis (SPMS) presents with a challenging clinical phenotype, and siponimod has a potential to treat the active clinical phenotype of this disease. This single-center longitudinal study aimed to determine the therapeutic effects of siponimod in patients with active SPMS over 12 months. METHODS The clinical and radiological parameters of 50 patients with active SPMS treated using siponimod were assessed at baseline and after a 1-year follow-up period using the annual relapse rate (ARR), the Expanded Disability Status Scale (EDSS), the occurrence of gadolinium-enhanced lesion (GdE+), the Modified Fatigue Impact Scale (MFIS), and the Symbol Digit Modalities Test. The urine bladder postvoid residual (PVR) volume was also measured in a subcohort of 39 participants. Participants with an EDSS score ≥5.0 at baseline were finally assessed separately in prespecified subgroup analyses. RESULTS There were significant reductions in ARR (p<0.001), GdE+ (p<0.001), and MFIS score (p=0.001) during the follow-up period. The progression of physical and cognitive disabilities remained stable (p>0.05). The PVR-volume analysis revealed a significant decrease in urine bladder PVR volume (p<0.001). These observations were consistent for the subgroup with EDSS score ≥5.0. CONCLUSIONS Siponimod demonstrated efficacy in reducing ARR, GdE+, fatigue levels, and PVR volume, while maintaining stability in the cognitive and physical disability statuses of patients with SPMS. Similar findings were documented in the subgroup with EDSS score ≥5.0.
Collapse
Affiliation(s)
- Konstantina Stavrogianni
- Second Department of Neurology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - Dimitrios K Kitsos
- Second Department of Neurology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Vasileios Giannopapas
- Second Department of Neurology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- Department of Physical Therapy, University of West Attica, Attica, Greece
- Laboratory of Neuromuscular and Cardiovascular Study of Motion-LANECASM, University of West Attica, Attica, Greece
| | - Maria-Ioanna Stefanou
- Second Department of Neurology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Niki Christouli
- Second Department of Neurology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Vassiliki Smyrni
- Second Department of Neurology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Athanasios K Chasiotis
- Second Department of Neurology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- Department of Physical Therapy, University of West Attica, Attica, Greece
- Laboratory of Neuromuscular and Cardiovascular Study of Motion-LANECASM, University of West Attica, Attica, Greece
| | - Alexandra Akrivaki
- Second Department of Neurology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Evangelia Dimitriadou
- Second Department of Neurology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria Chondrogianni
- Second Department of Neurology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Georgios Tsivgoulis
- Second Department of Neurology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Sotirios Giannopoulos
- Second Department of Neurology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
20
|
Yang F, Zhao LY, Yang WQ, Chao S, Ling ZX, Sun BY, Wei LP, Zhang LJ, Yu LM, Cai GY. Quantitative proteomics and multi-omics analysis identifies potential biomarkers and the underlying pathological molecular networks in Chinese patients with multiple sclerosis. BMC Neurol 2024; 24:423. [PMID: 39478468 PMCID: PMC11526627 DOI: 10.1186/s12883-024-03926-3] [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: 06/28/2024] [Accepted: 10/21/2024] [Indexed: 11/02/2024] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disorder caused by chronic inflammatory reactions in the central nervous system. Currently, little is known about the changes of plasma proteomic profiles in Chinese patients with MS (CpwMS) and its relationship with the altered profiles of multi-omics such as metabolomics and gut microbiome, as well as potential molecular networks that underlie the etiology of MS. To uncover the characteristics of proteomics landscape and potential multi-omics interaction networks in CpwMS, Plasma samples were collected from 22 CpwMS and 22 healthy controls (HCs) and analyzed using a Tandem Mass Tag (TMT)-based quantitative proteomics approach. Our results showed that the plasma proteomics pattern was significantly different in CpwMS compared to HCs. A total of 90 differentially expressed proteins (DEPs), such as LAMP1 and FCG2A, were identified in CpwMS plasma comparing to HCs. Furthermore, we also observed extensive and significant correlations between the altered proteomic profiles and the changes of metabolome, gut microbiome, as well as altered immunoinflammatory responses in MS-affected patients. For instance, the level of LAMP1 and ERN1 were significantly and positively correlated with the concentrations of metabolite L-glutamic acid and pro-inflammatory factor IL-17 (Padj < 0.05). However, they were negatively correlated with the amounts of other metabolites such as L-tyrosine and sphingosine 1-phosphate, as well as the concentrations of IL-8 and MIP-1α. This study outlined the underlying multi-omics integrated mechanisms that might regulate peripheral immunoinflammatory responses and MS progression. These findings are potentially helpful for developing new assisting diagnostic biomarker and therapeutic strategies for MS.
Collapse
Affiliation(s)
- Fan Yang
- Lishui Key Laboratory of Brain Health and Severe Brain Disorders, Department of Rehabilitation & Clinical Laboratory, Lishui Second People's Hospital, Lishui, China.
- Key Laboratory of Cell Engineering in Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, China.
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China.
| | - Long-You Zhao
- Lishui Key Laboratory of Brain Health and Severe Brain Disorders, Department of Rehabilitation & Clinical Laboratory, Lishui Second People's Hospital, Lishui, China
| | - Wen-Qi Yang
- Department of Clinical Laboratory & Gastrointestinal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Shan Chao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Zong-Xin Ling
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Bo-Yao Sun
- Department of Clinical Laboratory & Gastrointestinal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Li-Ping Wei
- Department of Clinical Laboratory & Gastrointestinal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Li-Juan Zhang
- Lishui Key Laboratory of Brain Health and Severe Brain Disorders, Department of Rehabilitation & Clinical Laboratory, Lishui Second People's Hospital, Lishui, China
| | - Li-Mei Yu
- Key Laboratory of Cell Engineering in Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, China.
| | - Guang-Yong Cai
- Lishui Key Laboratory of Brain Health and Severe Brain Disorders, Department of Rehabilitation & Clinical Laboratory, Lishui Second People's Hospital, Lishui, China.
| |
Collapse
|
21
|
Airaghi Leccardi MJI, Desbiolles BXE, Haddad AY, Joy BC, Song C, Sarkar D. Light-induced rolling of azobenzene polymer thin films for wrapping subcellular neuronal structures. Commun Chem 2024; 7:249. [PMID: 39478057 PMCID: PMC11525480 DOI: 10.1038/s42004-024-01335-8] [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: 08/11/2024] [Accepted: 10/18/2024] [Indexed: 11/02/2024] Open
Abstract
Neurons are essential cells composing our nervous system and orchestrating our body, thoughts, and emotions. Recently, research efforts have been focused on studying not only their collective structure and functions but also the single-cell properties as an individual complex system. Nanoscale technology has the potential to unravel mysteries in neuroscience and provide support to the neuron by measuring and influencing several aspects of the cell. As wearable devices interact with different parts of our body, we could envision a thousand times smaller interface to conform on and around subcellular regions of the neurons for unprecedented contact, probing, and control. However, a major challenge is to develop an interface that can morph to the extreme curvatures of subcellular structures. Here, we address this challenge with the development of a platform that conforms even to small neuronal processes. To achieve this, we produced a wireless platform made of an azobenzene polymer that undergoes on-demand light-induced folding with sub-micrometer radius of curvature. We show that these platforms can be fabricated with an adjustable form factor, micro-injected onto neuronal cultures, and can delicately wrap various morphologies of neuronal processes in vitro, toward obtaining seamless biointerfaces with an increased coupling with the cell membrane. Our in vitro testings did not show any adverse effects of the platforms in contact with the neurons. Additionally, for future functionality, nanoparticles or optoelectronic materials could be blended with the azobenzene polymer, and 2D materials on the platform surface could be safely folded to the high curvatures without mechanical failure, as per our calculations. Ultimately, this technology could lay the foundation for future integration of wirelessly actuated materials within or on its platform for neuromodulation, recording, and neuroprotection at the subcellular level.
Collapse
Affiliation(s)
| | | | - Anna Y Haddad
- MIT Media Lab, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Baju C Joy
- MIT Media Lab, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Chen Song
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Deblina Sarkar
- MIT Media Lab, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
22
|
Gadani SP, Singh S, Kim S, Hu J, Smith MD, Calabresi PA, Bhargava P. Spatial transcriptomics of meningeal inflammation reveals inflammatory gene signatures in adjacent brain parenchyma. eLife 2024; 12:RP88414. [PMID: 39475792 PMCID: PMC11524578 DOI: 10.7554/elife.88414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2024] Open
Abstract
While modern high efficacy disease modifying therapies have revolutionized the treatment of relapsing-remitting multiple sclerosis, they are less effective at controlling progressive forms of the disease. Meningeal inflammation is a recognized risk factor for cortical gray matter pathology which can result in disabling symptoms such as cognitive impairment and depression, but the mechanisms linking meningeal inflammation and gray matter pathology remain unclear. Here, we performed magnetic resonance imaging (MRI)-guided spatial transcriptomics in a mouse model of autoimmune meningeal inflammation to characterize the transcriptional signature in areas of meningeal inflammation and the underlying brain parenchyma. We found broadly increased activity of inflammatory signaling pathways at sites of meningeal inflammation, but only a subset of these pathways active in the adjacent brain parenchyma. Subclustering of regions adjacent to meningeal inflammation revealed the subset of immune programs induced in brain parenchyma, notably complement signaling and antigen processing/presentation. Trajectory gene and gene set modeling analysis confirmed variable penetration of immune signatures originating from meningeal inflammation into the adjacent brain tissue. This work contributes a valuable data resource to the field, provides the first detailed spatial transcriptomic characterization in a model of meningeal inflammation, and highlights several candidate pathways in the pathogenesis of gray matter pathology.
Collapse
Affiliation(s)
- Sachin P Gadani
- Division of Neuroimmunology, Department of Neurology, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Neurology, University of PittsburghPittsburghUnited States
| | - Saumitra Singh
- Division of Neuroimmunology, Department of Neurology, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Sophia Kim
- Division of Neuroimmunology, Department of Neurology, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Jingwen Hu
- Division of Neuroimmunology, Department of Neurology, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Matthew D Smith
- Division of Neuroimmunology, Department of Neurology, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Peter A Calabresi
- Division of Neuroimmunology, Department of Neurology, Johns Hopkins University School of MedicineBaltimoreUnited States
- Solomon Snyder, Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Pavan Bhargava
- Division of Neuroimmunology, Department of Neurology, Johns Hopkins University School of MedicineBaltimoreUnited States
| |
Collapse
|
23
|
Hurani M, Lavi I, Bloch S, Golan D. The prevalence of multiple sclerosis in Israel based on validation of a health care organization database. Sci Rep 2024; 14:25070. [PMID: 39443627 PMCID: PMC11499638 DOI: 10.1038/s41598-024-76282-4] [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: 06/12/2024] [Accepted: 10/11/2024] [Indexed: 10/25/2024] Open
Abstract
In Israel there is no registry of multiple sclerosis (MS), thus the prevalence of MS is unknown. Clalit Health Services (CHS) is Israel's largest health care organization. We have developed an algorithm to identify people with MS (PwMS) using CHS database and validated it against a gold-standard diagnosis by expert neurologists. People with a possible diagnosis of MS were identified in CHS database according to ICD-9 code or dispense of MS specific disease modifying treatment (DMT). The electronic medical records (EMRs) of a random sample of 25% of this population, stratified by age and sex were examined to determine positive predictive value (PPV). Another age- and sex- stratified random sample of 15% of pwMS under our care were searched in CHS database to determine database sensitivity (Sn). Finally, a random sample of 40% of people without MS were searched in CHS database to evaluate database specificity (Sp). The best case definition to retrieve pwMS was ICD-9 code 340 as an established diagnosis or at least one dispense of a DMT (PPV = 87%, Sn = 92%, Sp = 90%). Using this definition the prevalence of MS in Israel was 68, 82 and 95 per 100,000 population by the end of 2011, 2016 and 2021, respectively. The prevalence of MS in Israel is rising, in line with the worldwide trend.
Collapse
Affiliation(s)
- Maha Hurani
- Multiple Sclerosis and Neuroimmunology Center, Department of Neurology, Lady Davis Carmel Medical Center, 7 Mikhal St, Haifa, 3436212, Israel
| | - Idit Lavi
- Department of Community Medicine and Epidemiology, Lady Davis Carmel Medical Center, Haifa, Israel
| | - Sivan Bloch
- Multiple Sclerosis and Neuroimmunology Center, Department of Neurology, Lady Davis Carmel Medical Center, 7 Mikhal St, Haifa, 3436212, Israel
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Daniel Golan
- Multiple Sclerosis and Neuroimmunology Center, Department of Neurology, Lady Davis Carmel Medical Center, 7 Mikhal St, Haifa, 3436212, Israel.
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
24
|
Sokolowski I, Kucharska-Lusina A, Miller E, Poplawski T, Majsterek I. Exploring the Gene Expression and Plasma Protein Levels of HSP90, HSP60, and GDNF in Multiple Sclerosis Patients and Healthy Controls. Curr Issues Mol Biol 2024; 46:11668-11680. [PMID: 39451573 PMCID: PMC11505768 DOI: 10.3390/cimb46100693] [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: 09/24/2024] [Revised: 10/12/2024] [Accepted: 10/16/2024] [Indexed: 10/26/2024] Open
Abstract
Multiple sclerosis (MS) is a chronic neurodegenerative disease characterized by immune-mediated inflammation and neurodegeneration in the central nervous system (CNS). In this study; we aimed to investigate the gene expression and plasma protein levels of three neuroprotective genes-heat shock proteins (HSP90 and HSP60) and glial cell line-derived neurotrophic factor (GDNF)-in MS patients compared to healthy controls. Forty patients with relapsing-remitting MS and 40 healthy volunteers participated in this study. Gene expression was measured using reverse transcription quantitative real-time PCR, and protein levels were assessed via ELISA. The results showed a significant increase in HSP90 (1.7-fold) and HSP60 (2-fold) gene expression in MS patients compared to controls, along with corresponding increases in protein levels (1.5-fold for both HSP90 and HSP60). In contrast, GDNF gene expression and protein levels were significantly reduced in MS patients, with a 7-fold decrease in gene expression and a 1.6-fold reduction in protein levels. Notably, a non-linear relationship between GDNF gene expression and protein concentration was observed in MS patients, suggesting complex regulatory mechanisms influencing GDNF in the disease. The upregulation of HSP90 and HSP60 in MS highlights their roles in immune regulation and stress responses, while the reduction in GDNF indicates impaired neuroprotection. These findings suggest that HSP90, HSP60, and GDNF could serve as biomarkers for disease progression and as potential therapeutic targets in MS, offering promising avenues for future research and treatment development.
Collapse
Affiliation(s)
- Igor Sokolowski
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (I.S.); (A.K.-L.); (I.M.)
| | - Aleksandra Kucharska-Lusina
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (I.S.); (A.K.-L.); (I.M.)
| | - Elzbieta Miller
- Department of Neurological Rehabilitation, Medical University of Lodz, Milionowa 14, 93-113 Lodz, Poland;
| | - Tomasz Poplawski
- Department of Microbiology and Pharmaceutical Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland
| | - Ireneusz Majsterek
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (I.S.); (A.K.-L.); (I.M.)
| |
Collapse
|
25
|
Takano T, Takano C, Funakoshi H, Bando Y. Impact of Neuron-Derived HGF on c-Met and KAI-1 in CNS Glial Cells: Implications for Multiple Sclerosis Pathology. Int J Mol Sci 2024; 25:11261. [PMID: 39457044 PMCID: PMC11509024 DOI: 10.3390/ijms252011261] [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: 08/28/2024] [Revised: 10/10/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Demyelination and axonal degeneration are fundamental pathological characteristics of multiple sclerosis (MS), an inflammatory disease of the central nervous system (CNS). Although the molecular mechanisms driving these processes are not fully understood, hepatocyte growth factor (HGF) has emerged as a potential regulator of neuroinflammation and tissue protection in MS. Elevated HGF levels have been reported in MS patients receiving immunomodulatory therapy, indicating its relevance in disease modulation. This study investigated HGF's neuroprotective effects using transgenic mice that overexpressed HGF. The experimental autoimmune encephalomyelitis (EAE) model, which mimics MS pathology, was employed to assess demyelination and axonal damage in the CNS. HGF transgenic mice showed delayed EAE progression, with reduced CNS inflammation, decreased demyelination, and limited axonal degeneration. Scanning electron microscopy confirmed the preservation of myelin and axonal integrity in these mice. In addition, we explored HGF's effects using a cuprizone-induced demyelination model, which operates independently of the immune system. HGF transgenic mice exhibited significant protection against demyelination in this model as well. We also investigated the expression of key HGF receptors, particularly c-Met and KAI-1. While c-Met, which is associated with increased inflammation, was upregulated in EAE, its expression was significantly reduced in HGF transgenic mice, correlating with decreased neuroinflammation. Conversely, KAI-1, which has been linked to axonal protection and stability, showed enhanced expression in HGF transgenic mice, suggesting a protective mechanism against axonal degeneration. These findings underscore HGF's potential in preserving CNS structure and function, suggesting it may be a promising therapeutic target for MS, offering new hope for mitigating disease progression and enhancing neuroprotection.
Collapse
Affiliation(s)
- Takuma Takano
- Department of Functional Anatomy and Neuroscience, Asahikawa Medical University, Asahikawa 078-8510, Japan
- Department of Neurosurgery, Asahikawa Medical University, Asahikawa 078-8510, Japan
| | - Chie Takano
- Department of Functional Anatomy and Neuroscience, Asahikawa Medical University, Asahikawa 078-8510, Japan
- Department of Neurosurgery, Asahikawa Medical University, Asahikawa 078-8510, Japan
| | - Hiroshi Funakoshi
- Department of Advanced Medical Science, Asahikawa Medical University, Asahikawa 078-8510, Japan
| | - Yoshio Bando
- Department of Functional Anatomy and Neuroscience, Asahikawa Medical University, Asahikawa 078-8510, Japan
- Department of Anatomy, Akita University Graduate School of Medicine, Akita 010-08543, Japan
| |
Collapse
|
26
|
Arrambide G, Comabella M, Tur C. Big data and artificial intelligence applied to blood and CSF fluid biomarkers in multiple sclerosis. Front Immunol 2024; 15:1459502. [PMID: 39493759 PMCID: PMC11527669 DOI: 10.3389/fimmu.2024.1459502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 09/30/2024] [Indexed: 11/05/2024] Open
Abstract
Artificial intelligence (AI) has meant a turning point in data analysis, allowing predictions of unseen outcomes with precedented levels of accuracy. In multiple sclerosis (MS), a chronic inflammatory-demyelinating condition of the central nervous system with a complex pathogenesis and potentially devastating consequences, AI-based models have shown promising preliminary results, especially when using neuroimaging data as model input or predictor variables. The application of AI-based methodologies to serum/blood and CSF biomarkers has been less explored, according to the literature, despite its great potential. In this review, we aimed to investigate and summarise the recent advances in AI methods applied to body fluid biomarkers in MS, highlighting the key features of the most representative studies, while illustrating their limitations and future directions.
Collapse
Affiliation(s)
- Georgina Arrambide
- Multiple Sclerosis Centre of Catalonia (Cemcat), Department of Neurology, Hospital
Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | - Carmen Tur
- Multiple Sclerosis Centre of Catalonia (Cemcat), Department of Neurology, Hospital
Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
27
|
Ladakis DC, Harrison KL, Smith MD, Solem K, Gadani S, Jank L, Hwang S, Farhadi F, Dewey BE, Fitzgerald KC, Sotirchos ES, Saidha S, Calabresi PA, Bhargava P. Bile acid metabolites predict multiple sclerosis progression and supplementation is safe in progressive disease. MED 2024:S2666-6340(24)00378-7. [PMID: 39447576 DOI: 10.1016/j.medj.2024.09.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 07/31/2024] [Accepted: 09/25/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND Bile acid metabolism is altered in multiple sclerosis (MS) and tauroursodeoxycholic acid (TUDCA) supplementation ameliorated disease in mouse models of MS. METHODS Global metabolomics was performed in an observational cohort of people with MS, followed by pathway analysis to examine relationships between baseline metabolite levels and subsequent brain and retinal atrophy. A double-blind, placebo-controlled trial was completed in people with progressive MS (PMS), randomized to receive either TUDCA (2 g/day) or placebo for 16 weeks. Participants were followed with serial clinical and laboratory assessments. Primary outcomes were safety and tolerability of TUDCA, and exploratory outcomes included changes in clinical, laboratory, and gut microbiome parameters. FINDINGS In the observational cohort, higher primary bile acid levels at baseline predicted slower whole-brain atrophy, brain substructure atrophy, and specific retinal layer atrophy. In the clinical trial, 47 participants were included in our analyses (21 in placebo arm, 26 in TUDCA arm). Adverse events did not differ significantly between arms (p = 0.77). The TUDCA arm demonstrated increased serum levels of multiple bile acids. No significant differences were noted in clinical or fluid biomarker outcomes. Central memory CD4+ and Th1/17 cells decreased, while CD4+ naive cells increased in the TUDCA arm compared to placebo. Changes in the composition and function of gut microbiota were also noted between the two groups. CONCLUSIONS Bile acid metabolism in MS is linked to brain and retinal atrophy. TUDCA supplementation in PMS is safe, tolerable, and has measurable biological effects that warrant further evaluation in larger trials with a longer treatment duration. FUNDING National MS Society grant RG-1707-28601 to P.B., R01 NS082347 from the National Institute of Neurological Disorders and Stroke to P.A.C., and National MS Society grant RG-1606-08768 to S.S.
Collapse
Affiliation(s)
- Dimitrios C Ladakis
- Johns Hopkins University School of Medicine, Department of Neurology, Baltimore, MD 21205, USA
| | - Kimystian L Harrison
- Johns Hopkins University School of Medicine, Department of Neurology, Baltimore, MD 21205, USA
| | - Matthew D Smith
- Johns Hopkins University School of Medicine, Department of Neurology, Baltimore, MD 21205, USA
| | - Krista Solem
- Johns Hopkins University School of Medicine, Department of Neurology, Baltimore, MD 21205, USA
| | - Sachin Gadani
- Johns Hopkins University School of Medicine, Department of Neurology, Baltimore, MD 21205, USA
| | - Larissa Jank
- Johns Hopkins University School of Medicine, Department of Neurology, Baltimore, MD 21205, USA
| | - Soonmyung Hwang
- Johns Hopkins University School of Medicine, Department of Neurology, Baltimore, MD 21205, USA
| | - Farzaneh Farhadi
- Johns Hopkins University School of Medicine, Department of Neurology, Baltimore, MD 21205, USA
| | - Blake E Dewey
- Johns Hopkins University School of Medicine, Department of Neurology, Baltimore, MD 21205, USA
| | - Kathryn C Fitzgerald
- Johns Hopkins University School of Medicine, Department of Neurology, Baltimore, MD 21205, USA
| | - Elias S Sotirchos
- Johns Hopkins University School of Medicine, Department of Neurology, Baltimore, MD 21205, USA
| | - Shiv Saidha
- Johns Hopkins University School of Medicine, Department of Neurology, Baltimore, MD 21205, USA
| | - Peter A Calabresi
- Johns Hopkins University School of Medicine, Department of Neurology, Baltimore, MD 21205, USA
| | - Pavan Bhargava
- Johns Hopkins University School of Medicine, Department of Neurology, Baltimore, MD 21205, USA.
| |
Collapse
|
28
|
Fung WH, van Lingen MR, Broos JY, Lam KH, van Dam M, Fung WK, Noteboom S, Koubiyr I, de Vries HE, Jasperse B, Teunissen CE, Giera M, Killestein J, Hulst HE, Strijbis EMM, Schoonheim MM, Kooij G. 9-HODE associates with thalamic atrophy and predicts white matter damage in multiple sclerosis. Mult Scler Relat Disord 2024; 92:105946. [PMID: 39447246 DOI: 10.1016/j.msard.2024.105946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 10/11/2024] [Accepted: 10/15/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND Multiple sclerosis (MS) is characterized by extensive tissue damage leading to a range of complex symptoms, including physical disability and cognitive dysfunction. Recent work has indicated the clinical relevance of bioactive lipid mediators (LMs), which are known to orchestrate inflammation and its resolution and are deregulated in MS. However, it is unknown whether LM profiles relate to white matter (WM) damage. OBJECTIVES To investigate the potential association between plasma-derived LMs and MRI-quantified WM damage using fractional anisotropy (FA) and grey matter (GM) atrophy in dimethyl fumarate-treated relapsing remitting MS (RRMS) patients. METHODS Severity of FA-based WM damage and GM atrophy was determined in RRMS patients (n = 28) compared to age- and sex-matched controls (n = 31) at treatment initiation (baseline) and after 6 months. Plasma LMs were assessed using HPLC-MS/MS and baseline LMs were correlated to changes in FA and brain volumes. RESULTS We observed significant WM damage in RRMS patients (mean age 41.4 [SD 9.1]) at baseline and follow-up (z-score=-0.33 and 0.31, respectively) compared to controls (mean age 41.9 [SD 9.5]; p < 0.001 for both comparisons). Patients with severe WM damage showed a decline of thalamic volume (p = 0.02), and this decline correlated (r = 0.51, p < 0.001) with lower baseline levels of 9-HODE. This LM also predicted FA worsening (beta = 0.14, p < 0.001) over time at 6 months. CONCLUSION Despite the relatively small sample size, lower baseline levels of the LM 9-HODE correlated with more thalamic atrophy and predicted subsequent worsening of WM damage in RRMS patients.
Collapse
Affiliation(s)
- Wing Hee Fung
- MS Center Amsterdam, Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands; MS Center Amsterdam, Molecular Cell Biology and Immunology, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, the Netherlands.
| | - Marike R van Lingen
- MS Center Amsterdam, Anatomy & Neurosciences, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Jelle Y Broos
- MS Center Amsterdam, Molecular Cell Biology and Immunology, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, the Netherlands; Leiden University Medical Centre (LUMC), Center of Proteomics and Metabolomics, Leiden, the Netherlands
| | - Ka-Hoo Lam
- MS Center Amsterdam, Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Maureen van Dam
- MS Center Amsterdam, Anatomy & Neurosciences, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Wing Ka Fung
- MS Center Amsterdam, Molecular Cell Biology and Immunology, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, the Netherlands
| | - Samantha Noteboom
- MS Center Amsterdam, Anatomy & Neurosciences, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Ismail Koubiyr
- MS Center Amsterdam, Anatomy & Neurosciences, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Helga E de Vries
- MS Center Amsterdam, Molecular Cell Biology and Immunology, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, the Netherlands
| | - Bas Jasperse
- MS Center Amsterdam, Radiology & Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
| | - Charlotte E Teunissen
- MS Center Amsterdam, Neurochemistry Laboratory, Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, the Netherlands
| | - Martin Giera
- Department of Medical, Health and Neuropsychology, Leiden University, Leiden, the Netherlands
| | - Joep Killestein
- MS Center Amsterdam, Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Hanneke E Hulst
- Department of Medical, Health and Neuropsychology, Leiden University, Leiden, the Netherlands
| | - Eva M M Strijbis
- MS Center Amsterdam, Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Menno M Schoonheim
- MS Center Amsterdam, Anatomy & Neurosciences, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Gijs Kooij
- MS Center Amsterdam, Molecular Cell Biology and Immunology, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, Amsterdam UMC, Amsterdam, the Netherlands
| |
Collapse
|
29
|
Ghezelhesari EM, Heydari A, Ebrahimipour H, Nahayati MA, Khadivzadeh T. Meta-synthesis of the motherhood-related needs of women with multiple sclerosis. BMC Womens Health 2024; 24:559. [PMID: 39396006 PMCID: PMC11470578 DOI: 10.1186/s12905-024-03397-1] [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: 11/22/2023] [Accepted: 09/30/2024] [Indexed: 10/14/2024] Open
Abstract
BACKGROUND The treatment of multiple sclerosis, an autoimmune disease that predominantly affects women of reproductive age, has undergone considerable advancements. Nevertheless, their unique needs, particularly those related to motherhood, continue to be disregarded. This study aims to ascertain the needs of women diagnosed with MS about motherhood. METHODS Using the Noblit and Hare methods, meta-synthesis research was conducted on 10 qualitative studies. The relevant studies were obtained by conducting a comprehensive search using keywords in the PubMed, Scopus, Web of Science (ISI), and ProQuest databases without any time limitations until June 2023. The CASP checklist was used to assess the quality of the articles, while the technique of Brown and Clark (2006) was utilized for data synthesis. RESULTS Four key themes emerged from this meta-synthesis, highlighting the significance of a support system, psychoeducational counseling, knowledge promotion, and continuous, comprehensive, and unique care. CONCLUSION Identifying the needs of women diagnosed with MS and addressing these needs concerning motherhood can enhance the quality of services rendered and enhance women's contentment with their choice to embark on motherhood. Furthermore, recognizing these needs during subsequent stages can be utilized in formulating tailored programs catering to this specific cohort of women.
Collapse
Affiliation(s)
- Elaheh Mansouri Ghezelhesari
- Candidate of Reproductive Health, Student Research Committee, Faculty of Nursing and Midwifery, Mashhad University of Medical Science, Mashhad, Iran
| | - Abbas Heydari
- Nursing and Midwifery Care Research Center, School of Nursing and Midwifery, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hosein Ebrahimipour
- Department of Health Economics and Management, School of Health, Mashhad University of Medical Sciences, Mashhad, Iran
- Health Sciences Research Center, Torbat Heydarieh University of Medical Sciences, Torbat Heydarieh, Iran
| | - Mohammad Ali Nahayati
- Department of Neurology, Ghaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Talat Khadivzadeh
- Nursing and Midwifery Care Research Center, School of Nursing and Midwifery, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Midwifery, School of Nursing and Midwifery, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
30
|
Touil H, Luquez T, Comandante-Lou N, Lee AJ, Fujita M, Habeck C, Kroshilina A, Hegewisch-Solloa E, McInvale J, Zuroff L, Isnard S, Walker E, Zhang L, Routy JP, Zhang Y, Mace EM, Klotz L, Wiendl H, Xia Z, Bar-Or A, Menon V, Stern Y, De Jager PL. Relation of CMV and brain atrophy to trajectories of immunosenescence in diverse populations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.07.614568. [PMID: 39416188 PMCID: PMC11482892 DOI: 10.1101/2024.10.07.614568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Immunosenescence (ISC), the aging of the immune system, has largely been studied in populations of European descent. Here, circulating immune cell cytometric data from African-American, Hispanic, and non-Hispanic White participants were generated. Known and novel age effects were identified using either a meta-analysis approach or a parallel genetic approach. Most results are consistent across the three populations, but some cell populations display evidence of heterogeneity, such as a PD-L1 + CD56 + NK cell subset. The study estimated "Immunological Age" (IA) during physiologic aging. While we found no relation of IA to Multiple Sclerosis, IA is associated with entorhinal cortex atrophy, a presymptomatic feature of Alzheimer's disease, linking neurodegeneration and peripheral immunity. ISC trajectories were also inferred, highlighting age, CMV status, and genetic ancestry as key influences. Our assessment offers reference ISC trajectories for personalization of assessments of immune function over the life course in diverse populations.
Collapse
|
31
|
Baller EB, Luo AC, Schindler MK, Cooper EC, Pecsok MK, Cieslak MC, Martin ML, Bar-Or A, Elahi A, Perrone CM, Reid D, Spangler BC, Satterthwaite TD, Shinohara RT. Association of Anxiety with Uncinate Fasciculus Lesion Burden in Multiple Sclerosis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.10.08.24315108. [PMID: 39417125 PMCID: PMC11482984 DOI: 10.1101/2024.10.08.24315108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Importance Multiple sclerosis (MS) is an immune-mediated neurological disorder that affects 2.4 million people world-wide, and up to 60% experience anxiety. Objective We investigated how anxiety in MS is associated with white matter lesion burden in the uncinate fasciculus (UF). Design Retrospective case-control study of participants who received research-quality 3-tesla (3T) neuroimaging as part of MS clinical care from 2010-2018. Analyses were performed from June 1st to September 30th, 2024. Setting Single-center academic medical specialty MS clinic. Participants Participants were identified from the electronic medical record. All participants were diagnosed by an MS specialist and completed research-quality MRI at 3T. After excluding participants with poor image quality, 372 were stratified into three groups which were balanced for age and sex: 1) MS without anxiety (MS+noA, n=99); 2) MS with mild anxiety (MS+mildA, n=249); and 3) MS with severe anxiety (MS+severeA, n=24). Exposure Anxiety diagnosis and anxiolytic medication. Main Outcome and Measure We first evaluated whether MS+severeA patients had greater lesion burden in the UF than MS+noA. Next, we examined whether increasing anxiety severity was associated with greater UF lesion burden. Generalized additive models were employed, with the burden of lesions (e.g. proportion of fascicle impacted) within the UF as the outcome measure and sex and spline of age as covariates. Results UF burden was higher in MS+severeA as compared to MS+noA (T=2.02, P=0.045, Cohen's f 2=0.19). A dose-response effect was also found, where higher mean UF burden was associated with higher anxiety severity (T=2.08, P=0.038, Cohen's f 2=0.10). Conclusions and Relevance We demonstrate that overall lesion burden in UF was associated with the presence and severity of anxiety in patients with MS. Future studies linking white matter lesion burden in UF with treatment prognosis are warranted.
Collapse
Affiliation(s)
- Erica B. Baller
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA USA
- Penn Lifespan Informatics and Neuroimaging Center (PennLINC), Philadelphia, PA USA
| | - Audrey C. Luo
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA USA
- Penn Lifespan Informatics and Neuroimaging Center (PennLINC), Philadelphia, PA USA
| | - Matthew K. Schindler
- Department of Neurology, University of Pennsylvania, Philadelphia, PA USA
- Center for Neuroinflammation and Neurotherapeutics, University of Pennsylvania, Philadelphia, PA USA
| | - Elena C. Cooper
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA USA
- Penn Lifespan Informatics and Neuroimaging Center (PennLINC), Philadelphia, PA USA
| | - Margaret K. Pecsok
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA USA
| | - Matthew C. Cieslak
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA USA
- Penn Lifespan Informatics and Neuroimaging Center (PennLINC), Philadelphia, PA USA
| | - Melissa Lynne Martin
- Penn Statistics in Imaging and Visualization Center (PennSIVE), Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA USA
| | - Amit Bar-Or
- Department of Neurology, University of Pennsylvania, Philadelphia, PA USA
- Center for Neuroinflammation and Neurotherapeutics, University of Pennsylvania, Philadelphia, PA USA
| | - Ameena Elahi
- Department of Information Services, University of Pennsylvania, Philadelphia, PA USA
| | - Christopher M. Perrone
- Department of Neurology, University of Pennsylvania, Philadelphia, PA USA
- Center for Neuroinflammation and Neurotherapeutics, University of Pennsylvania, Philadelphia, PA USA
| | - Donovan Reid
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA USA
| | - Bailey C. Spangler
- Penn Statistics in Imaging and Visualization Center (PennSIVE), Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA USA
| | - Theodore D. Satterthwaite
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA USA
- Penn Lifespan Informatics and Neuroimaging Center (PennLINC), Philadelphia, PA USA
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA USA
| | - Russell T. Shinohara
- Penn Statistics in Imaging and Visualization Center (PennSIVE), Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA USA
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA USA
| |
Collapse
|
32
|
Arslan MB, Öge-Daşdöğen Ö. Investigation of comparative nonword repetition performance in multiple sclerosis: Group differences, subtype variations, and disability effects. APPLIED NEUROPSYCHOLOGY. ADULT 2024:1-8. [PMID: 39382371 DOI: 10.1080/23279095.2024.2408393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
This study investigated Nonword Repetition (NWR) tasks in individuals with Multiple Sclerosis (MS) compared to healthy controls (HC), focusing on phonological working memory (WMP). Significant differences were found in NWR acurracy (NWRacc) score between MS subgroups and HC (H = 48.2, p < 0.001). NWRacc decreased as the number of syllables increased in both groups, indicating increased cognitive load. All MS subtypes showed lower NWRacc compared to HC across varying syllable lengths (Mann Whitney U Test: two syllables U = 64.5, p < 0.001; three syllables U = 183, p < 0.001; four syllables U = 248, p < 0.001; five syllables U = 283.5, p < 0.001). However, no significant differences were found within MS subtypes based on syllable length. NWRacc did not differ between mild and severe MS groups. Overall, the NWR test effectively assessed WMP in MS, highlighting its utility in diagnosing and addressing language-cognitive challenges in individuals with MS. This underscores the importance of tailored intervention strategies to mitigate these challenges.
Collapse
|
33
|
Garton T, Gadani SP, Gill AJ, Calabresi PA. Neurodegeneration and demyelination in multiple sclerosis. Neuron 2024; 112:3231-3251. [PMID: 38889714 PMCID: PMC11466705 DOI: 10.1016/j.neuron.2024.05.025] [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: 03/07/2024] [Revised: 05/20/2024] [Accepted: 05/23/2024] [Indexed: 06/20/2024]
Abstract
Progressive multiple sclerosis (PMS) is an immune-initiated neurodegenerative condition that lacks effective therapies. Although peripheral immune infiltration is a hallmark of relapsing-remitting MS (RRMS), PMS is associated with chronic, tissue-restricted inflammation and disease-associated reactive glial states. The effector functions of disease-associated microglia, astrocytes, and oligodendrocyte lineage cells are beginning to be defined, and recent studies have made significant progress in uncovering their pathologic implications. In this review, we discuss the immune-glia interactions that underlie demyelination, failed remyelination, and neurodegeneration with a focus on PMS. We highlight the common and divergent immune mechanisms by which glial cells acquire disease-associated phenotypes. Finally, we discuss recent advances that have revealed promising novel therapeutic targets for the treatment of PMS and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Thomas Garton
- Division of Neuroimmunology, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sachin P Gadani
- Division of Neuroimmunology, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alexander J Gill
- Division of Neuroimmunology, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peter A Calabresi
- Division of Neuroimmunology, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
34
|
Lu Q, Ma J, Zhao Y, Ding G, Wang Y, Qiao X, Cheng X. Disruption of blood-brain barrier and endothelial-to-mesenchymal transition are attenuated by Astragalus polysaccharides mediated through upregulation of ETS1 expression in experimental autoimmune encephalomyelitis. Biomed Pharmacother 2024; 180:117521. [PMID: 39383730 DOI: 10.1016/j.biopha.2024.117521] [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: 06/25/2024] [Revised: 09/27/2024] [Accepted: 10/04/2024] [Indexed: 10/11/2024] Open
Abstract
Blood-brain barrier (BBB) breakdown, an early hallmark of multiple sclerosis (MS), remains crucial for MS progression. Our previous works have confirmed that Astragalus polysaccharides (APS) can significantly ameliorate demyelination and disease progression in experimental autoimmune encephalomyelitis (EAE) mice. However, it remains unclear whether APS protects BBB and the potential mechanism. In this study, we found that APS effectively reduced BBB leakage in EAE mice, which was accompanied by a decreased level of endothelial-to-mesenchymal transition (EndoMT) in the central nervous system (CNS). We further induced EndoMT in the mouse brain endothelial cells (bEnd.3) by interleukin-1β (IL-1β) in vitro. The results showed that APS treatment could inhibit IL-1β-induced EndoMT and endothelial cell dysfunction. In addition, the transcription factor ETS1 is a central regulator of EndoMT related to the compromise of BBB. We tested the regulation of APS on ETS1 and identified the expression of ETS1 was upregulated in both EAE mice and bEnd.3 cells by APS. ETS1 knockdown facilitated EndoMT and endothelial cell dysfunction, which completely abolished the regulatory effect of APS. Collectively, APS treatment could protect BBB integrity by inhibiting EndoMT, which might be associated with upregulating ETS1 expression. Our findings indicated that APS has potential value in the prevention of MS.
Collapse
Affiliation(s)
- Qijin Lu
- Institute of Clinical Immunology, Yue-yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Jinyun Ma
- Institute of Clinical Immunology, Yue-yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Yan Zhao
- Institute of Clinical Immunology, Yue-yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Guiqing Ding
- Institute of Clinical Immunology, Yue-yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Yuanhua Wang
- Institute of Clinical Immunology, Yue-yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Xi Qiao
- Institute of Clinical Immunology, Yue-yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Xiaodong Cheng
- Institute of Clinical Immunology, Yue-yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China.
| |
Collapse
|
35
|
Vlad B, Neidhart S, Hilty M, Asplund Högelin K, Reichen I, Ziegler M, Khademi M, Lutterotti A, Regeniter A, Martin R, Al Nimer F, Jelcic I. Intrathecal immune reactivity against Measles-, Rubella-, and Varicella Zoster viruses is associated with cerebrospinal fluid inflammation in multiple sclerosis. Mult Scler 2024:13524585241279645. [PMID: 39377663 DOI: 10.1177/13524585241279645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/09/2024]
Abstract
BACKGROUND/OBJECTIVES We aimed to determine in multiple sclerosis (MS) whether intrathecal immunoglobulin G (IgG) production against measles- (M), rubella- (R), and varicella zoster (Z) viruses, which is called MRZ reaction (MRZR) and considered the most specific soluble biomarker for MS, is associated with demographic and basic cerebrospinal fluid (CSF) parameters reflecting inflammation. METHODS We analyzed the presence of positive MRZR and associations with demographic and clinical routine CSF parameters in 513 patients with MS and 182 non-MS patients. RESULTS Comparing MS patients versus non-MS patients, positive MRZR (38.8% versus 2.2%; specificity 97.8%; positive likelihood ratio, PLR 17.7) had a better specificity and PLR for MS than CSF-specific OCB (89.5% versus 22.0%; specificity 78.0%; PLR 4.1). A positive MRZR in MS patients was associated with female sex (p = 0.0001), pleocytosis (p < 0.0001), higher frequency of presence of plasma cells in CSF (p = 0.0248), normal CSF/serum albumin ratio (p = 0.0005), and intrathecal production of total IgG or CSF-specific OCB (both p < 0.0001), but not with intrathecal production of total IgA or IgM. CONCLUSIONS This study confirms the MRZR as a highly specific marker of MS and shows that MRZR-positive MS patients more frequently are female and show inflammatory changes of basic CSF parameters than MRZR-negative MS patients.
Collapse
Affiliation(s)
- Benjamin Vlad
- Neuroimmunology and Multiple Sclerosis Research Section, Department of Neurology, University of Zurich and University Hospital, Zurich, Switzerland
| | - Stephan Neidhart
- Neuroimmunology and Multiple Sclerosis Research Section, Department of Neurology, University of Zurich and University Hospital, Zurich, Switzerland; Swiss Epilepsy Center (Klinik Lengg), Zurich, Switzerland
| | - Marc Hilty
- Neuroimmunology and Multiple Sclerosis Research Section, Department of Neurology, University of Zurich and University Hospital, Zurich, Switzerland
- Department of Neurology, Hirslanden Klinik Zurich, Zurich, Switzerland
| | - Klara Asplund Högelin
- Center for Molecular Medicine, Neuroimmunology Unit, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Ina Reichen
- Neuroimmunology and Multiple Sclerosis Research Section, Department of Neurology, University of Zurich and University Hospital, Zurich, Switzerland
- Neuroimmunology Outpatient Clinic, Center for Multiple Sclerosis, Neurocenter Bellevue, Zurich, Switzerland
| | - Mario Ziegler
- Neuroimmunology and Multiple Sclerosis Research Section, Department of Neurology, University of Zurich and University Hospital, Zurich, Switzerland
| | - Mohsen Khademi
- Center for Molecular Medicine, Neuroimmunology Unit, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Andreas Lutterotti
- Neuroimmunology and Multiple Sclerosis Research Section, Department of Neurology, University of Zurich and University Hospital, Zurich, Switzerland
- Clinical Research Priority Program MS (CRPP), PrecisionMS of the University of Zurich, Zurich, Switzerland
- Neuroimmunology Outpatient Clinic, Center for Multiple Sclerosis, Neurocenter Bellevue, Zurich, Switzerland
| | - Axel Regeniter
- Infectious Disease Serology and Immunology, Medica Medizinische Laboratorien Dr. F. Kaeppeli AG, Zurich, Switzerland
| | - Roland Martin
- Neuroimmunology and Multiple Sclerosis Research Section, Department of Neurology, University of Zurich and University Hospital, Zurich, Switzerland
- Clinical Research Priority Program MS (CRPP), PrecisionMS of the University of Zurich, Zurich, Switzerland
- Center for Molecular Medicine, Therapeutic Immune Design Unit, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Faiez Al Nimer
- Center for Molecular Medicine, Neuroimmunology Unit, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Ilijas Jelcic
- Neuroimmunology and Multiple Sclerosis Research Section, Department of Neurology, University of Zurich and University Hospital, Zurich, Switzerland
- Clinical Research Priority Program MS (CRPP), PrecisionMS of the University of Zurich, Zurich, Switzerland
- Neuroimmunology Outpatient Clinic, Center for Multiple Sclerosis, Neurocenter Bellevue, Zurich, Switzerland
| |
Collapse
|
36
|
Gurski F, Shirvanchi K, Rajendran V, Rajendran R, Megalofonou FF, Böttiger G, Stadelmann C, Bhushan S, Ergün S, Karnati S, Berghoff M. Anti-inflammatory and remyelinating effects of fexagratinib in experimental multiple sclerosis. Br J Pharmacol 2024. [PMID: 39367768 DOI: 10.1111/bph.17341] [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: 04/25/2024] [Revised: 07/04/2024] [Accepted: 07/15/2024] [Indexed: 10/07/2024] Open
Abstract
BACKGROUND AND PURPOSE FGF, VEGFR-2 and CSF1R signalling pathways play a key role in the pathogenesis of multiple sclerosis (MS). Selective inhibition of FGFR by infigratinib in MOG35-55-induced experimental autoimmune encephalomyelitis (EAE) prevented severe first clinical episodes by 40%; inflammation and neurodegeneration were reduced, and remyelination was enhanced. Multi-kinase inhibition of FGFR1-3, CSFR and VEGFR-2 by fexagratinib (formerly known as AZD4547) may be more efficient in reducing inflammation, neurodegeneration and regeneration in the disease model. EXPERIMENTAL APPROACH Female C57BL/6J mice were treated with fexagratinib (6.25 or 12.5 mg·kg-1) orally or placebo over 10 days either from time of EAE induction (prevention experiment) or onset of symptoms (suppression experiment). Effects on inflammation, neurodegeneration and remyelination were assessed at the peak of the disease (Day 18/20 post immunization) and the chronic phase of EAE (Day 41/42). KEY RESULTS In the prevention experiment, treatment with 6.25 or 12.5 mg·kg-1 fexagratinib prevented severe first clinical episodes by 66.7% or 84.6% respectively. Mice treated with 12.5 mg·kg-1 fexagratinib hardly showed any symptoms in the chronic phase of EAE. In the suppression experiment, fexagratinib resulted in a long-lasting reduction of severe symptoms by 91 or 100%. Inflammation and demyelination were reduced, and axonal density, numbers of oligodendrocytes and their precursor cells, and remyelinated axons were increased by both experimental approaches. CONCLUSION AND IMPLICATIONS Multi-kinase inhibition by fexagratinib in a well-tolerated dose of 1 mg·kg-1 in humans may be a promising approach to reduce inflammation and neurodegeneration, to slow down disease progression and support remyelination in patients.
Collapse
Affiliation(s)
- Fynn Gurski
- Experimental Neurology, Department of Neurology, University of Giessen, Giessen, Germany
| | - Kian Shirvanchi
- Experimental Neurology, Department of Neurology, University of Giessen, Giessen, Germany
| | - Vinothkumar Rajendran
- Experimental Neurology, Department of Neurology, University of Giessen, Giessen, Germany
| | - Ranjithkumar Rajendran
- Experimental Neurology, Department of Neurology, University of Giessen, Giessen, Germany
| | | | - Gregor Böttiger
- Experimental Neurology, Department of Neurology, University of Giessen, Giessen, Germany
| | - Christine Stadelmann
- Institute of Neuropathology, University Medical Centre Göttingen, Göttingen, Germany
| | - Sudhanshu Bhushan
- Institute for Anatomy and Cell Biology, University of Giessen, Giessen, Germany
| | - Süleyman Ergün
- Institute of Anatomy and Cell Biology, University of Würzburg, Würzburg, Germany
| | - Srikanth Karnati
- Institute of Anatomy and Cell Biology, University of Würzburg, Würzburg, Germany
| | - Martin Berghoff
- Experimental Neurology, Department of Neurology, University of Giessen, Giessen, Germany
| |
Collapse
|
37
|
Song Y, Li J, Wu Y. Evolving understanding of autoimmune mechanisms and new therapeutic strategies of autoimmune disorders. Signal Transduct Target Ther 2024; 9:263. [PMID: 39362875 PMCID: PMC11452214 DOI: 10.1038/s41392-024-01952-8] [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: 02/20/2024] [Revised: 07/09/2024] [Accepted: 08/07/2024] [Indexed: 10/05/2024] Open
Abstract
Autoimmune disorders are characterized by aberrant T cell and B cell reactivity to the body's own components, resulting in tissue destruction and organ dysfunction. Autoimmune diseases affect a wide range of people in many parts of the world and have become one of the major concerns in public health. In recent years, there have been substantial progress in our understanding of the epidemiology, risk factors, pathogenesis and mechanisms of autoimmune diseases. Current approved therapeutic interventions for autoimmune diseases are mainly non-specific immunomodulators and may cause broad immunosuppression that leads to serious adverse effects. To overcome the limitations of immunosuppressive drugs in treating autoimmune diseases, precise and target-specific strategies are urgently needed. To date, significant advances have been made in our understanding of the mechanisms of immune tolerance, offering a new avenue for developing antigen-specific immunotherapies for autoimmune diseases. These antigen-specific approaches have shown great potential in various preclinical animal models and recently been evaluated in clinical trials. This review describes the common epidemiology, clinical manifestation and mechanisms of autoimmune diseases, with a focus on typical autoimmune diseases including multiple sclerosis, type 1 diabetes, rheumatoid arthritis, systemic lupus erythematosus, and sjögren's syndrome. We discuss the current therapeutics developed in this field, highlight the recent advances in the use of nanomaterials and mRNA vaccine techniques to induce antigen-specific immune tolerance.
Collapse
Affiliation(s)
- Yi Song
- Institute of Immunology, PLA, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jian Li
- Chongqing International Institute for Immunology, Chongqing, China.
| | - Yuzhang Wu
- Institute of Immunology, PLA, Third Military Medical University (Army Medical University), Chongqing, China.
- Chongqing International Institute for Immunology, Chongqing, China.
| |
Collapse
|
38
|
Miscioscia A, Mainero C, Treaba CA, Silvestri E, Scialpi G, Berardi A, Causin F, Anglani MG, Rinaldi F, Perini P, Puthenparampil M, Bertoldo A, Gallo P. The contribution of paramagnetic rim and cortical lesions to physical and cognitive disability at multiple sclerosis clinical onset: evaluating the power of MRI and OCT biomarkers. J Neurol 2024; 271:6702-6714. [PMID: 39155316 DOI: 10.1007/s00415-024-12622-8] [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: 06/12/2024] [Revised: 07/31/2024] [Accepted: 08/02/2024] [Indexed: 08/20/2024]
Abstract
BACKGROUND In multiple sclerosis (MS), imaging biomarkers play a crucial role in characterizing the disease at the time of diagnosis. MRI and optical coherence tomography (OCT) provide readily available biomarkers that may help to define the patient's clinical profile. However, the evaluation of cortical and paramagnetic rim lesions (CL, PRL), as well as retinal atrophy, is not routinely performed in clinic. OBJECTIVE To identify the most significant MRI and OCT biomarkers associated with early clinical disability in MS. METHODS Brain, spinal cord (SC) MRI, and OCT scans were acquired from 45 patients at MS diagnosis to obtain: brain PRL and non-PRL, CL, SC lesion volumes and counts, brain volumetric metrics, SC C2-C3 cross-sectional area, and retinal layer thickness. Regression models assessed relationships with physical disability (Expanded Disability Status Scale [EDSS]) and cognitive performance (Brief International Cognitive Assessment for Multiple Sclerosis [BICAMS]). RESULTS In a stepwise regression (R2 = 0.526), PRL (β = 0.001, p = 0.023) and SC lesion volumes (β = 0.001, p = 0.017) were the most significant predictors of EDSS, while CL volume and age were strongly associated with BICAMS scores. Moreover, in a model where PRL and non-PRL were pooled, only the contribution of SC lesion volume was retained in EDSS prediction. OCT measures did not show associations with disability at the onset. CONCLUSION At MS onset, PRL and SC lesions exhibit the strongest association with physical disability, while CL strongly contribute to cognitive performance. Incorporating the evaluation of PRL and CL into the initial MS patient assessment could help define their clinical profile, thus supporting the treatment choice.
Collapse
Affiliation(s)
- Alessandro Miscioscia
- A. A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Bldg 149, 13th Street, Charlestown, MA, 02129, USA.
- Department of Neuroscience, University of Padua, Padua, Italy.
- Multiple Sclerosis Centre of the Veneto Region (CeSMuV), Padua University Hospital, Padua, Italy.
| | - Caterina Mainero
- A. A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Bldg 149, 13th Street, Charlestown, MA, 02129, USA
- Harvard Medical School, Boston, MA, USA
| | - Constantina A Treaba
- A. A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Bldg 149, 13th Street, Charlestown, MA, 02129, USA
- Harvard Medical School, Boston, MA, USA
| | - Erica Silvestri
- Department of Information Engineering, University of Padua, Padua, Italy
| | - Graziana Scialpi
- Multiple Sclerosis Centre of the Veneto Region (CeSMuV), Padua University Hospital, Padua, Italy
| | - Angela Berardi
- Multiple Sclerosis Centre of the Veneto Region (CeSMuV), Padua University Hospital, Padua, Italy
| | | | | | - Francesca Rinaldi
- Multiple Sclerosis Centre of the Veneto Region (CeSMuV), Padua University Hospital, Padua, Italy
| | - Paola Perini
- Multiple Sclerosis Centre of the Veneto Region (CeSMuV), Padua University Hospital, Padua, Italy
| | - Marco Puthenparampil
- Department of Neuroscience, University of Padua, Padua, Italy
- Multiple Sclerosis Centre of the Veneto Region (CeSMuV), Padua University Hospital, Padua, Italy
| | | | - Paolo Gallo
- Department of Neuroscience, University of Padua, Padua, Italy
- Multiple Sclerosis Centre of the Veneto Region (CeSMuV), Padua University Hospital, Padua, Italy
| |
Collapse
|
39
|
Summers RA, Fagiani F, Rowitch DH, Absinta M, Reich DS. Novel human iPSC models of neuroinflammation in neurodegenerative disease and regenerative medicine. Trends Immunol 2024; 45:799-813. [PMID: 39307583 PMCID: PMC11471369 DOI: 10.1016/j.it.2024.08.004] [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: 07/27/2024] [Revised: 08/23/2024] [Accepted: 08/25/2024] [Indexed: 10/13/2024]
Abstract
The importance of neuroinflammation in neurodegenerative diseases is becoming increasingly evident, and, in parallel, human induced pluripotent stem cell (hiPSC) models of physiology and pathology are emerging. Here, we review new advancements in the differentiation of hiPSCs into glial, neural, and blood-brain barrier (BBB) cell types, and the integration of these cells into complex organoids and chimeras. These advancements are relevant for modeling neuroinflammation in the context of prevalent neurodegenerative disorders, such as Alzheimer's disease (AD), Parkinson's disease (PD), and multiple sclerosis (MS). With awareness of current limitations, recent progress in the development and application of various hiPSC-derived models shows potential for aiding the identification of candidate therapeutic targets and immunotherapy approaches.
Collapse
Affiliation(s)
- Rose Ana Summers
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK; Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Francesca Fagiani
- Translational Neuropathology Unit, Institute of Experimental Neurology, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - David H Rowitch
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Martina Absinta
- Translational Neuropathology Unit, Institute of Experimental Neurology, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Daniel S Reich
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
40
|
Olejnik P, Roszkowska Z, Adamus S, Kasarełło K. Multiple sclerosis: a narrative overview of current pharmacotherapies and emerging treatment prospects. Pharmacol Rep 2024; 76:926-943. [PMID: 39177889 PMCID: PMC11387431 DOI: 10.1007/s43440-024-00642-0] [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: 06/13/2024] [Revised: 08/20/2024] [Accepted: 08/20/2024] [Indexed: 08/24/2024]
Abstract
Multiple sclerosis (MS) is a chronic autoimmune disease characterized by pathological processes of demyelination, subsequent axonal loss, and neurodegeneration within the central nervous system. Despite the availability of numerous disease-modifying therapies that effectively manage this condition, there is an emerging need to identify novel therapeutic targets, particularly for progressive forms of MS. Based on contemporary insights into disease pathophysiology, ongoing efforts are directed toward developing innovative treatment modalities. Primarily, monoclonal antibodies have been extensively investigated for their efficacy in influencing specific pathological pathways not yet targeted. Emerging approaches emphasizing cellular mechanisms, such as chimeric antigen receptor T cell therapy targeting immunological cells, are attracting increasing interest. The evolving understanding of microglia and the involvement of ferroptotic mechanisms in MS pathogenesis presents further avenues for targeted therapies. Moreover, innovative treatment strategies extend beyond conventional approaches to encompass interventions that target alterations in microbiota composition and dietary modifications. These adjunctive therapies hold promise as complementary methods for the holistic management of MS. This narrative review aims to summarize current therapies and outline potential treatment methods for individuals with MS.
Collapse
Affiliation(s)
- Piotr Olejnik
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Zuzanna Roszkowska
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Sylwia Adamus
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
- Biomedical Physics Division, Faculty of Physics, University of Warsaw, Warsaw, Poland
| | - Kaja Kasarełło
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland.
| |
Collapse
|
41
|
Russell RD, He J, Black LJ, Begley A. Evaluating Experiences in a Digital Nutrition Education Program for People With Multiple Sclerosis: A Qualitative Study. Health Expect 2024; 27:e70012. [PMID: 39207907 PMCID: PMC11361266 DOI: 10.1111/hex.70012] [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: 04/08/2024] [Revised: 08/11/2024] [Accepted: 08/18/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Multiple sclerosis (MS) is a complex immune-mediated disease with no currently known cure. There is growing evidence to support the role of diet in reducing some of the symptoms and disease progression in MS, and we previously developed and tested the feasibility of a digital nutrition education program for people with MS. OBJECTIVE The aim of this study was to explore factors that influenced engagement in the digital nutrition education program, including features influencing capability, opportunity and motivation to change their dietary behaviours. METHODS Semi-structured interviews were conducted with people who had MS, and who completed some or all of the program until data saturation was reached. Interviews were analysed inductively using thematic analysis. Themes were deductively mapped against the COM-B (Capability, Opportunity, Motivation, Behaviour) behaviour change model. RESULTS Sixteen interviews were conducted with participants who completed all (n = 10) or some of the program (n = 6). Four themes emerged: (1) acquiring and validating nutrition knowledge; (2) influence of time and social support; (3) getting in early to improve health and (4) accounting for food literacy experiences. DISCUSSION This is the first online nutrition program with suitable behavioural supports for people with MS. It highlights the importance of disease-specific and evidence-based nutrition education to support people with MS to make dietary changes. Acquiring nutrition knowledge, coupled with practical support mechanisms, such as recipe booklets and goal setting, emerged as crucial for facilitating engagement with the program. CONCLUSIONS When designing education programs for people with MS and other neurological conditions, healthcare professionals and program designers should consider flexible delivery and building peer support to address the needs and challenges faced by participants. PATIENT OR PUBLIC CONTRIBUTION Members of the MS Nutrition Research Program Stakeholder Reference Group, which includes people with MS and MS health professionals, provided input during the development of the nutrition education program and study design stages.
Collapse
Affiliation(s)
- R. D. Russell
- Curtin School of Population HealthCurtin UniversityPerthAustralia
| | - J. He
- Curtin School of Population HealthCurtin UniversityPerthAustralia
| | - L. J. Black
- Curtin School of Population HealthCurtin UniversityPerthAustralia
- School of Exercise and Nutrition Sciences, Institute for Physical Activity and Nutrition (IPAN)Deakin UniversityMelbourneAustralia
| | - A. Begley
- Curtin School of Population HealthCurtin UniversityPerthAustralia
| |
Collapse
|
42
|
Yazdan Panah M, Vaheb S, Moases Ghaffary E, Shaygannejad V, Zabeti A, Mirmosayyeb O. Bone loss and fracture in people with multiple sclerosis: A systematic review and meta-analysis. Mult Scler Relat Disord 2024; 90:105773. [PMID: 39068819 DOI: 10.1016/j.msard.2024.105773] [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: 05/22/2024] [Revised: 06/29/2024] [Accepted: 07/14/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND People with multiple sclerosis (PwMS) exhibit reduced bone mineral density (BMD) across several anatomical regions. Studies have indicated that PwMS are at a heightened risk of fractures due to decreased BMD and increased prevalence of osteopenia and osteoporosis. This study aimed to investigate the prevalence and risk of osteopenia, osteoporosis, and fracture among PwMS. METHODS Relevant studies were identified through comprehensive searches of databases (PubMed/MEDLINE, Scopus, Embase, and Web of Science) from January 1, 2000, to January 21, 2024. R software version 4.4.0 and random-effects models were employed to estimate the pooled prevalence, odds ratio (OR), and risk ratio (RR) of osteopenia, osteoporosis, and fracture among PwMS, along with their respective 95 % confidence intervals (CIs). RESULTS From a total of 2039 articles, 51 studies with 1,503,785 PwMS met our inclusion criteria. The pooled prevalence of osteopenia, osteoporosis, and overall fracture among PwMS was 41.41 % (95 % CI: 36.14% to 46.69 %, I2=97 %), 14.21 % (95 % CI: 10.75 % to 17.68 %, I2=99 %), and 12.84 % (95 % CI: 8.49 % to 17.19 %, I2 = 100 %), respectively. The likelihood of osteopenia (OR=2.02, 95 % CI: 1.46 to 2.8, p-value<0.01, I2=17 %) and osteoporosis (OR=1.71, 95 % CI: 1.27 to 2.31, p-value<0.01, I2=74 %), as well as the probability of overall fracture (RR=1.86, 95 % CI: 1.61 to 2.14, p-value<0.01, I2=74 %) were significantly higher in PwMS than healthy controls (HCs). CONCLUSION PwMS were at a substantially increased risk of developing osteopenia (2-fold), osteoporosis (1.7-fold), and overall fractures (1.9-fold). Well-designed studies are needed to explore these associations further.
Collapse
Affiliation(s)
- Mohammad Yazdan Panah
- Student Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran; Isfahan Neurosciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Saeed Vaheb
- Isfahan Neurosciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Elham Moases Ghaffary
- Isfahan Neurosciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Vahid Shaygannejad
- Isfahan Neurosciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran; Department of Neurology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Aram Zabeti
- Department of Neurology, University of Cincinnati, Cincinnati, OH, United States
| | - Omid Mirmosayyeb
- Isfahan Neurosciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran; Department of Neurology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
43
|
Yang Z, Bao K, Yang Z, Chen S, Zheng Y. Elaborating the knowledge structure and emerging research trends of physical activity for multiple sclerosis: A bibliometric analysis from 1994 to 2023. Mult Scler Relat Disord 2024; 90:105817. [PMID: 39191096 DOI: 10.1016/j.msard.2024.105817] [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: 05/08/2024] [Revised: 07/23/2024] [Accepted: 08/12/2024] [Indexed: 08/29/2024]
Abstract
BACKGROUND Multiple sclerosis is a common inflammatory neurological disease among young adults and is the tenth leading cause of the global burden of disease. Existing common treatments such as pharmacological and palliative therapies do not control the neurodegenerative process or cure multiple sclerosis. Numerous epidemiological surveys, randomised controlled trials, and systematic reviews with meta-analyses support the effects of physical activity on health-related outcomes among patients with multiple sclerosis. Moreover, bibliometric analysis can provide a broad evidence synthesis beyond systematic reviews and meta-analyses, allowing researchers and other stakeholders to obtain a one-stop overview of this research field. Therefore, this bibliometric analysis aims to provide insight into the knowledge structure of the field of physical activity for multiple sclerosis over the past three decades, and to predict emerging research trends. METHODS This study strictly complied with step-by-step guidelines of bibliometric analysis, combining performance analysis and science mapping. Four indexes from the Web of Science Core Collection were selected as data sources, and articles and review articles in the field of physical activity for multiple sclerosis from 1994 to 2023 were included in this analysis. Mircrosoft Excel, RStudio, VOSviewer 1.6.20, and CiteSpace 6.3.R1 (64-bit) Advanced were used to perform performance analysis and science mapping. RESULTS Over the past three decades, this field published a total of 1,271 documents, with the scientific output showing a rapid upward trend over the past two decades. Robbert W Motl was the most prolific author in this field, with a total of 300 publications. The USA contributed nearly half of the publications in this field (549 documents), and the University of Illinois System was the institution with the highest number of publications (222 documents). Multiple Sclerosis and Related Disorders was the journal that published the highest number of documents in this field (117 documents), while more than a third of this field's publications were included in the category: Clinical Neurology (438 documents). The Reference co-citation analysis identified three main research trends, including shifts in research methodology, changes in health outcomes in randomised controlled trials, and shifts in different types of physical activity interventions. Combining the results from reference co-citation analysis and citation burst analysis, the combination of behaviour change technique and telerehabilitation may be the emerging research trend. CONCLUSION This bibliometric analysis identifies rapid growth in the field of physical activity for multiple sclerosis over the past two decades. Moreover, the combination of performance analysis and science mapping provides insight into knowledge structure in this field and informed future research trends for researchers and the relevant stakeholders.
Collapse
Affiliation(s)
- Zhen Yang
- Physical Activity, Sports & Health Research Group, Department of Movement Sciences, KU Leuven, Leuven, Belgium
| | - Kaiming Bao
- Division of Biokinesiology and Physical Therapy, University of Southern California, Los Angeles, USA
| | - Ziyan Yang
- The University of Sydney Business School, University of Sydney, Sydney, Australia
| | - Sitong Chen
- Centre for Mental Health, Shenzhen University, Shenzhen, China
| | - Yong Zheng
- College of Physical Education, China West Normal University, Nanchong, Sichuan, China.
| |
Collapse
|
44
|
Wurdack K, Prüss H, Finke C. Evolution from viral encephalitis to autoimmune encephalitis to multiple sclerosis: a case report. J Neurol 2024; 271:7035-7038. [PMID: 39217218 PMCID: PMC11446979 DOI: 10.1007/s00415-024-12659-9] [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: 06/03/2024] [Revised: 08/18/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND There are established associations between viral and autoimmune encephalitis as well as between autoimmune encephalitis and demyelinating central nervous system (CNS) diseases. Here, we report the evolution from varicella zoster virus (VZV) encephalitis to limbic autoimmune encephalitis (AIE) to multiple sclerosis (MS) in one patient. CASE REPORT A woman in her mid-thirties presented with headache, aphasia, and a generalized tonic-clonic seizure. Cerebrospinal fluid (CSF) VZV polymerase chain reaction was positive and treatment with acyclovir was administered for VZV encephalitis. Five months later, the patient presented with cognitive deficits and MRI showed new bilateral hippocampal T2-hyperintensities. CSF analyses revealed pleocytosis and neuropil antibodies in tissue-staining. A diagnosis of limbic AIE was established and treatment with IV steroids and IV immunoglobulins initiated. One year later, the patient developed paresthesia of both legs and magnetic resonance imaging studies now showed new supratentorial and spinal demyelinating lesions. The patient was diagnosed with MS and treatment was changed to rituximab. CONCLUSIONS This unique case report links three important neuroimmunological entities in characterizing the evolution from infectious to autoimmune encephalitis to multiple sclerosis in one patient. Identification of such rare clinical constellations is critical for correct treatment choice and provides important novel insights into the pathophysiology of neuroimmunological disorders including viral triggers and overlap manifestations of autoimmune CNS diseases.
Collapse
Affiliation(s)
- Katharina Wurdack
- Department of Neurology, Charité Universitätsmedizin Berlin, Berlin, Germany.
| | - Harald Prüss
- Department of Neurology, Charité Universitätsmedizin Berlin, Berlin, Germany
- German Center for Neurodegenerative Disorders Berlin, Berlin, Germany
| | - Carsten Finke
- Department of Neurology, Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin School of Mind and Brain, Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
45
|
Samara A, Xiang B, Judge B, Ciotti JR, Yablonskiy DA, Cross AH, Brier MR. Increased periventricular thalamic damage gradient in multiple sclerosis detected by quantitative gradient echo MRI. Mult Scler Relat Disord 2024; 90:105834. [PMID: 39208571 DOI: 10.1016/j.msard.2024.105834] [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: 02/26/2024] [Revised: 08/08/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
OBJECTIVE Thalamic tissue damage in multiple sclerosis (MS) follows a 'surface-in' gradient from the ventricular surface. The clinical consequences of this gradient are not completely understood. Using quantitative gradient-recalled echo (qGRE) MRI, we evaluated a periventricular thalamic gradient of tissue integrity in MS and its relationship with clinical variables. METHODS Structural and qGRE MRI scans were acquired for a cohort of MS patients and healthy controls (HC). qGRE-derived R2t* values were used as a measure of tissue integrity. Thalamic segmentations were divided into 1-mm concentric bands radiating from the ventricular surface, excluding the CSF-adjacent band. Median R2t* values within these bands were used to calculate the periventricular thalamic gradient. RESULTS We included 44 MS patients and 17 HC. R2t* increased slightly with distance from the ventricular surface in HC. MS patients had a steeper periventricular thalamic gradient compared to HC (mean slope 0.55 vs. 0.36; p < 0.001), which correlated with longer disease duration (β = 0.001 /year; p = 0.027) and higher Expanded Disability Status Scale (EDSS) score (β = 0.07 /EDSS point; p = 0.019). Left and right thalamus were symmetrically affected. CONCLUSIONS We detected an increased thalamic gradient in MS in vivo using qGRE MRI, which correlated with disease duration and greater clinical disability. These findings further support the 'surface-in' pathology hypothesis in MS and suggest a CSF-mediated process given symmetric bi-thalamic involvement.
Collapse
Affiliation(s)
- Amjad Samara
- Department of Neurology, Washington University School of Medicine, St. Louis, St. Louis, MO 63110, USA
| | - Biao Xiang
- Department of Neurology, Washington University School of Medicine, St. Louis, St. Louis, MO 63110, USA
| | - Bradley Judge
- Department of Radiology, Washington University School of Medicine, St. Louis, St. Louis, MO 63110, USA
| | - John R Ciotti
- Department of Neurology, University of South Florida, Tampa, FL, USA
| | - Dmitriy A Yablonskiy
- Department of Radiology, Washington University School of Medicine, St. Louis, St. Louis, MO 63110, USA
| | - Anne H Cross
- Department of Neurology, Washington University School of Medicine, St. Louis, St. Louis, MO 63110, USA
| | - Matthew R Brier
- Department of Neurology, Washington University School of Medicine, St. Louis, St. Louis, MO 63110, USA; Department of Radiology, Washington University School of Medicine, St. Louis, St. Louis, MO 63110, USA.
| |
Collapse
|
46
|
Mahmoudi F, Mirmosayyeb O, Shaabani E, Ghaffary EM, Nelson F. COVID-19 vaccination in patients with multiple sclerosis: what you need to know - a review. Health Sci Rep 2024; 7:e70119. [PMID: 39377025 PMCID: PMC11456691 DOI: 10.1002/hsr2.70119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 09/06/2024] [Accepted: 09/18/2024] [Indexed: 10/09/2024] Open
Abstract
Background The appearance of severe acute respiratory syndrome Coronavirus 2 (SARS-CoV-2) initiated the COVID-19 pandemic, resulting in millions of confirmed cases and numerous fatalities. In response, rapid vaccine development efforts were launched to mitigate the pandemic's impact. Despite the high efficacy of COVID-19 vaccines, they are also associated with several common side effects/complications, some of them specific to the multiple sclerosis population. Our goal is to review various types of COVID-19 vaccines, assessing their efficacy, adverse events, their association with an MS relapse following vaccination, and the influence of disease modifying therapies (DMTs) on vaccines' efficacy. Methods The review was based on a database search that included PubMed/Medline, Embase, Scopus, and the Web of Science conducted from January 2020 to July 2024 using the following MeSH terms: MS, COVID-19, COVID-19 vaccination, vaccine side effects, and vaccine hesitancy. Results Receiving any type of COVID-19 vaccine is a safer and more reliable approach to building immunity compared to becoming infected with the virus. Complications tend to be mild to moderate, occasionally severe. DMTs could affect the humoral response to the COVID-19 vaccine. Among all DMTs, a notable reduction in the humoral response has been observed in patients who received anti-CD20 and sphingosine-1-phosphate (S1P) receptor modulator drugs after their COVID-19 vaccination. Conclusion Despite certain drawbacks, the benefits of the COVID-19 vaccine significantly outweigh the associated risks, making it a recommended course of action for people with multiple sclerosis (pwMS). However, physicians need to be mindful of potential complications especially in patients undergoing anti CD20 and manage them appropriately.
Collapse
Affiliation(s)
| | - Omid Mirmosayyeb
- Department of Neurology, Jacobs School of Medicine and Biomedical SciencesUniversity at Buffalo, State University of New YorkBuffaloNew YorkUSA
| | - Elnaz Shaabani
- Koch Institute for Integrative Cancer Research at MITMassachusettsUSA
| | | | - Flavia Nelson
- Department of NeurologyUniversity of MiamiMiamiFloridaUSA
| |
Collapse
|
47
|
Cordeiro B, Ahn JJ, Gawde S, Ucciferri C, Alvarez-Sanchez N, Revelo XS, Stickle N, Massey K, Brooks DG, Guthridge JM, Pardo G, Winer DA, Axtell RC, Dunn SE. Obesity intensifies sex-specific interferon signaling to selectively worsen central nervous system autoimmunity in females. Cell Metab 2024; 36:2298-2314.e11. [PMID: 39168127 PMCID: PMC11463735 DOI: 10.1016/j.cmet.2024.07.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 04/29/2024] [Accepted: 07/23/2024] [Indexed: 08/23/2024]
Abstract
Obesity has been implicated in the rise of autoimmunity in women. We report that obesity induces a serum protein signature that is associated with T helper 1 (Th1), interleukin (IL)-17, and multiple sclerosis (MS) signaling pathways selectively in human females. Females, but not male mice, subjected to diet-induced overweightness/obesity (DIO) exhibited upregulated Th1/IL-17 inflammation in the central nervous system during experimental autoimmune encephalomyelitis, a model of MS. This was associated with worsened disability and a heightened expansion of myelin-specific Th1 cells in the peripheral lymphoid organs. Moreover, at steady state, DIO increased serum levels of interferon (IFN)-α and potentiated STAT1 expression and IFN-γ production by naive CD4+ T cells uniquely in female mice. This T cell phenotype was driven by increased adiposity and was prevented by the removal of ovaries or knockdown of the type I IFN receptor in T cells. Our findings offer a mechanistic explanation of how obesity enhances autoimmunity.
Collapse
Affiliation(s)
- Brendan Cordeiro
- Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, ON M5B 1W8, Canada
| | | | - Saurabh Gawde
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; Department of Microbiology and Immunology, Oklahoma University Health Science Center, Oklahoma City, OK 73104, USA
| | - Carmen Ucciferri
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Nuria Alvarez-Sanchez
- Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, ON M5B 1W8, Canada
| | - Xavier S Revelo
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Natalie Stickle
- Bioinformatics and High Performance Computing Core, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Kaylea Massey
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - David G Brooks
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C4, Canada
| | - Joel M Guthridge
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Gabriel Pardo
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; Multiple Sclerosis Center of Excellence, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Daniel A Winer
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada; Buck Institute for Research on Aging, Novato, CA 94945, USA; Division of Cellular and Molecular Biology, Diabetes Research Group, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, ON M5S 1A8, Canada
| | - Robert C Axtell
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; Department of Microbiology and Immunology, Oklahoma University Health Science Center, Oklahoma City, OK 73104, USA.
| | - Shannon E Dunn
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada; Women's College Research Institute, Women's College Hospital, Toronto, ON M5G 1N8, Canada; Sunnybrook Research Institute, Sunnybrook Hospital, Toronto, ON M4M 3M5, Canada.
| |
Collapse
|
48
|
Rodríguez-Fuentes G, Ferreiro-Gómez E, Campo-Prieto P, Cancela-Carral JM. Exergames and Immersive Virtual Reality as a Novel Therapy Approach in Multiple Sclerosis: Randomised Feasibility Study. J Clin Med 2024; 13:5845. [PMID: 39407905 PMCID: PMC11478282 DOI: 10.3390/jcm13195845] [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: 07/31/2024] [Revised: 09/17/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
Background: Multiple sclerosis is a chronic, inflammatory, neurodegenerative autoimmune disease caused by myelin loss in the central nervous system, which leads to motor and non-motor problems. The main objective of this study was to explore whether an immersive virtual reality (IVR) exercise programme would be feasible as a form of physical therapy for people with MS (pwMS). Methods: 18 participants (13 women; 45.06 years) were assigned to an experimental group (EG, n = 8) and a control group (CG, n = 10). The EG took part in a twice-weekly IVR exergame physical therapy programme-ExeRVIEM programme. A randomised, single-blind clinical trial was conducted and was registered in clinicaltrials (NCT05870254). Results: The intervention was feasible and safe. The participants completed the programme with no adverse effects (no symptoms on the Simulator Sickness Questionnaire), high usability (System Usability Scale 90.31%), and outstandingly positive post-game experiences (Game Experience Questionnaire 3.10/4). In addition, the GE significantly improved several of their functional capacities: increased lower limb strength (Five Times Sit-to-Stand Test p = 0.042), improved functional mobility, and reduced fall risk (Timed Up and Go Test-simple p = 0.009; Timed Up and Go Test-cognitive p = 0.003). There were no statistically significant differences between the groups. Conclusions: The findings support that the use of exergames and IVR as physical therapy in pwMS is feasible and safe. Furthermore, there is the suggestion of possible benefits to participants' functional abilities, all of which position IVR as a promising tool for the rehabilitation of this neurodegenerative pathology affecting young adults.
Collapse
Affiliation(s)
- Gustavo Rodríguez-Fuentes
- Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Fisioterapia, Universidade de Vigo, E-36005 Pontevedra, Spain;
- HealthyFit Research Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, E-36312 Pontevedra, Spain;
| | | | - Pablo Campo-Prieto
- Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Fisioterapia, Universidade de Vigo, E-36005 Pontevedra, Spain;
- HealthyFit Research Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, E-36312 Pontevedra, Spain;
| | - José Mª Cancela-Carral
- HealthyFit Research Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, E-36312 Pontevedra, Spain;
- Departamento de Didácticas Especiais, Facultade de Ciencias da Educación e do Deporte, Universidade de Vigo, E-36005 Pontevedra, Spain
| |
Collapse
|
49
|
Kavaka V, Mutschler L, de la Rosa Del Val C, Eglseer K, Gómez Martínez AM, Flierl-Hecht A, Ertl-Wagner B, Keeser D, Mortazavi M, Seelos K, Zimmermann H, Haas J, Wildemann B, Kümpfel T, Dornmair K, Korn T, Hohlfeld R, Kerschensteiner M, Gerdes LA, Beltrán E. Twin study identifies early immunological and metabolic dysregulation of CD8 + T cells in multiple sclerosis. Sci Immunol 2024; 9:eadj8094. [PMID: 39331727 DOI: 10.1126/sciimmunol.adj8094] [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: 07/17/2023] [Accepted: 09/05/2024] [Indexed: 09/29/2024]
Abstract
Multiple sclerosis (MS) is an inflammatory neurological disease of the central nervous system with a subclinical phase preceding frank neuroinflammation. CD8+ T cells are abundant within MS lesions, but their potential role in disease pathology remains unclear. Using high-throughput single-cell RNA sequencing and single-cell T cell receptor analysis, we compared CD8+ T cell clones from the blood and cerebrospinal fluid (CSF) of monozygotic twin pairs in which the cotwin had either no or subclinical neuroinflammation (SCNI). We identified peripheral MS-associated immunological and metabolic alterations indicative of an enhanced migratory, proinflammatory, and activated CD8+ T cell phenotype, which was also evident in cotwins with SCNI and in an independent validation cohort of people with MS. Together, our in-depth single-cell analysis indicates a disease-driving proinflammatory role of infiltrating CD8+ T cells and identifies potential immunological and metabolic therapeutic targets in both prodromal and definitive stages of the disease.
Collapse
Affiliation(s)
- Vladyslav Kavaka
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, Ludwig Maximilian University of Munich, Martinsried, Germany
| | - Luisa Mutschler
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, Ludwig Maximilian University of Munich, Martinsried, Germany
| | - Clara de la Rosa Del Val
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, Ludwig Maximilian University of Munich, Martinsried, Germany
- Graduate School of Systemic Neurosciences, Ludwig Maximilian University of Munich, Munich, Germany
| | - Klara Eglseer
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, Ludwig Maximilian University of Munich, Martinsried, Germany
| | - Ana M Gómez Martínez
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, Ludwig Maximilian University of Munich, Martinsried, Germany
| | - Andrea Flierl-Hecht
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, Ludwig Maximilian University of Munich, Martinsried, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Birgit Ertl-Wagner
- Department of Radiology, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- Department of Medical Imaging, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Daniel Keeser
- Department of Psychiatry and Psychotherapy, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Martin Mortazavi
- Department of Radiology, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- Department of Psychiatry and Psychotherapy, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Klaus Seelos
- Institute of Neuroradiology, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Hanna Zimmermann
- Institute of Neuroradiology, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Jürgen Haas
- Molecular Neuroimmunology Group, Department of Neurology, University of Heidelberg, Heidelberg, Germany
| | - Brigitte Wildemann
- Molecular Neuroimmunology Group, Department of Neurology, University of Heidelberg, Heidelberg, Germany
| | - Tania Kümpfel
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, Ludwig Maximilian University of Munich, Martinsried, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Klaus Dornmair
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, Ludwig Maximilian University of Munich, Martinsried, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Thomas Korn
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
- Institute for Experimental Neuroimmunology, Technical University of Munich School of Medicine, Munich, Germany
- Department of Neurology, Technical University of Munich School of Medicine, Munich, Germany
| | - Reinhard Hohlfeld
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, Ludwig Maximilian University of Munich, Martinsried, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Martin Kerschensteiner
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, Ludwig Maximilian University of Munich, Martinsried, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Lisa Ann Gerdes
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, Ludwig Maximilian University of Munich, Martinsried, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Eduardo Beltrán
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, Ludwig Maximilian University of Munich, Martinsried, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
50
|
Wuerch EC, Mirzaei R, Yong VW. Niacin produces an inconsistent treatment response in the EAE model of multiple sclerosis. J Neuroimmunol 2024; 394:578421. [PMID: 39088907 DOI: 10.1016/j.jneuroim.2024.578421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/21/2024] [Accepted: 07/25/2024] [Indexed: 08/03/2024]
Abstract
Niacin was found in the lysolecithin model of multiple sclerosis (MS) to promote the phagocytic clearance of debris and enhance remyelination. Lysolecithin lesions have prominent microglia/macrophages but lack lymphocytes that populate plaques of MS or its experimental autoimmune encephalomyelitis (EAE) model. Thus, the current study assessed the efficacy of niacin in EAE. We found that niacin inconsistently affects EAE clinical score, and largely does not ameliorate neuropathology. In culture, niacin enhances phagocytosis by macrophages, but does not reduce T cell proliferation. We suggest that studies of niacin for potential remyelination in MS should include a therapeutic that targets adaptive immunity.
Collapse
Affiliation(s)
- Emily C Wuerch
- Hotchkiss Brain Institute and the Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | - Reza Mirzaei
- Hotchkiss Brain Institute and the Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | - V Wee Yong
- Hotchkiss Brain Institute and the Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|