1
|
Bjørklund G, Wallace DR, Hangan T, Butnariu M, Gurgas L, Peana M. Cerebral iron accumulation in multiple sclerosis: Pathophysiology and therapeutic implications. Autoimmun Rev 2025; 24:103741. [PMID: 39756528 DOI: 10.1016/j.autrev.2025.103741] [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: 12/14/2024] [Revised: 01/02/2025] [Accepted: 01/02/2025] [Indexed: 01/07/2025]
Abstract
Multiple sclerosis (MS) is a chronic autoimmune disorder of the central nervous system characterized by demyelination, neuroinflammation, and neurodegeneration. Recent studies highlight the role of cerebral iron (Fe) accumulation in exacerbating MS pathophysiology. Fe, essential for neural function, contributes to oxidative stress and inflammation when dysregulated, particularly in the brain's gray matter and demyelinated lesions. Advanced imaging techniques, including susceptibility-weighted and quantitative susceptibility mapping, have revealed abnormal Fe deposition patterns in MS patients, suggesting its involvement in disease progression. Iron's interaction with immune cells, such as microglia, releases pro-inflammatory cytokines, further amplifying neuroinflammation and neuronal damage. These findings implicate Fe dysregulation as a significant factor in MS progression, contributing to clinical manifestations like cognitive impairment. Therapeutic strategies targeting Fe metabolism, including Fe chelation therapies, show promise in reducing Fe-related damage, instilling optimism about the future of MS treatment. However, challenges such as crossing the blood-brain barrier and maintaining Fe homeostasis remain. Emerging approaches, such as Fe-targeted nanotherapeutics and biologics, offer new possibilities for personalized treatments. However, the journey is far from over. Continued research into the molecular mechanisms of Fe-induced neuroinflammation and oxidative damage is essential. Through this research, we can develop effective interventions that could slow MS progression and improve patient outcomes.
Collapse
Affiliation(s)
- Geir Bjørklund
- Council for Nutritional and Environmental Medicine (CONEM), Mo i Rana, Norway.
| | - David R Wallace
- Department of Pharmacology, Oklahoma State University Center for Health Sciences, Tulsa, OK, United States
| | - Tony Hangan
- Faculty of Medicine, Ovidius University of Constanta, Constanta, Romania
| | - Monica Butnariu
- University of Life Sciences "King Mihai I" from Timisoara, Timis, Romania; CONEM Romania Biotechnology and Environmental Sciences Group, University of Life Sciences "King Mihai I" from Timisoara, Timis, Romania
| | - Leonard Gurgas
- Faculty of Medicine, Ovidius University of Constanta, Constanta, Romania
| | - Massimiliano Peana
- Department of Chemical, Physical, Mathematical and Natural Sciences, University of Sassari, Italy
| |
Collapse
|
2
|
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
|
3
|
Ziccardi S, Tamanti A, Ruggieri C, Guandalini M, Marastoni D, Camera V, Montibeller L, Mazziotti V, Rossi S, Calderone M, Pizzini FB, Montemezzi S, Magliozzi R, Calabrese M. CSF Parvalbumin Levels at Multiple Sclerosis Diagnosis Predict Future Worse Cognition, Physical Disability, Fatigue, and Gray Matter Damage. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2024; 11:e200301. [PMID: 39178066 PMCID: PMC11368234 DOI: 10.1212/nxi.0000000000200301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 07/11/2024] [Indexed: 08/25/2024]
Abstract
BACKGROUND AND OBJECTIVES Cognitive impairment (CI) in multiple sclerosis (MS) is frequent and determined by a complex interplay between inflammatory and neurodegenerative processes. We aimed to investigate whether CSF parvalbumin (PVALB), measured at the time of diagnosis, may have a prognostic role in patients with MS. METHODS In this cohort study, CSF analysis of PVALB and Nf-L levels was performed on all patients at diagnosis (T0) and combined with physical, cognitive, and MRI assessment after an average of 4 years of follow-up (T4) from diagnosis. Cognitive performance was evaluated with a comprehensive neuropsychologic battery: both global (cognitively normal, CN, mildly CI, mCI, and severely CI, sCI) and domain cognitive status (normal/impaired in memory, attention/information processing speed, and executive functions) were considered. Cortical thickness and gray matter volume data were acquired using 3T MRI scanner. RESULTS A total of 72 patients with MS were included. At diagnosis, PVALB levels were higher in those patients who showed a worsening physical disability after 4 years of follow-up (p = 0.011). CSF PVALB levels were higher in sCI patients than in CN (p = 0.033). Moreover, higher PVALB levels significantly correlated with worse global cognitive (p = 0.024) and memory functioning (p = 0.044). A preliminary clinical threshold for PVALB levels at diagnosis was proposed (2.57 ng/mL), which maximizes the risk of showing CI (in particular, sCI) at follow-up, with a sensitivity of 91% (specificity 30%). No significant results were found for these associations with Nf-L. In addition, patients with higher levels of PVALB at diagnosis showed higher cognitive (p = 0.024) and global fatigue (p = 0.043) at follow-up. Finally, higher PVALB levels also correlated significantly with more pronounced CTh/volume at T4 in the inferior frontal gyrus (p = 0.044), postcentral gyrus (p = 0.025), frontal pole (p = 0.042), transverse temporal gyrus (p = 0.008), and cerebellar cortex (p = 0.041) and higher atrophy (change T0-T4) in the right thalamus (p = 0.038), pericalcarine cortex (p = 0.009), lingual gyrus (p = 0.045), and medial frontal gyrus (p = 0.028). DISCUSSION The significant association found between parvalbumin levels in the CSF at diagnosis and cognitive, clinical, and neuroradiologic worsening after 4 years of follow-up support the idea that parvalbumin, in addition to Nf-L, might represent a new potential prognostic biomarker, reflecting MS neurodegenerative processes occurring since early disease stages.
Collapse
Affiliation(s)
- Stefano Ziccardi
- From the Department of Neurosciences (S.Z., A.T., C.R., M.G., D.M., V.C., L.M., V.M., S.R., R.M., M. Calabrese), Biomedicine and Movement, University of Verona; Department of Oncology and Molecular Medicine (S.R.), Istituto Superiore di Sanità, Rome; Radiology Unit (M. Calderone), Cmsr Veneto Medica s.r.l., Altavilla Vicentina, Vicenza; and Institute of Radiology (F.B.P., S.M.), University of Verona, Italy
| | - Agnese Tamanti
- From the Department of Neurosciences (S.Z., A.T., C.R., M.G., D.M., V.C., L.M., V.M., S.R., R.M., M. Calabrese), Biomedicine and Movement, University of Verona; Department of Oncology and Molecular Medicine (S.R.), Istituto Superiore di Sanità, Rome; Radiology Unit (M. Calderone), Cmsr Veneto Medica s.r.l., Altavilla Vicentina, Vicenza; and Institute of Radiology (F.B.P., S.M.), University of Verona, Italy
| | - Claudia Ruggieri
- From the Department of Neurosciences (S.Z., A.T., C.R., M.G., D.M., V.C., L.M., V.M., S.R., R.M., M. Calabrese), Biomedicine and Movement, University of Verona; Department of Oncology and Molecular Medicine (S.R.), Istituto Superiore di Sanità, Rome; Radiology Unit (M. Calderone), Cmsr Veneto Medica s.r.l., Altavilla Vicentina, Vicenza; and Institute of Radiology (F.B.P., S.M.), University of Verona, Italy
| | - Maddalena Guandalini
- From the Department of Neurosciences (S.Z., A.T., C.R., M.G., D.M., V.C., L.M., V.M., S.R., R.M., M. Calabrese), Biomedicine and Movement, University of Verona; Department of Oncology and Molecular Medicine (S.R.), Istituto Superiore di Sanità, Rome; Radiology Unit (M. Calderone), Cmsr Veneto Medica s.r.l., Altavilla Vicentina, Vicenza; and Institute of Radiology (F.B.P., S.M.), University of Verona, Italy
| | - Damiano Marastoni
- From the Department of Neurosciences (S.Z., A.T., C.R., M.G., D.M., V.C., L.M., V.M., S.R., R.M., M. Calabrese), Biomedicine and Movement, University of Verona; Department of Oncology and Molecular Medicine (S.R.), Istituto Superiore di Sanità, Rome; Radiology Unit (M. Calderone), Cmsr Veneto Medica s.r.l., Altavilla Vicentina, Vicenza; and Institute of Radiology (F.B.P., S.M.), University of Verona, Italy
| | - Valentina Camera
- From the Department of Neurosciences (S.Z., A.T., C.R., M.G., D.M., V.C., L.M., V.M., S.R., R.M., M. Calabrese), Biomedicine and Movement, University of Verona; Department of Oncology and Molecular Medicine (S.R.), Istituto Superiore di Sanità, Rome; Radiology Unit (M. Calderone), Cmsr Veneto Medica s.r.l., Altavilla Vicentina, Vicenza; and Institute of Radiology (F.B.P., S.M.), University of Verona, Italy
| | - Luigi Montibeller
- From the Department of Neurosciences (S.Z., A.T., C.R., M.G., D.M., V.C., L.M., V.M., S.R., R.M., M. Calabrese), Biomedicine and Movement, University of Verona; Department of Oncology and Molecular Medicine (S.R.), Istituto Superiore di Sanità, Rome; Radiology Unit (M. Calderone), Cmsr Veneto Medica s.r.l., Altavilla Vicentina, Vicenza; and Institute of Radiology (F.B.P., S.M.), University of Verona, Italy
| | - Valentina Mazziotti
- From the Department of Neurosciences (S.Z., A.T., C.R., M.G., D.M., V.C., L.M., V.M., S.R., R.M., M. Calabrese), Biomedicine and Movement, University of Verona; Department of Oncology and Molecular Medicine (S.R.), Istituto Superiore di Sanità, Rome; Radiology Unit (M. Calderone), Cmsr Veneto Medica s.r.l., Altavilla Vicentina, Vicenza; and Institute of Radiology (F.B.P., S.M.), University of Verona, Italy
| | - Stefania Rossi
- From the Department of Neurosciences (S.Z., A.T., C.R., M.G., D.M., V.C., L.M., V.M., S.R., R.M., M. Calabrese), Biomedicine and Movement, University of Verona; Department of Oncology and Molecular Medicine (S.R.), Istituto Superiore di Sanità, Rome; Radiology Unit (M. Calderone), Cmsr Veneto Medica s.r.l., Altavilla Vicentina, Vicenza; and Institute of Radiology (F.B.P., S.M.), University of Verona, Italy
| | - Milena Calderone
- From the Department of Neurosciences (S.Z., A.T., C.R., M.G., D.M., V.C., L.M., V.M., S.R., R.M., M. Calabrese), Biomedicine and Movement, University of Verona; Department of Oncology and Molecular Medicine (S.R.), Istituto Superiore di Sanità, Rome; Radiology Unit (M. Calderone), Cmsr Veneto Medica s.r.l., Altavilla Vicentina, Vicenza; and Institute of Radiology (F.B.P., S.M.), University of Verona, Italy
| | - Francesca Benedetta Pizzini
- From the Department of Neurosciences (S.Z., A.T., C.R., M.G., D.M., V.C., L.M., V.M., S.R., R.M., M. Calabrese), Biomedicine and Movement, University of Verona; Department of Oncology and Molecular Medicine (S.R.), Istituto Superiore di Sanità, Rome; Radiology Unit (M. Calderone), Cmsr Veneto Medica s.r.l., Altavilla Vicentina, Vicenza; and Institute of Radiology (F.B.P., S.M.), University of Verona, Italy
| | - Stefania Montemezzi
- From the Department of Neurosciences (S.Z., A.T., C.R., M.G., D.M., V.C., L.M., V.M., S.R., R.M., M. Calabrese), Biomedicine and Movement, University of Verona; Department of Oncology and Molecular Medicine (S.R.), Istituto Superiore di Sanità, Rome; Radiology Unit (M. Calderone), Cmsr Veneto Medica s.r.l., Altavilla Vicentina, Vicenza; and Institute of Radiology (F.B.P., S.M.), University of Verona, Italy
| | - Roberta Magliozzi
- From the Department of Neurosciences (S.Z., A.T., C.R., M.G., D.M., V.C., L.M., V.M., S.R., R.M., M. Calabrese), Biomedicine and Movement, University of Verona; Department of Oncology and Molecular Medicine (S.R.), Istituto Superiore di Sanità, Rome; Radiology Unit (M. Calderone), Cmsr Veneto Medica s.r.l., Altavilla Vicentina, Vicenza; and Institute of Radiology (F.B.P., S.M.), University of Verona, Italy
| | - Massimiliano Calabrese
- From the Department of Neurosciences (S.Z., A.T., C.R., M.G., D.M., V.C., L.M., V.M., S.R., R.M., M. Calabrese), Biomedicine and Movement, University of Verona; Department of Oncology and Molecular Medicine (S.R.), Istituto Superiore di Sanità, Rome; Radiology Unit (M. Calderone), Cmsr Veneto Medica s.r.l., Altavilla Vicentina, Vicenza; and Institute of Radiology (F.B.P., S.M.), University of Verona, Italy
| |
Collapse
|
4
|
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
|
5
|
Marastoni D, Turano E, Tamanti A, Colato E, Pisani AI, Scartezzini A, Carotenuto S, Mazziotti V, Camera V, Anni D, Ziccardi S, Guandalini M, Pizzini FB, Virla F, Mariotti R, Magliozzi R, Bonetti B, Steinman L, Calabrese M. Association of Levels of CSF Osteopontin With Cortical Atrophy and Disability in Early Multiple Sclerosis. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2024; 11:e200265. [PMID: 38917380 PMCID: PMC11203401 DOI: 10.1212/nxi.0000000000200265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 04/29/2024] [Indexed: 06/27/2024]
Abstract
BACKGROUND AND OBJECTIVES To evaluate CSF inflammatory markers with accumulation of cortical damage as well as disease activity in patients with early relapsing-remitting MS (RRMS). METHODS CSF levels of osteopontin (OPN) and 66 inflammatory markers were assessed using an immune-assay multiplex technique in 107 patients with RRMS (82 F/25 M, mean age 35.7 ± 11.8 years). All patients underwent regular clinical assessment and yearly 3T MRI scans for 2 years while 39 patients had a 4-year follow-up. White matter lesion number and volume, cortical lesions (CLs) and volume, and global cortical thickness (CTh) were evaluated together with the 'no evidence of disease activity' (NEDA-3) status, defined by no relapses, no disability worsening, and no MRI activity, including CLs. RESULTS The random forest algorithm selected OPN, CXCL13, TWEAK, TNF, IL19, sCD30, sTNFR1, IL35, IL16, and sCD163 as significantly associated with changes in global CTh. OPN and CXCL13 were most related to accumulation of atrophy after 2 and 4 years. In a multivariate linear regression model on CSF markers, OPN (p < 0.001), CXCL13 (p = 0.001), and sTNFR1 (p = 0.024) were increased in those patients with accumulating atrophy (adjusted R-squared 0.615). The 10 markers were added in a model that included all clinical, demographic, and MRI variables: OPN (p = 0.002) and IL19 (p = 0.022) levels were confirmed to be significantly increased in patients developing more CTh change over the follow-up (adjusted R-squared 0.619). CXCL13 and OPN also revealed the best association with NEDA-3 after 2 years, with OPN significantly linked to disability accumulation (OR 2.468 [1.46-5.034], p = 0.004) at the multivariate logistic regression model. DISCUSSION These data confirm and expand our knowledge on the prognostic role of the CSF inflammatory profile in predicting changes in cortical pathology and disease activity in early MS. The data emphasize a crucial role of OPN.
Collapse
Affiliation(s)
- Damiano Marastoni
- From the Neurology B (D.M., E.T., A.T., E.C., A.I.P., A.S., S.C., V.M., V.C., D.A., S.Z., M.G., F.V., R. Magliozzi, M.C.); Anatomy and Histology section (E.T., F.V., R. Mariotti), Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy; Department of Anatomy and Neurosciences (E.C.), MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands; Neuroradiology and Radiology Units (F.B.P.), Department of Engineering for Innovation Medicine, University of Verona, Italy; Department of Brain Sciences (R. Magliozzi), Faculty of Medicine, Imperial College London, United Kingdom; Neurology A (B.B.), Azienda Ospedaliera Universitaria Integrata di Verona, Italy; and Department of Neurology and Neurological Sciences Stanford University (L.S.), CA
| | - Ermanna Turano
- From the Neurology B (D.M., E.T., A.T., E.C., A.I.P., A.S., S.C., V.M., V.C., D.A., S.Z., M.G., F.V., R. Magliozzi, M.C.); Anatomy and Histology section (E.T., F.V., R. Mariotti), Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy; Department of Anatomy and Neurosciences (E.C.), MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands; Neuroradiology and Radiology Units (F.B.P.), Department of Engineering for Innovation Medicine, University of Verona, Italy; Department of Brain Sciences (R. Magliozzi), Faculty of Medicine, Imperial College London, United Kingdom; Neurology A (B.B.), Azienda Ospedaliera Universitaria Integrata di Verona, Italy; and Department of Neurology and Neurological Sciences Stanford University (L.S.), CA
| | - Agnese Tamanti
- From the Neurology B (D.M., E.T., A.T., E.C., A.I.P., A.S., S.C., V.M., V.C., D.A., S.Z., M.G., F.V., R. Magliozzi, M.C.); Anatomy and Histology section (E.T., F.V., R. Mariotti), Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy; Department of Anatomy and Neurosciences (E.C.), MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands; Neuroradiology and Radiology Units (F.B.P.), Department of Engineering for Innovation Medicine, University of Verona, Italy; Department of Brain Sciences (R. Magliozzi), Faculty of Medicine, Imperial College London, United Kingdom; Neurology A (B.B.), Azienda Ospedaliera Universitaria Integrata di Verona, Italy; and Department of Neurology and Neurological Sciences Stanford University (L.S.), CA
| | - Elisa Colato
- From the Neurology B (D.M., E.T., A.T., E.C., A.I.P., A.S., S.C., V.M., V.C., D.A., S.Z., M.G., F.V., R. Magliozzi, M.C.); Anatomy and Histology section (E.T., F.V., R. Mariotti), Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy; Department of Anatomy and Neurosciences (E.C.), MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands; Neuroradiology and Radiology Units (F.B.P.), Department of Engineering for Innovation Medicine, University of Verona, Italy; Department of Brain Sciences (R. Magliozzi), Faculty of Medicine, Imperial College London, United Kingdom; Neurology A (B.B.), Azienda Ospedaliera Universitaria Integrata di Verona, Italy; and Department of Neurology and Neurological Sciences Stanford University (L.S.), CA
| | - Anna Isabella Pisani
- From the Neurology B (D.M., E.T., A.T., E.C., A.I.P., A.S., S.C., V.M., V.C., D.A., S.Z., M.G., F.V., R. Magliozzi, M.C.); Anatomy and Histology section (E.T., F.V., R. Mariotti), Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy; Department of Anatomy and Neurosciences (E.C.), MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands; Neuroradiology and Radiology Units (F.B.P.), Department of Engineering for Innovation Medicine, University of Verona, Italy; Department of Brain Sciences (R. Magliozzi), Faculty of Medicine, Imperial College London, United Kingdom; Neurology A (B.B.), Azienda Ospedaliera Universitaria Integrata di Verona, Italy; and Department of Neurology and Neurological Sciences Stanford University (L.S.), CA
| | - Arianna Scartezzini
- From the Neurology B (D.M., E.T., A.T., E.C., A.I.P., A.S., S.C., V.M., V.C., D.A., S.Z., M.G., F.V., R. Magliozzi, M.C.); Anatomy and Histology section (E.T., F.V., R. Mariotti), Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy; Department of Anatomy and Neurosciences (E.C.), MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands; Neuroradiology and Radiology Units (F.B.P.), Department of Engineering for Innovation Medicine, University of Verona, Italy; Department of Brain Sciences (R. Magliozzi), Faculty of Medicine, Imperial College London, United Kingdom; Neurology A (B.B.), Azienda Ospedaliera Universitaria Integrata di Verona, Italy; and Department of Neurology and Neurological Sciences Stanford University (L.S.), CA
| | - Silvia Carotenuto
- From the Neurology B (D.M., E.T., A.T., E.C., A.I.P., A.S., S.C., V.M., V.C., D.A., S.Z., M.G., F.V., R. Magliozzi, M.C.); Anatomy and Histology section (E.T., F.V., R. Mariotti), Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy; Department of Anatomy and Neurosciences (E.C.), MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands; Neuroradiology and Radiology Units (F.B.P.), Department of Engineering for Innovation Medicine, University of Verona, Italy; Department of Brain Sciences (R. Magliozzi), Faculty of Medicine, Imperial College London, United Kingdom; Neurology A (B.B.), Azienda Ospedaliera Universitaria Integrata di Verona, Italy; and Department of Neurology and Neurological Sciences Stanford University (L.S.), CA
| | - Valentina Mazziotti
- From the Neurology B (D.M., E.T., A.T., E.C., A.I.P., A.S., S.C., V.M., V.C., D.A., S.Z., M.G., F.V., R. Magliozzi, M.C.); Anatomy and Histology section (E.T., F.V., R. Mariotti), Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy; Department of Anatomy and Neurosciences (E.C.), MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands; Neuroradiology and Radiology Units (F.B.P.), Department of Engineering for Innovation Medicine, University of Verona, Italy; Department of Brain Sciences (R. Magliozzi), Faculty of Medicine, Imperial College London, United Kingdom; Neurology A (B.B.), Azienda Ospedaliera Universitaria Integrata di Verona, Italy; and Department of Neurology and Neurological Sciences Stanford University (L.S.), CA
| | - Valentina Camera
- From the Neurology B (D.M., E.T., A.T., E.C., A.I.P., A.S., S.C., V.M., V.C., D.A., S.Z., M.G., F.V., R. Magliozzi, M.C.); Anatomy and Histology section (E.T., F.V., R. Mariotti), Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy; Department of Anatomy and Neurosciences (E.C.), MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands; Neuroradiology and Radiology Units (F.B.P.), Department of Engineering for Innovation Medicine, University of Verona, Italy; Department of Brain Sciences (R. Magliozzi), Faculty of Medicine, Imperial College London, United Kingdom; Neurology A (B.B.), Azienda Ospedaliera Universitaria Integrata di Verona, Italy; and Department of Neurology and Neurological Sciences Stanford University (L.S.), CA
| | - Daniela Anni
- From the Neurology B (D.M., E.T., A.T., E.C., A.I.P., A.S., S.C., V.M., V.C., D.A., S.Z., M.G., F.V., R. Magliozzi, M.C.); Anatomy and Histology section (E.T., F.V., R. Mariotti), Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy; Department of Anatomy and Neurosciences (E.C.), MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands; Neuroradiology and Radiology Units (F.B.P.), Department of Engineering for Innovation Medicine, University of Verona, Italy; Department of Brain Sciences (R. Magliozzi), Faculty of Medicine, Imperial College London, United Kingdom; Neurology A (B.B.), Azienda Ospedaliera Universitaria Integrata di Verona, Italy; and Department of Neurology and Neurological Sciences Stanford University (L.S.), CA
| | - Stefano Ziccardi
- From the Neurology B (D.M., E.T., A.T., E.C., A.I.P., A.S., S.C., V.M., V.C., D.A., S.Z., M.G., F.V., R. Magliozzi, M.C.); Anatomy and Histology section (E.T., F.V., R. Mariotti), Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy; Department of Anatomy and Neurosciences (E.C.), MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands; Neuroradiology and Radiology Units (F.B.P.), Department of Engineering for Innovation Medicine, University of Verona, Italy; Department of Brain Sciences (R. Magliozzi), Faculty of Medicine, Imperial College London, United Kingdom; Neurology A (B.B.), Azienda Ospedaliera Universitaria Integrata di Verona, Italy; and Department of Neurology and Neurological Sciences Stanford University (L.S.), CA
| | - Maddalena Guandalini
- From the Neurology B (D.M., E.T., A.T., E.C., A.I.P., A.S., S.C., V.M., V.C., D.A., S.Z., M.G., F.V., R. Magliozzi, M.C.); Anatomy and Histology section (E.T., F.V., R. Mariotti), Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy; Department of Anatomy and Neurosciences (E.C.), MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands; Neuroradiology and Radiology Units (F.B.P.), Department of Engineering for Innovation Medicine, University of Verona, Italy; Department of Brain Sciences (R. Magliozzi), Faculty of Medicine, Imperial College London, United Kingdom; Neurology A (B.B.), Azienda Ospedaliera Universitaria Integrata di Verona, Italy; and Department of Neurology and Neurological Sciences Stanford University (L.S.), CA
| | - Francesca B Pizzini
- From the Neurology B (D.M., E.T., A.T., E.C., A.I.P., A.S., S.C., V.M., V.C., D.A., S.Z., M.G., F.V., R. Magliozzi, M.C.); Anatomy and Histology section (E.T., F.V., R. Mariotti), Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy; Department of Anatomy and Neurosciences (E.C.), MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands; Neuroradiology and Radiology Units (F.B.P.), Department of Engineering for Innovation Medicine, University of Verona, Italy; Department of Brain Sciences (R. Magliozzi), Faculty of Medicine, Imperial College London, United Kingdom; Neurology A (B.B.), Azienda Ospedaliera Universitaria Integrata di Verona, Italy; and Department of Neurology and Neurological Sciences Stanford University (L.S.), CA
| | - Federica Virla
- From the Neurology B (D.M., E.T., A.T., E.C., A.I.P., A.S., S.C., V.M., V.C., D.A., S.Z., M.G., F.V., R. Magliozzi, M.C.); Anatomy and Histology section (E.T., F.V., R. Mariotti), Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy; Department of Anatomy and Neurosciences (E.C.), MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands; Neuroradiology and Radiology Units (F.B.P.), Department of Engineering for Innovation Medicine, University of Verona, Italy; Department of Brain Sciences (R. Magliozzi), Faculty of Medicine, Imperial College London, United Kingdom; Neurology A (B.B.), Azienda Ospedaliera Universitaria Integrata di Verona, Italy; and Department of Neurology and Neurological Sciences Stanford University (L.S.), CA
| | - Raffaella Mariotti
- From the Neurology B (D.M., E.T., A.T., E.C., A.I.P., A.S., S.C., V.M., V.C., D.A., S.Z., M.G., F.V., R. Magliozzi, M.C.); Anatomy and Histology section (E.T., F.V., R. Mariotti), Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy; Department of Anatomy and Neurosciences (E.C.), MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands; Neuroradiology and Radiology Units (F.B.P.), Department of Engineering for Innovation Medicine, University of Verona, Italy; Department of Brain Sciences (R. Magliozzi), Faculty of Medicine, Imperial College London, United Kingdom; Neurology A (B.B.), Azienda Ospedaliera Universitaria Integrata di Verona, Italy; and Department of Neurology and Neurological Sciences Stanford University (L.S.), CA
| | - Roberta Magliozzi
- From the Neurology B (D.M., E.T., A.T., E.C., A.I.P., A.S., S.C., V.M., V.C., D.A., S.Z., M.G., F.V., R. Magliozzi, M.C.); Anatomy and Histology section (E.T., F.V., R. Mariotti), Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy; Department of Anatomy and Neurosciences (E.C.), MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands; Neuroradiology and Radiology Units (F.B.P.), Department of Engineering for Innovation Medicine, University of Verona, Italy; Department of Brain Sciences (R. Magliozzi), Faculty of Medicine, Imperial College London, United Kingdom; Neurology A (B.B.), Azienda Ospedaliera Universitaria Integrata di Verona, Italy; and Department of Neurology and Neurological Sciences Stanford University (L.S.), CA
| | - Bruno Bonetti
- From the Neurology B (D.M., E.T., A.T., E.C., A.I.P., A.S., S.C., V.M., V.C., D.A., S.Z., M.G., F.V., R. Magliozzi, M.C.); Anatomy and Histology section (E.T., F.V., R. Mariotti), Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy; Department of Anatomy and Neurosciences (E.C.), MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands; Neuroradiology and Radiology Units (F.B.P.), Department of Engineering for Innovation Medicine, University of Verona, Italy; Department of Brain Sciences (R. Magliozzi), Faculty of Medicine, Imperial College London, United Kingdom; Neurology A (B.B.), Azienda Ospedaliera Universitaria Integrata di Verona, Italy; and Department of Neurology and Neurological Sciences Stanford University (L.S.), CA
| | - Lawrence Steinman
- From the Neurology B (D.M., E.T., A.T., E.C., A.I.P., A.S., S.C., V.M., V.C., D.A., S.Z., M.G., F.V., R. Magliozzi, M.C.); Anatomy and Histology section (E.T., F.V., R. Mariotti), Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy; Department of Anatomy and Neurosciences (E.C.), MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands; Neuroradiology and Radiology Units (F.B.P.), Department of Engineering for Innovation Medicine, University of Verona, Italy; Department of Brain Sciences (R. Magliozzi), Faculty of Medicine, Imperial College London, United Kingdom; Neurology A (B.B.), Azienda Ospedaliera Universitaria Integrata di Verona, Italy; and Department of Neurology and Neurological Sciences Stanford University (L.S.), CA
| | - Massimiliano Calabrese
- From the Neurology B (D.M., E.T., A.T., E.C., A.I.P., A.S., S.C., V.M., V.C., D.A., S.Z., M.G., F.V., R. Magliozzi, M.C.); Anatomy and Histology section (E.T., F.V., R. Mariotti), Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy; Department of Anatomy and Neurosciences (E.C.), MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands; Neuroradiology and Radiology Units (F.B.P.), Department of Engineering for Innovation Medicine, University of Verona, Italy; Department of Brain Sciences (R. Magliozzi), Faculty of Medicine, Imperial College London, United Kingdom; Neurology A (B.B.), Azienda Ospedaliera Universitaria Integrata di Verona, Italy; and Department of Neurology and Neurological Sciences Stanford University (L.S.), CA
| |
Collapse
|
6
|
Aliyu M, Zohora FT, Ceylan A, Hossain F, Yazdani R, Azizi G. Immunopathogenesis of multiple sclerosis: molecular and cellular mechanisms and new immunotherapeutic approaches. Immunopharmacol Immunotoxicol 2024; 46:355-377. [PMID: 38634438 DOI: 10.1080/08923973.2024.2330642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 03/09/2024] [Indexed: 04/19/2024]
Abstract
BACKGROUND Multiple sclerosis (MS) is a central nervous system (CNS) demyelinating autoimmune disease with increasing global prevalence. It predominantly affects females, especially those of European descent. The interplay between environmental factors and genetic predisposition plays a crucial role in MS etiopathogenesis. METHODS We searched recent relevant literature on reputable databases, which include, PubMed, Embase, Web of Science, Scopus, and ScienceDirect using the following keywords: multiple sclerosis, pathogenesis, autoimmunity, demyelination, therapy, and immunotherapy. RESULTS Various animal models have been employed to investigate the MS etiopathogenesis and therapeutics. Autoreactive T cells within the CNS recruit myeloid cells through chemokine expression, leading to the secretion of inflammatory cytokines driving the MS pathogenesis, resulting in demyelination, gliosis, and axonal loss. Key players include T cell lymphocytes (CD4+ and CD8+), B cells, and neutrophils. Signaling dysregulation in inflammatory pathways and the immunogenetic basis of MS are essential considerations for any successful therapy to MS. Data indicates that B cells and neutrophils also have significant roles in MS, despite the common belief that T cells are essential. High neutrophil-to-lymphocyte ratios correlate with MS severity, indicating their contribution to disease progression. Dysregulated signaling pathways further exacerbate MS progression. CONCLUSION MS remains incurable, but disease-modifying therapies, monoclonal antibodies, and immunomodulatory drugs offer hope for patients. Research on the immunogenetics and immunoregulatory functions of gut microbiota is continuing to provide light on possible treatment avenues. Understanding the complex interplay between genetic predisposition, environmental factors, and immune dysregulation is critical for developing effective treatments for MS.
Collapse
Affiliation(s)
- Mansur Aliyu
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, International Campus, TUMS-IC, Tehran, Iran
- Department of Medical Microbiology, Faculty of Clinical Science, College of Health Sciences, Bayero University, Kano, Nigeria
| | - Fatema Tuz Zohora
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Subang Jaya, Malaysia
| | - Ayca Ceylan
- Medical Faculty, Department of Pediatrics, Division of Immunology and Allergy, Selcuk University, Konya, Turkey
| | - Fariha Hossain
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Subang Jaya, Malaysia
| | - Reza Yazdani
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Gholamreza Azizi
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, USA
- Non-communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| |
Collapse
|
7
|
Lazzarotto A, Hamzaoui M, Tonietto M, Dubessy AL, Khalil M, Pirpamer L, Ropele S, Enzinger C, Battaglini M, Stromillo ML, De Stefano N, Filippi M, Rocca MA, Gallo P, Gasperini C, Stankoff B, Bodini B. Time is myelin: early cortical myelin repair prevents atrophy and clinical progression in multiple sclerosis. Brain 2024; 147:1331-1343. [PMID: 38267729 PMCID: PMC10994569 DOI: 10.1093/brain/awae024] [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: 07/07/2023] [Revised: 12/15/2023] [Accepted: 01/13/2024] [Indexed: 01/26/2024] Open
Abstract
Cortical myelin loss and repair in multiple sclerosis (MS) have been explored in neuropathological studies, but the impact of these processes on neurodegeneration and the irreversible clinical progression of the disease remains unknown. Here, we evaluated in vivo cortical demyelination and remyelination in a large cohort of people with all clinical phenotypes of MS followed up for 5 years using magnetization transfer imaging (MTI), a technique that has been shown to be sensitive to myelin content changes in the cortex. We investigated 140 people with MS (37 clinically isolated syndrome, 71 relapsing-MS, 32 progressive-MS), who were clinically assessed at baseline and after 5 years and, along with 84 healthy controls, underwent a 3 T-MRI protocol including MTI at baseline and after 1 year. Changes in cortical volume over the radiological follow-up were computed with a Jacobian integration method. Magnetization transfer ratio was employed to calculate for each patient an index of cortical demyelination at baseline and of dynamic cortical demyelination and remyelination over the follow-up period. The three indices of cortical myelin content change were heterogeneous across patients but did not significantly differ across clinical phenotypes or treatment groups. Cortical remyelination, which tended to fail in the regions closer to CSF (-11%, P < 0.001), was extensive in half of the cohort and occurred independently of age, disease duration and clinical phenotype. Higher indices of cortical dynamic demyelination (β = 0.23, P = 0.024) and lower indices of cortical remyelination (β = -0.18, P = 0.03) were significantly associated with greater cortical atrophy after 1 year, independently of age and MS phenotype. While the extent of cortical demyelination predicted a higher probability of clinical progression after 5 years in the entire cohort [odds ratio (OR) = 1.2; P = 0.043], the impact of cortical remyelination in reducing the risk of accumulating clinical disability after 5 years was significant only in the subgroup of patients with shorter disease duration and limited extent of demyelination in cortical regions (OR = 0.86, P = 0.015, area under the curve = 0.93). In this subgroup, a 30% increase in cortical remyelination nearly halved the risk of clinical progression at 5 years, independently of clinical relapses. Overall, our results highlight the critical role of cortical myelin dynamics in the cascade of events leading to neurodegeneration and to the subsequent accumulation of irreversible disability in MS. Our findings suggest that early-stage myelin repair compensating for cortical myelin loss has the potential to prevent neuro-axonal loss and its long-term irreversible clinical consequences in people with MS.
Collapse
Affiliation(s)
- Andrea Lazzarotto
- Department of Neuroscience, Sorbonne Université, Paris Brain Institute, CNRS, Inserm, 75013 Paris, France
- AP-HP, Hôpital Universitaire Pitié-Salpêtrière, 75013 Paris, France
- Padova Neuroscience Center, University of Padua, 35122 Padua, Italy
| | - Mariem Hamzaoui
- Department of Neuroscience, Sorbonne Université, Paris Brain Institute, CNRS, Inserm, 75013 Paris, France
| | - Matteo Tonietto
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Service Hospitalier Frédéric Joliot, 91400 Orsay, France
- Roche Pharma Research & Early Development, F. Hoffmann-La Roche Ltd., CH-4070 Basel, Switzerland
| | | | - Michael Khalil
- Department of Neurology, Medical University of Graz, 8036 Graz, Austria
| | - Lukas Pirpamer
- Department of Neurology, Medical University of Graz, 8036 Graz, Austria
- Medical Image Analysis Center (MIAC) and Department of Biomedical Engineering, University of Basel, CH-4051 Basel, Switzerland
| | - Stefan Ropele
- Department of Neurology, Medical University of Graz, 8036 Graz, Austria
| | | | - Marco Battaglini
- Department of Medicine, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy
| | - Maria Laura Stromillo
- Department of Medicine, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy
| | - Nicola De Stefano
- Department of Medicine, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy
| | - Massimo Filippi
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, Neurology Unit, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Maria Assunta Rocca
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, Neurology Unit, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Paolo Gallo
- Padova Neuroscience Center, University of Padua, 35122 Padua, Italy
- Multiple Sclerosis Centre of Veneto Region, 35128 Padua, Italy
| | | | - Bruno Stankoff
- Department of Neuroscience, Sorbonne Université, Paris Brain Institute, CNRS, Inserm, 75013 Paris, France
- AP-HP, Hôpital Universitaire Pitié-Salpêtrière, 75013 Paris, France
| | - Benedetta Bodini
- Department of Neuroscience, Sorbonne Université, Paris Brain Institute, CNRS, Inserm, 75013 Paris, France
- AP-HP, Hôpital Universitaire Pitié-Salpêtrière, 75013 Paris, France
| |
Collapse
|
8
|
Dziedzic A, Maciak K, Miller ED, Starosta M, Saluk J. Targeting Vascular Impairment, Neuroinflammation, and Oxidative Stress Dynamics with Whole-Body Cryotherapy in Multiple Sclerosis Treatment. Int J Mol Sci 2024; 25:3858. [PMID: 38612668 PMCID: PMC11011409 DOI: 10.3390/ijms25073858] [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: 01/31/2024] [Revised: 03/05/2024] [Accepted: 03/28/2024] [Indexed: 04/14/2024] Open
Abstract
Multiple sclerosis (MS), traditionally perceived as a neurodegenerative disease, exhibits significant vascular alternations, including blood-brain barrier (BBB) disruption, which may predispose patients to increased cardiovascular risks. This vascular dysfunction is intricately linked with the infiltration of immune cells into the central nervous system (CNS), which plays a significant role in perpetuating neuroinflammation. Additionally, oxidative stress serves not only as a byproduct of inflammatory processes but also as an active contributor to neural damage. The synthesis of these multifaceted aspects highlights the importance of understanding their cumulative impact on MS progression. This review reveals that the triad of vascular damage, chronic inflammation, and oxidative imbalance may be considered interdependent processes that exacerbate each other, underscoring the need for holistic and multi-targeted therapeutic approaches in MS management. There is a necessity for reevaluating MS treatment strategies to encompass these overlapping pathologies, offering insights for future research and potential therapeutic interventions. Whole-body cryotherapy (WBCT) emerges as one of the potential avenues for holistic MS management approaches which may alleviate the triad of MS progression factors in multiple ways.
Collapse
Affiliation(s)
- Angela Dziedzic
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland; (A.D.); (K.M.)
| | - Karina Maciak
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland; (A.D.); (K.M.)
| | - Elżbieta Dorota Miller
- Department of Neurological Rehabilitation, Medical University of Lodz, Milionowa 14, 93-113 Lodz, Poland; (E.D.M.); (M.S.)
| | - Michał Starosta
- Department of Neurological Rehabilitation, Medical University of Lodz, Milionowa 14, 93-113 Lodz, Poland; (E.D.M.); (M.S.)
| | - Joanna Saluk
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland; (A.D.); (K.M.)
| |
Collapse
|
9
|
Nimmo J, Byrne R, Daskoulidou N, Watkins L, Carpanini S, Zelek W, Morgan B. The complement system in neurodegenerative diseases. Clin Sci (Lond) 2024; 138:387-412. [PMID: 38505993 PMCID: PMC10958133 DOI: 10.1042/cs20230513] [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: 10/31/2023] [Revised: 02/15/2024] [Accepted: 03/01/2024] [Indexed: 03/21/2024]
Abstract
Complement is an important component of innate immune defence against pathogens and crucial for efficient immune complex disposal. These core protective activities are dependent in large part on properly regulated complement-mediated inflammation. Dysregulated complement activation, often driven by persistence of activating triggers, is a cause of pathological inflammation in numerous diseases, including neurological diseases. Increasingly, this has become apparent not only in well-recognized neuroinflammatory diseases like multiple sclerosis but also in neurodegenerative and neuropsychiatric diseases where inflammation was previously either ignored or dismissed as a secondary event. There is now a large and rapidly growing body of evidence implicating complement in neurological diseases that cannot be comprehensively addressed in a brief review. Here, we will focus on neurodegenerative diseases, including not only the 'classical' neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease, but also two other neurological diseases where neurodegeneration is a neglected feature and complement is implicated, namely, schizophrenia, a neurodevelopmental disorder with many mechanistic features of neurodegeneration, and multiple sclerosis, a demyelinating disorder where neurodegeneration is a major cause of progressive decline. We will discuss the evidence implicating complement as a driver of pathology in these diverse diseases and address briefly the potential and pitfalls of anti-complement drug therapy for neurodegenerative diseases.
Collapse
Affiliation(s)
- Jacqui Nimmo
- UK Dementia Research Institute Cardiff, Cardiff University, Cardiff CF24 4HQ, U.K
| | - Robert A.J. Byrne
- UK Dementia Research Institute Cardiff, Cardiff University, Cardiff CF24 4HQ, U.K
| | - Nikoleta Daskoulidou
- UK Dementia Research Institute Cardiff, Cardiff University, Cardiff CF24 4HQ, U.K
| | - Lewis M. Watkins
- UK Dementia Research Institute Cardiff, Cardiff University, Cardiff CF24 4HQ, U.K
| | - Sarah M. Carpanini
- UK Dementia Research Institute Cardiff, Cardiff University, Cardiff CF24 4HQ, U.K
| | - Wioleta M. Zelek
- UK Dementia Research Institute Cardiff, Cardiff University, Cardiff CF24 4HQ, U.K
| | - B. Paul Morgan
- UK Dementia Research Institute Cardiff, Cardiff University, Cardiff CF24 4HQ, U.K
| |
Collapse
|
10
|
Wu E, Cheng M, Zhang X, Wu T, Sheng S, Sheng M, Wei L, Zhang L, Shao W. Exploration of potential shared gene signatures between periodontitis and multiple sclerosis. BMC Oral Health 2024; 24:75. [PMID: 38218802 PMCID: PMC10788039 DOI: 10.1186/s12903-023-03846-7] [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: 09/13/2023] [Accepted: 12/31/2023] [Indexed: 01/15/2024] Open
Abstract
BACKGROUND Although periodontitis has previously been reported to be linked with multiple sclerosis (MS), but the molecular mechanisms and pathological interactions between the two remain unclear. This study aims to explore potential crosstalk genes and pathways between periodontitis and MS. METHODS Periodontitis and MS data were obtained from the Gene Expression Omnibus (GEO) database. Shared genes were identified by differential expression analysis and weighted gene co-expression network analysis (WGCNA). Then, enrichment analysis for the shared genes was carried out by multiple methods. The least absolute shrinkage and selection operator (LASSO) regression was used to obtain potential shared diagnostic genes. Furthermore, the expression profile of 28 immune cells in periodontitis and MS was examined using single-sample GSEA (ssGSEA). Finally, real-time quantitative fluorescent PCR (qRT-PCR) and immune histochemical staining were employed to validate Hub gene expressions in periodontitis and MS samples. RESULTS FAM46C, SLC7A7, LY96, CFI, DDIT4L, CD14, C5AR1, and IGJ genes were the shared genes between periodontitis, and MS. GO analysis revealed that the shared genes exhibited the greatest enrichment in response to molecules of bacterial origin. LASSO analysis indicated that CFI, DDIT4L, and FAM46C were the most effective shared diagnostic biomarkers for periodontitis and MS, which were further validated by qPCR and immunohistochemical staining. ssGSEA analysis revealed that T and B cells significantly influence the development of MS and periodontitis. CONCLUSIONS FAM46C, SLC7A7, LY96, CFI, DDIT4L, CD14, C5AR1, and IGJ were the most important crosstalk genes between periodontitis, and MS. Further studies found that CFI, DDIT4L, and FAM46C were potential biomarkers in periodontitis and MS.
Collapse
Affiliation(s)
- Erli Wu
- College & Hospital of Stomatology, Key Lab. of Oral Diseases Research of Anhui Province, Anhui Medical University, Hefei, 230032, China
| | - Ming Cheng
- College & Hospital of Stomatology, Key Lab. of Oral Diseases Research of Anhui Province, Anhui Medical University, Hefei, 230032, China
| | - Xinjing Zhang
- College & Hospital of Stomatology, Key Lab. of Oral Diseases Research of Anhui Province, Anhui Medical University, Hefei, 230032, China
| | - Tiangang Wu
- College & Hospital of Stomatology, Key Lab. of Oral Diseases Research of Anhui Province, Anhui Medical University, Hefei, 230032, China
| | - Shuyan Sheng
- First Clinical Medical College (First Affiliated Hospital), Anhui Medical University, Hefei, 230032, China
| | - Mengfei Sheng
- Department of Microbiology and Parasitology, Anhui Provincial Laboratory of Pathogen Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Ling Wei
- The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
| | - Lei Zhang
- College & Hospital of Stomatology, Key Lab. of Oral Diseases Research of Anhui Province, Anhui Medical University, Hefei, 230032, China.
- Department of Periodontology, Anhui Stomatology Hospital affiliated to Anhui Medical University, Hefei, 230032, China.
| | - Wei Shao
- College & Hospital of Stomatology, Key Lab. of Oral Diseases Research of Anhui Province, Anhui Medical University, Hefei, 230032, China.
- Department of Microbiology and Parasitology, Anhui Provincial Laboratory of Pathogen Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
11
|
Okar SV, Fagiani F, Absinta M, Reich DS. Imaging of brain barrier inflammation and brain fluid drainage in human neurological diseases. Cell Mol Life Sci 2024; 81:31. [PMID: 38212566 PMCID: PMC10838199 DOI: 10.1007/s00018-023-05073-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 11/22/2023] [Accepted: 11/29/2023] [Indexed: 01/13/2024]
Abstract
The intricate relationship between the central nervous system (CNS) and the immune system plays a crucial role in the pathogenesis of various neurological diseases. Understanding the interactions among the immunopathological processes at the brain borders is essential for advancing our knowledge of disease mechanisms and developing novel diagnostic and therapeutic approaches. In this review, we explore the emerging role of neuroimaging in providing valuable insights into brain barrier inflammation and brain fluid drainage in human neurological diseases. Neuroimaging techniques have enabled us not only to visualize and assess brain structures, but also to study the dynamics of the CNS in health and disease in vivo. By analyzing imaging findings, we can gain a deeper understanding of the immunopathology observed at the brain-immune interface barriers, which serve as critical gatekeepers that regulate immune cell trafficking, cytokine release, and clearance of waste products from the brain. This review explores the integration of neuroimaging data with immunopathological findings, providing valuable insights into brain barrier integrity and immune responses in neurological diseases. Such integration may lead to the development of novel diagnostic markers and targeted therapeutic approaches that can benefit patients with neurological disorders.
Collapse
Affiliation(s)
- Serhat V Okar
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Francesca Fagiani
- Translational Neuropathology Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Martina Absinta
- Translational Neuropathology Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy.
- Division of Neuroscience, Vita-Salute San Raffaele University, 20132, Milan, Italy.
| | - Daniel S Reich
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
12
|
Wang H, Yu L, Cheng L, Guo Z. The roles of lncRNAs in Th17-associated diseases, with special focus on JAK/STAT signaling pathway. Clin Exp Med 2023; 23:3349-3359. [PMID: 37743424 DOI: 10.1007/s10238-023-01181-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 08/28/2023] [Indexed: 09/26/2023]
Abstract
One of the most crucial T cell subsets in a variety of autoimmune and chronic inflammatory illnesses is T helper (Th) 17 cells. Th17 cells appear to have an essential role in the clearance of extracellular pathogens during infections. However, Th17 cells are also involved in inflammation and have been implicated in the pathogenesis of several autoimmune diseases and human inflammatory conditions. Due to the involvement of Th17 cells in the onset of Th17-associated diseases, understanding molecular mechanisms of Th17 cell functions may open the door to developing tailored therapies to address these difficult disorders. However, the molecular mechanisms governing Th17 differentiation in various diseases are still not well understood. The JAK/STAT signaling pathway plays a critical role in immune responses and has been linked to various aspects of Th17 cell differentiation and function. In this article, we conducted a comprehensive review of various molecular mechanisms (JAK/STAT, microRNAs, etc.), that can affect the differentiation of Th17 cells in various Th17-associated diseases.
Collapse
Affiliation(s)
- Han Wang
- Department of Clinical Laboratory, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130021, China
| | - Lanlan Yu
- Department of Clinical Laboratory, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130021, China
| | - Li Cheng
- Department of Clinical Laboratory, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130021, China
| | - Zhigang Guo
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, 130031, China.
| |
Collapse
|
13
|
Hofmann A, Krajnc N, Dal-Bianco A, Riedl CJ, Zrzavy T, Lerma-Martin C, Kasprian G, Weber CE, Pezzini F, Leutmezer F, Rommer P, Bsteh G, Platten M, Gass A, Berger T, Eisele P, Magliozzi R, Schirmer L, Hametner S. Myeloid cell iron uptake pathways and paramagnetic rim formation in multiple sclerosis. Acta Neuropathol 2023; 146:707-724. [PMID: 37715818 PMCID: PMC10564819 DOI: 10.1007/s00401-023-02627-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 08/01/2023] [Accepted: 08/23/2023] [Indexed: 09/18/2023]
Abstract
In multiple sclerosis (MS), sustained inflammatory activity can be visualized by iron-sensitive magnetic resonance imaging (MRI) at the edges of chronic lesions. These paramagnetic rim lesions (PRLs) are associated with clinical worsening, although the cell type-specific and molecular pathways of iron uptake and metabolism are not well known. We studied two postmortem cohorts: an exploratory formalin-fixed paraffin-embedded (FFPE) tissue cohort of 18 controls and 24 MS cases and a confirmatory snap-frozen cohort of 6 controls and 14 MS cases. Besides myelin and non-heme iron imaging, the haptoglobin-hemoglobin scavenger receptor CD163, the iron-metabolizing markers HMOX1 and HAMP as well as immune-related markers P2RY12, CD68, C1QA and IL10 were visualized in myeloid cell (MC) subtypes at RNA and protein levels across different MS lesion areas. In addition, we studied PRLs in vivo in a cohort of 98 people with MS (pwMS) via iron-sensitive 3 T MRI and haptoglobin genotyping by PCR. CSF samples were available from 38 pwMS for soluble CD163 (sCD163) protein level measurements by ELISA. In postmortem tissues, we observed that iron uptake was linked to rim-associated C1QA-expressing MC subtypes, characterized by upregulation of CD163, HMOX1, HAMP and, conversely, downregulation of P2RY12. We found that pwMS with [Formula: see text] 4 PRLs had higher sCD163 levels in the CSF than pwMS with [Formula: see text] 3 PRLs with sCD163 correlating with the number of PRLs. The number of PRLs was associated with clinical worsening but not with age, sex or haptoglobin genotype of pwMS. However, pwMS with Hp2-1/Hp2-2 haplotypes had higher clinical disability scores than pwMS with Hp1-1. In summary, we observed upregulation of the CD163-HMOX1-HAMP axis in MC subtypes at chronic active lesion rims, suggesting haptoglobin-bound hemoglobin but not transferrin-bound iron as a critical source for MC-associated iron uptake in MS. The correlation of CSF-associated sCD163 with PRL counts in MS highlights the relevance of CD163-mediated iron uptake via haptoglobin-bound hemoglobin. Also, while Hp haplotypes had no noticeable influence on PRL counts, pwMS carriers of a Hp2 allele might have a higher risk to experience clinical worsening.
Collapse
Affiliation(s)
- Annika Hofmann
- Department of Neurology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Nik Krajnc
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Assunta Dal-Bianco
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Christian J Riedl
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Tobias Zrzavy
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Celia Lerma-Martin
- Department of Neurology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Gregor Kasprian
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
- Division of Neuroradiology and Musculoskeletal Radiology, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Claudia E Weber
- Department of Neurology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Francesco Pezzini
- Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of Verona, Verona, Italy
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Fritz Leutmezer
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Paulus Rommer
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Gabriel Bsteh
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Michael Platten
- Department of Neurology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Mannheim Center for Translational Neuroscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Mannheim Institute for Innate Immunity, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany
- DKTK Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center, INF 280, Heidelberg, Germany
| | - Achim Gass
- Department of Neurology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Mannheim Center for Translational Neuroscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Thomas Berger
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Philipp Eisele
- Department of Neurology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Mannheim Center for Translational Neuroscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Roberta Magliozzi
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Lucas Schirmer
- Department of Neurology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
- Mannheim Center for Translational Neuroscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
- Mannheim Institute for Innate Immunity, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
- Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany.
| | - Simon Hametner
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria.
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
14
|
Alruwaili M, Al-kuraishy HM, Alexiou A, Papadakis M, ALRashdi BM, Elhussieny O, Saad HM, Batiha GES. Pathogenic Role of Fibrinogen in the Neuropathology of Multiple Sclerosis: A Tale of Sorrows and Fears. Neurochem Res 2023; 48:3255-3269. [PMID: 37442896 PMCID: PMC10514123 DOI: 10.1007/s11064-023-03981-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 06/20/2023] [Accepted: 06/29/2023] [Indexed: 07/15/2023]
Abstract
Multiple sclerosis (MS) is an autoimmune demyelinating neurodegenerative disease of the central nervous system (CNS) due to injury of the myelin sheath by immune cells. The clotting factor fibrinogen is involved in the pathogenesis of MS by triggering microglia and the progress of neuroinflammation. Fibrinogen level is correlated with MS severity; consequently, inhibition of the fibrinogen cascade may reduce MS neuropathology. Thus, this review aimed to clarify the potential role of fibrinogen in the pathogenesis of MS and how targeting of fibrinogen affects MS neuropathology. Accumulation of fibrinogen in the CNS may occur independently or due to disruption of blood-brain barrier (BBB) integrity in MS. Fibrinogen acts as transduction and increases microglia activation which induces the progression of inflammation, oxidative stress, and neuronal injury. Besides, brain fibrinogen impairs the remyelination process by inhibiting the differentiation of oligodendrocyte precursor cells. These findings proposed that fibrinogen is associated with MS neuropathology through interruption of BBB integrity, induction of neuroinflammation, and demyelination with inhibition of the remyelination process by suppressing oligodendrocytes. Therefore, targeting of fibrinogen and/or CD11b/CD18 receptors by metformin and statins might decrease MS neuropathology. In conclusion, inhibiting the expression of CD11b/CD18 receptors by metformin and statins may decrease the pro-inflammatory effect of fibrinogen on microglia which is involved in the progression of MS.
Collapse
Affiliation(s)
- Mubarak Alruwaili
- Department of Internal Medicine, College of Medicine, Jouf University, Sakaka, Saudi Arabia
| | - Hayder M. Al-kuraishy
- Department of Pharmacology, Toxicology and Medicine, Medical Faculty, College of Medicine, Al-Mustansiriyah University, P.O. Box 14132, Baghdad, Iraq
| | - Athanasios Alexiou
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, NSW 2770 Australia
- AFNP Med, 1030 Vienna, Austria
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, University of Witten-Herdecke, Heusnerstrasse 40, 42283 Wuppertal, Germany
| | - Barakat M. ALRashdi
- Biology Department, College of Science, Jouf University, Sakaka, 41412 Saudi Arabia
| | - Omnya Elhussieny
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Matrouh University, Marsa Matruh, 51744 Egypt
| | - Hebatallah M. Saad
- Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Marsa Matruh, 51744 Egypt
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511 Egypt
| |
Collapse
|
15
|
Tian M, Tang L. Good efficacy achieved by telitacicept, corticosteroids and immunosuppressants in the treatment of SLE combined with MOG-AD. Rheumatol Adv Pract 2023; 7:rkad088. [PMID: 37937177 PMCID: PMC10627278 DOI: 10.1093/rap/rkad088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/10/2023] [Indexed: 11/09/2023] Open
Affiliation(s)
- Mengxue Tian
- Department of Rheumatology and Immunology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lin Tang
- Department of Rheumatology and Immunology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
16
|
Saadh MJ, Arellano MTC, Saini RS, Amin AH, Sharma N, Arias-Gonzáles JL, Alsandook T, Cotrina-Aliaga JC, Akhavan-Sigari R. Molecular mechanisms of long non-coding RNAs in differentiation of T Helper17 cells. Int Immunopharmacol 2023; 123:110728. [PMID: 37572506 DOI: 10.1016/j.intimp.2023.110728] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 07/20/2023] [Accepted: 07/27/2023] [Indexed: 08/14/2023]
Abstract
T helper (Th) 17 cells are one of the most important T cell subsets in a number of autoimmune and chronic inflammatory diseases. During infections, Th17 cells appear to play an important role in the clearance of extracellular pathogens. Th17 cells, on the other hand, are engaged in inflammation and have been linked to the pathophysiology of a number of autoimmune illnesses and human inflammatory disorders. A diverse group of RNA molecules known as lncRNAs serve critical functions in gene expression regulation. They may interact with a wide range of molecules, including DNA, RNA, and proteins, and have a complex structure. LncRNAs, which have restricted or no protein-coding activity, are implicated in a number of illnesses due to their regulatory impact on a variety of biological processes such as cell proliferation, apoptosis, and differentiation. Several lncRNAs have been associated with Th7 cell development in the context of immune cell differentiation. In this article, we cover new studies on the involvement of lncRNAs in Th17 cell differentiation in a variety of disorders, including auto-immune diseases, malignancies, asthma, heart disease, and infections.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman 11831, Jordan; Applied Science Research Center. Applied Science Private University, Amman, Jordan.
| | | | | | - Ali H Amin
- Deanship of Scientific Research, Umm Al-Qura University, Makkah 21955, Saudi Arabia.
| | - Nidhi Sharma
- Department of Computer Engineering & Application, GLA University, Mathura, India.
| | | | - Tahani Alsandook
- Dentistry Department, Al-Turath University College, Baghdad, Iraq.
| | | | - Reza Akhavan-Sigari
- Department of Neurosurgery, University Medical Center Tuebingen, Germany; Department of Health Care Management and Clinical Research, Collegium Humanum Warsaw Management University Warsaw, Poland.
| |
Collapse
|
17
|
Mendiola AS, Yan Z, Dixit K, Johnson JR, Bouhaddou M, Meyer-Franke A, Shin MG, Yong Y, Agrawal A, MacDonald E, Muthukumar G, Pearce C, Arun N, Cabriga B, Meza-Acevedo R, Alzamora MDPS, Zamvil SS, Pico AR, Ryu JK, Krogan NJ, Akassoglou K. Defining blood-induced microglia functions in neurodegeneration through multiomic profiling. Nat Immunol 2023; 24:1173-1187. [PMID: 37291385 PMCID: PMC10307624 DOI: 10.1038/s41590-023-01522-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 04/24/2023] [Indexed: 06/10/2023]
Abstract
Blood protein extravasation through a disrupted blood-brain barrier and innate immune activation are hallmarks of neurological diseases and emerging therapeutic targets. However, how blood proteins polarize innate immune cells remains largely unknown. Here, we established an unbiased blood-innate immunity multiomic and genetic loss-of-function pipeline to define the transcriptome and global phosphoproteome of blood-induced innate immune polarization and its role in microglia neurotoxicity. Blood induced widespread microglial transcriptional changes, including changes involving oxidative stress and neurodegenerative genes. Comparative functional multiomics showed that blood proteins induce distinct receptor-mediated transcriptional programs in microglia and macrophages, such as redox, type I interferon and lymphocyte recruitment. Deletion of the blood coagulation factor fibrinogen largely reversed blood-induced microglia neurodegenerative signatures. Genetic elimination of the fibrinogen-binding motif to CD11b in Alzheimer's disease mice reduced microglial lipid metabolism and neurodegenerative signatures that were shared with autoimmune-driven neuroinflammation in multiple sclerosis mice. Our data provide an interactive resource for investigation of the immunology of blood proteins that could support therapeutic targeting of microglia activation by immune and vascular signals.
Collapse
Affiliation(s)
- Andrew S Mendiola
- Gladstone Institutes, San Francisco, CA, USA
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
| | - Zhaoqi Yan
- Gladstone Institutes, San Francisco, CA, USA
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
| | - Karuna Dixit
- Gladstone Institutes, San Francisco, CA, USA
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
| | | | - Mehdi Bouhaddou
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA, USA
- Quantitative Biosciences Institute, University of California, San Francisco, CA, USA
| | | | | | - Yu Yong
- Gladstone Institutes, San Francisco, CA, USA
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
| | | | - Eilidh MacDonald
- Gladstone Institutes, San Francisco, CA, USA
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
| | | | - Clairice Pearce
- Gladstone Institutes, San Francisco, CA, USA
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
| | - Nikhita Arun
- Gladstone Institutes, San Francisco, CA, USA
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
| | - Belinda Cabriga
- Gladstone Institutes, San Francisco, CA, USA
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
| | - Rosa Meza-Acevedo
- Gladstone Institutes, San Francisco, CA, USA
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
| | - Maria Del Pilar S Alzamora
- Gladstone Institutes, San Francisco, CA, USA
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
| | - Scott S Zamvil
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | | | - Jae Kyu Ryu
- Gladstone Institutes, San Francisco, CA, USA
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Nevan J Krogan
- Gladstone Institutes, San Francisco, CA, USA
- Quantitative Biosciences Institute, University of California, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA
| | - Katerina Akassoglou
- Gladstone Institutes, San Francisco, CA, USA.
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA.
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, USA.
| |
Collapse
|
18
|
Rahmat-Zaie R, Amini J, Haddadi M, Beyer C, Sanadgol N, Zendedel A. TNF-α/STAT1/CXCL10 mutual inflammatory axis that contributes to the pathogenesis of experimental models of multiple sclerosis: A promising signaling pathway for targeted therapies. Cytokine 2023; 168:156235. [PMID: 37267677 DOI: 10.1016/j.cyto.2023.156235] [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: 12/24/2022] [Revised: 05/07/2023] [Accepted: 05/16/2023] [Indexed: 06/04/2023]
Abstract
BACKGROUND Identifying mutual neuroinflammatory axis in different experimental models of multiple sclerosis (MS) is essential to evaluate the de- and re-myelination processes and improve therapeutic interventions' reproducibility. METHODS The expression profile data set of EAE (GSE47900) and cuprizone (GSE100663) models were downloaded from the Gene Expression Omnibus database. The R package and GEO2R software processed these raw chip data. Gene Ontology (GO) functional analysis, KEGG pathway analysis, and protein-protein interaction network analysis were performed to investigate interactions between common differentially expressed genes (DEGs) in all models. Finally, the ELISA method assessed the protein level of highlighted mutual cytokines in serum. RESULTS Our data introduced 59 upregulated [CXCL10, CCL12, and GBP6 as most important] and 17 downregulated [Serpinb1a, Prr18, and Ugt8a as most important] mutual genes. The signal transducer and activator of transcription 1 (STAT1) and CXCL10 were the most crucial hub proteins among mutual upregulated genes. These mutual genes were found to be mainly involved in the TNF-α, TLRs, and complement cascade signaling, and animal models shared 26 mutual genes with MS individuals. Finally, significant upregulation of serum level of TNF-α/IL-1β/CXCL10 cytokines was confirmed in all models in a relatively similar pattern. CONCLUSION For the first time, our study revealed the common neuroinflammatory pathway in animal models of MS and introduced candidate hub genes for better evaluating the preclinical efficacy of pharmacological interventions and designing prospective targeted therapies.
Collapse
Affiliation(s)
- Roya Rahmat-Zaie
- Department of Biology, Faculty of Sciences, University of Zabol, Zabol, Iran
| | - Javad Amini
- Department of Medical Biotechnology and Molecular Science, North Khorasan University of Medical Science, Bojnurd, Iran
| | - Mohammad Haddadi
- Department of Biology, Faculty of Sciences, University of Zabol, Zabol, Iran
| | - Cordian Beyer
- Institute of Neuroanatomy, RWTH University Hospital Aachen, 52074 Aachen, Germany
| | - Nima Sanadgol
- Department of Biology, Faculty of Sciences, University of Zabol, Zabol, Iran; Institute of Neuroanatomy, RWTH University Hospital Aachen, 52074 Aachen, Germany.
| | - Adib Zendedel
- Institute of Anatomy, Department of Biomedicine, University of Basel, 4001 Basel, Switzerland
| |
Collapse
|
19
|
Evans R, Watkins LM, Hawkins K, Santiago G, Demetriou C, Naughton M, Dittmer M, Rees MI, Fitzgerald D, Morgan BP, Neal JW, Howell OW. Complement activation and increased anaphylatoxin receptor expression are associated with cortical grey matter lesions and the compartmentalised inflammatory response of multiple sclerosis. Front Cell Neurosci 2023; 17:1094106. [PMID: 37032838 PMCID: PMC10073739 DOI: 10.3389/fncel.2023.1094106] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 02/07/2023] [Indexed: 04/11/2023] Open
Abstract
Background The extent of cortical pathology is an important determinant of multiple sclerosis (MS) severity. Cortical demyelination and neurodegeneration are related to inflammation of the overlying leptomeninges, a more inflammatory CSF milieu and with parenchymal microglia and astroglia activation. These are all components of the compartmentalised inflammatory response. Compartmentalised inflammation is a feature of progressive MS, which is not targeted by disease modifying therapies. Complement is differentially expressed in the MS CSF and complement, and complement receptors, are associated with demyelination and neurodegeneration. Methods To better understand if complement activation in the leptomeninges is associated with underlying cortical demyelination, inflammation, and microglial activation, we performed a neuropathological study of progressive MS (n = 22, 14 females), neuroinflammatory (n = 8), and non-neurological disease controls (n = 10). We then quantified the relative extent of demyelination, connective tissue inflammation, complement, and complement receptor positive microglia/macrophages. Results Complement was elevated at the leptomeninges, subpial, and within and around vessels of the cortical grey matter. The extent of complement C1q immunoreactivity correlated with connective tissue infiltrates, whilst activation products C4d, Bb, and C3b associated with grey matter demyelination, and C3a receptor 1+ and C5a receptor 1+ microglia/macrophages closely apposed C3b labelled cells. The density of C3a receptor 1+ and C5a receptor 1+ cells was increased at the expanding edge of subpial and leukocortical lesions. C5a receptor 1+ cells expressed TNFα, iNOS and contained puncta immunoreactive for proteolipid protein, neurofilament and synaptophysin, suggesting their involvement in grey matter lesion expansion. Interpretation The presence of products of complement activation at the brain surfaces, their association with the extent of underlying pathology and increased complement anaphylatoxin receptor positive microglia/macrophages at expanding cortical grey matter lesions, could represent a target to modify compartmentalised inflammation and cortical demyelination.
Collapse
Affiliation(s)
- Rhian Evans
- Faculty of Medicine, Health and Life Sciences, Swansea University Medical School, Swansea, United Kingdom
| | - Lewis M. Watkins
- Faculty of Medicine, Health and Life Sciences, Swansea University Medical School, Swansea, United Kingdom
| | - Kristen Hawkins
- Faculty of Medicine, Health and Life Sciences, Swansea University Medical School, Swansea, United Kingdom
| | - Gabriella Santiago
- Faculty of Medicine, Health and Life Sciences, Swansea University Medical School, Swansea, United Kingdom
| | - Constantinos Demetriou
- Faculty of Medicine, Health and Life Sciences, Swansea University Medical School, Swansea, United Kingdom
| | - Michelle Naughton
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast, United Kingdom
| | - Marie Dittmer
- Centre for Experimental Medicine, Queen’s University Belfast, Belfast, United Kingdom
| | - Mark I. Rees
- Faculty of Medicine and Health, The University of Sydney, Darlington, NSW, Australia
| | - Denise Fitzgerald
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast, United Kingdom
| | - B. Paul Morgan
- School of Medicine, UK Dementia Research Institute Cardiff and Systems Immunity Research Institute, Cardiff University, Cardiff, United Kingdom
| | - James W. Neal
- Faculty of Medicine, Health and Life Sciences, Swansea University Medical School, Swansea, United Kingdom
| | - Owain W. Howell
- Faculty of Medicine, Health and Life Sciences, Swansea University Medical School, Swansea, United Kingdom
- *Correspondence: Owain W. Howell,
| |
Collapse
|
20
|
Chronic Bedridden Condition Is Reflected by Substantial Changes in Plasma Inflammatory Profile. Biomolecules 2022; 12:biom12121867. [PMID: 36551295 PMCID: PMC9775060 DOI: 10.3390/biom12121867] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/05/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022] Open
Abstract
Absent or reduced physical activity and spontaneous movement over days, weeks, or even years may lead to problems in almost every major organ/system in the human body. In this study, we investigated whether the dysregulation and alteration of plasma protein inflammatory profiling can stratify chronic bedridden conditions observed in 22 elderly chronic bedridden (CBR) individuals with respect to 11 age-matched active (OLD) controls. By using a combination of immune-assay multiplex techniques, a complex of 27 inflammatory mediators was assessed in the plasma collected from the two groups. A specific plasma protein signature is indeed able to distinguish IPO individuals from age-matched OLD controls; while significantly (p < 0.001) higher protein levels of IL-2, IL-7, and IL-12p70 were measured in the plasma of CBR with respect to OLD individuals, significantly (p < 0.01) higher levels of seven inflammatory mediators, including IL-9, PDGF-b, CCL4 (MIP-1b), CCL5 (RANTES), IL-1Ra, CXCL10 (IP10), and CCL2 (MCP-1), were identified in OLD individuals with respect to CBR individuals. These data suggest that the chronic absence of physical activity may contribute to the dysregulation of a complex molecular pattern occurring with ageing and that specific plasma protein signatures may represent potential biomarkers as well as new potential therapeutic targets for new treatments aimed at improving health expectancy.
Collapse
|
21
|
Magliozzi R, Fadda G, Brown RA, Bar‐Or A, Howell OW, Hametner S, Marastoni D, Poli A, Nicholas R, Calabrese M, Monaco S, Reynolds R. "Ependymal-in" Gradient of Thalamic Damage in Progressive Multiple Sclerosis. Ann Neurol 2022; 92:670-685. [PMID: 35748636 PMCID: PMC9796378 DOI: 10.1002/ana.26448] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 06/17/2022] [Accepted: 06/22/2022] [Indexed: 01/01/2023]
Abstract
Leptomeningeal and perivenular infiltrates are important contributors to cortical grey matter damage and disease progression in multiple sclerosis (MS). Whereas perivenular inflammation induces vasculocentric lesions, leptomeningeal involvement follows a subpial "surface-in" gradient. To determine whether similar gradient of damage occurs in deep grey matter nuclei, we examined the dorsomedial thalamic nuclei and cerebrospinal fluid (CSF) samples from 41 postmortem secondary progressive MS cases compared with 5 non-neurological controls and 12 controls with other neurological diseases. CSF/ependyma-oriented gradient of reduction in NeuN+ neuron density was present in MS thalamic lesions compared to controls, greatest (26%) in subventricular locations at the ependyma/CSF boundary and least with increasing distance (12% at 10 mm). Concomitant graded reduction in SMI31+ axon density was observed, greatest (38%) at 2 mm from the ependyma/CSF boundary and least at 10 mm (13%). Conversely, gradient of major histocompatibility complex (MHC)-II+ microglia density increased by over 50% at 2 mm at the ependyma/CSF boundary and only by 15% at 10 mm and this gradient inversely correlated with the neuronal (R = -0.91, p < 0.0001) and axonal (R = -0.79, p < 0.0001) thalamic changes. Observed gradients were also detected in normal-appearing thalamus and were associated with rapid/severe disease progression; presence of leptomeningeal tertiary lymphoid-like structures; large subependymal infiltrates, enriched in CD20+ B cells and occasionally containing CXCL13+ CD35+ follicular dendritic cells; and high CSF protein expression of a complex pattern of soluble inflammatory/neurodegeneration factors, including chitinase-3-like-1, TNFR1, parvalbumin, neurofilament-light-chains and TNF. Substantial "ependymal-in" gradient of pathological cell alterations, accompanied by presence of intrathecal inflammation, compartmentalized either in subependymal lymphoid perivascular infiltrates or in CSF, may play a key role in MS progression. SUMMARY FOR SOCIAL MEDIA: Imaging and neuropathological evidences demonstrated the unique feature of "surface-in" gradient of damage in multiple sclerosis (MS) since early pediatric stages, often associated with more severe brain atrophy and disease progression. In particular, increased inflammation in the cerebral meninges has been shown to be strictly associated with an MS-specific gradient of neuronal, astrocyte, and oligodendrocyte loss accompanied by microglial activation in subpial cortical layers, which is not directly related to demyelination. To determine whether a similar gradient of damage occurs in deep grey matter nuclei, we examined the potential neuronal and microglia alterations in the dorsomedial thalamic nuclei from postmortem secondary progressive MS cases in combination with detailed neuropathological characterization of the inflammatory features and protein profiling of paired CSF samples. We observed a substantial "subependymal-in" gradient of neuro-axonal loss and microglia activation in active thalamic lesions of progressive MS cases, in particular in the presence of increased leptomeningeal and cerebrospinal fluid (CSF) inflammation. This altered graded pathology was found associated with more severe and rapid progressive MS and increased inflammatory degree either in large perivascular subependymal infiltrates, enriched in B cells, or within the paired CSF, in particular with elevated levels of a complex pattern of soluble inflammatory and neurodegeneration factors, including chitinase 3-like-1, TNFR1, parvalbumin, neurofilament light-chains and TNF. These data support a key role for chronic, intrathecally compartmentalized inflammation in specific disease endophenotypes. CSF biomarkers, together with advance imaging tools, may therefore help to improve not only the disease diagnosis but also the early identification of specific MS subgroups that would benefit of more personalized treatments. ANN NEUROL 2022;92:670-685.
Collapse
Affiliation(s)
- Roberta Magliozzi
- Neurology Section of Department of Neurological and Movement SciencesUniversity of VeronaVeronaItaly,Department of Brain Sciences, Faculty of MedicineImperial College LondonLondonUK
| | - Giulia Fadda
- Center for Neuroinflammation and Experimental Therapeutics and the Department of Neurology, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | | | - Amit Bar‐Or
- Center for Neuroinflammation and Experimental Therapeutics and the Department of Neurology, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Owain W. Howell
- Department of Brain Sciences, Faculty of MedicineImperial College LondonLondonUK,Institute of Life SciencesSwansea UniversitySwanseaUK
| | - Simon Hametner
- Brain Research CenterMedical University of ViennaViennaAustria
| | - Damiano Marastoni
- Neurology Section of Department of Neurological and Movement SciencesUniversity of VeronaVeronaItaly
| | - Alberto Poli
- Neurology Section of Department of Neurological and Movement SciencesUniversity of VeronaVeronaItaly
| | - Richard Nicholas
- Department of Brain Sciences, Faculty of MedicineImperial College LondonLondonUK
| | - Massimiliano Calabrese
- Neurology Section of Department of Neurological and Movement SciencesUniversity of VeronaVeronaItaly
| | - Salvatore Monaco
- Neurology Section of Department of Neurological and Movement SciencesUniversity of VeronaVeronaItaly
| | - Richard Reynolds
- Department of Brain Sciences, Faculty of MedicineImperial College LondonLondonUK,Centre for Molecular Neuropathology, Lee Kong Chian School of MedicineNanyang Technological UniversitySingaporeSingapore
| |
Collapse
|
22
|
Kaisey M, Lashgari G, Fert-Bober J, Ontaneda D, Solomon AJ, Sicotte NL. An Update on Diagnostic Laboratory Biomarkers for Multiple Sclerosis. Curr Neurol Neurosci Rep 2022; 22:675-688. [PMID: 36269540 DOI: 10.1007/s11910-022-01227-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/12/2022] [Indexed: 01/27/2023]
Abstract
PURPOSE For many patients, the multiple sclerosis (MS) diagnostic process can be lengthy, costly, and fraught with error. Recent research aims to address the unmet need for an accurate and simple diagnostic process through discovery of novel diagnostic biomarkers. This review summarizes recent studies on MS diagnostic fluid biomarkers, with a focus on blood biomarkers, and includes discussion of technical limitations and practical applicability. RECENT FINDINGS This line of research is in its early days. Accurate and easily obtainable biomarkers for MS have not yet been identified and validated, but several approaches to uncover them are underway. Continue efforts to define laboratory diagnostic biomarkers are likely to play an increasingly important role in defining MS at the earliest stages, leading to better long-term clinical outcomes.
Collapse
Affiliation(s)
- Marwa Kaisey
- Cedars-Sinai Medical Center Department of Neurology, 127 S. San Vicente Blvd, A6600, Los Angeles, CA, 90048, USA.
| | - Ghazal Lashgari
- Cedars-Sinai Medical Center Department of Neurology, 127 S. San Vicente Blvd, A6600, Los Angeles, CA, 90048, USA
| | - Justyna Fert-Bober
- Cedars-Sinai Medical Center Department of Neurology, 127 S. San Vicente Blvd, A6600, Los Angeles, CA, 90048, USA
| | - Daniel Ontaneda
- Mellen Center for Multiple Sclerosis, Neurological Institute, Cleveland Clinic, 9500 Euclid Ave. U10 Mellen Center, Cleveland, OH, 44106, USA
| | - Andrew J Solomon
- Department of Neurological Sciences, Larner College of Medicine at the University of Vermont University Health Center, Arnold 2, 1 South Prospect Street, Burlington, VT, 05401, USA
| | - Nancy L Sicotte
- Cedars-Sinai Medical Center Department of Neurology, 127 S. San Vicente Blvd, A6600, Los Angeles, CA, 90048, USA
| |
Collapse
|
23
|
Alberio T, Brughera M, Lualdi M. Current Insights on Neurodegeneration by the Italian Proteomics Community. Biomedicines 2022; 10:biomedicines10092297. [PMID: 36140397 PMCID: PMC9496271 DOI: 10.3390/biomedicines10092297] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/29/2022] [Accepted: 09/13/2022] [Indexed: 12/02/2022] Open
Abstract
The growing number of patients affected by neurodegenerative disorders represents a huge problem for healthcare systems, human society, and economics. In this context, omics strategies are crucial for the identification of molecular factors involved in disease pathobiology, and for the discovery of biomarkers that allow early diagnosis, patients’ stratification, and treatment response prediction. The integration of different omics data is a required step towards the goal of personalized medicine. The Italian proteomics community is actively developing and applying proteomics approaches to the study of neurodegenerative disorders; moreover, it is leading the mitochondria-focused initiative of the Human Proteome Project, which is particularly important given the central role of mitochondrial impairment in neurodegeneration. Here, we describe how Italian research groups in proteomics have contributed to the knowledge of many neurodegenerative diseases, through the elucidation of the pathobiology of these disorders, and through the discovery of disease biomarkers. In particular, we focus on the central role of post-translational modifications analysis, the implementation of network-based approaches in functional proteomics, the integration of different omics in a systems biology view, and the development of novel platforms for biomarker discovery for the high-throughput quantification of thousands of proteins at a time.
Collapse
|
24
|
Saez-Calveras N, Stuve O. The role of the complement system in Multiple Sclerosis: A review. Front Immunol 2022; 13:970486. [PMID: 36032156 PMCID: PMC9399629 DOI: 10.3389/fimmu.2022.970486] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 07/21/2022] [Indexed: 11/13/2022] Open
Abstract
The complement system has been involved in the pathogenesis of multiple neuroinflammatory and neurodegenerative conditions. In this review, we evaluated the possible role of complement activation in multiple sclerosis (MS) with a focus in progressive MS, where the disease pathogenesis remains to be fully elucidated and treatment options are limited. The evidence for the involvement of the complement system in the white matter plaques and gray matter lesions of MS stems from immunohistochemical analysis of post-mortem MS brains, in vivo serum and cerebrospinal fluid biomarker studies, and animal models of Experimental Autoimmune Encephalomyelitis (EAE). Complement knock-out studies in these animal models have revealed that this system may have a “double-edge sword” effect in MS. On the one hand, complement proteins may aid in promoting the clearance of myelin degradation products and other debris through myeloid cell-mediated phagocytosis. On the other, its aberrant activation may lead to demyelination at the rim of progressive MS white matter lesions as well as synapse loss in the gray matter. The complement system may also interact with known risk factors of MS, including as Epstein Barr Virus (EBV) infection, and perpetuate the activation of CNS self-reactive B cell populations. With the mounting evidence for the involvement of complement in MS, the development of complement modulating therapies for this condition is appealing. Herein, we also reviewed the pharmacological complement inhibitors that have been tested in MS animal models as well as in clinical trials for other neurologic diseases. The potential use of these agents, such as the C5-binding antibody eculizumab in MS will require a detailed understanding of the role of the different complement effectors in this disease and the development of better CNS delivery strategies for these compounds.
Collapse
Affiliation(s)
- Nil Saez-Calveras
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Olaf Stuve
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Neurology Section, Veterans Affairs (VA) North Texas Health Care System, Dallas, TX, United States
- *Correspondence: Olaf Stuve,
| |
Collapse
|
25
|
Pan L, Peng C, Wang L, Li L, Huang S, Fei C, Wang N, Chu F, Peng D, Duan X. Network pharmacology and experimental validation-based approach to understand the effect and mechanism of Taohong Siwu Decoction against ischemic stroke. JOURNAL OF ETHNOPHARMACOLOGY 2022; 294:115339. [PMID: 35525530 DOI: 10.1016/j.jep.2022.115339] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 04/16/2022] [Accepted: 04/29/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Taohong Siwu Decoction (THSWD) is a classic prescription of traditional Chinese medicine that is mainly used for promoting blood circulation and alleviating blood stasis. THSWD is composed of Prunus persica (L.) Batsch, Carthamus tinctorius L., Ligusticum chuanxiong hort, Angelica sinensis (Oliv.) Diels, Rehmannia glutinosa (Gaertn.) DC, and Paeoniae Radix Alba. This prescription eliminates blood stasis, supplements blood, and dredges the body as an auxiliary treatment. AIM OF THE STUDY To investigate the mechanistic effects of THSWD in the treatment of cerebral ischemia. MATERIALS AND METHODS we downloaded 39 blood components for THSWD from the PharmMapper database for target prediction studies and identified the targets of cerebral ischemia. We identified the intersection between the components and targets, constructed a protein-protein interaction (PPI) network, carried out GO and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis. a rat model of cerebral ischemia was established in rats, and the results of network pharmacology were verified by in vivo experiments. RESULTS Established a component-target-pathway network, further transcriptomics analysis identified a total of 11 target genes (Plau, Fabp4, Mmp9, Mmp12, Cfd, Lcn2, Trem1, Lgals3, Hmox1, Selp and Slc6a4), a total of seven pathways (focal adhesion, complement and coagulation cascades, Staphylococcus aureus infection, malaria, transcriptional dysregulation in cancer, progesterone-mediated oocyte maturation, and the PI3K-Akt signaling pathway), because both targets genes and the complement and coagulation cascade signaling pathways mediate inflammatory responses, the signaling pathways associated with the complement and coagulation cascades were selected for experimental verification. We detected inflammatory factors and several key proteins in the complement and coagulation cascade signaling pathway (C1qb, C1qc, C3ar1, C5ar1, and Cfd). Analysis showed that THSWD can reduce the release of inflammatory factors and inhibit activation of the complement signaling pathways, thereby protecting against ischemic stroke disease. CONCLUSIONS Our findings provide preliminary clarification of the predominant mechanism of action of THSWD when used to treat ischemic stroke.
Collapse
Affiliation(s)
- Lingyu Pan
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Can Peng
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Lei Wang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Lili Li
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Shi Huang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Changyi Fei
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Ni Wang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Furui Chu
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Daiyin Peng
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China; College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Xianchun Duan
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China.
| |
Collapse
|
26
|
Baldacchino K, Peveler WJ, Lemgruber L, Smith RS, Scharler C, Hayden L, Komarek L, Lindsay SL, Barnett SC, Edgar JM, Linington C, Thümmler K. Myelinated axons are the primary target of hemin-mediated oxidative damage in a model of the central nervous system. Exp Neurol 2022; 354:114113. [PMID: 35569511 DOI: 10.1016/j.expneurol.2022.114113] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 04/29/2022] [Accepted: 05/08/2022] [Indexed: 12/01/2022]
Abstract
Iron released from oligodendrocytes during demyelination or derived from haemoglobin breakdown products is believed to amplify oxidative tissue injury in multiple sclerosis (MS). However, the pathophysiological significance of iron-containing haemoglobin breakdown products themselves is rarely considered in the context of MS and their cellular specificity and mode of action remain unclear. Using myelinating cell cultures, we now report the cytotoxic potential of hemin (ferriprotoporphyrin IX chloride), a major degradation product of haemoglobin, is 25-fold greater than equimolar concentrations of free iron in myelinating cultures; a model that reproduces the complex multicellular environment of the CNS. At low micro molar concentrations (3.3 - 10 μM) we observed hemin preferentially binds to myelin and axons to initiate a complex detrimental response that results in targeted demyelination and axonal loss but spares neuronal cell bodies, astrocytes and the majority of oligodendroglia. Demyelination and axonal loss in this context are executed by a combination of mechanisms that include iron-dependent peroxidation by reactive oxygen species (ROS) and ferroptosis. These effects are microglial-independent, do not require any initiating inflammatory insult and represent a direct effect that compromises the structural integrity of myelinated axons in the CNS. Our data identify hemin-mediated demyelination and axonal loss as a novel mechanism by which intracerebral degradation of haemoglobin may contribute to lesion development in MS.
Collapse
Affiliation(s)
- Karl Baldacchino
- Institute of Infection, Immunity and Inflammation, University of Glasgow, G12 8TA Glasgow, United Kingdom
| | - William J Peveler
- WestCHEM, School of Chemistry, University of Glasgow, Joseph Black Building, G12 8QQ Glasgow, UK
| | - Leandro Lemgruber
- Glasgow Imaging Facility, Institute of Infection, Immunity and Inflammation, University of Glasgow, University Avenue, Glasgow G12 8QQ, UK
| | - Rebecca Sherrard Smith
- Institute of Infection, Immunity and Inflammation, University of Glasgow, G12 8TA Glasgow, United Kingdom
| | - Cornelia Scharler
- Institute of Experimental and Clinical Cell Therapy, Paracelsus Medical University, Salzburg, Austria
| | - Lorna Hayden
- Institute of Infection, Immunity and Inflammation, University of Glasgow, G12 8TA Glasgow, United Kingdom
| | - Lina Komarek
- Institute of Infection, Immunity and Inflammation, University of Glasgow, G12 8TA Glasgow, United Kingdom
| | - Susan L Lindsay
- Institute of Infection, Immunity and Inflammation, University of Glasgow, G12 8TA Glasgow, United Kingdom
| | - Susan C Barnett
- Institute of Infection, Immunity and Inflammation, University of Glasgow, G12 8TA Glasgow, United Kingdom
| | - Julia M Edgar
- Institute of Infection, Immunity and Inflammation, University of Glasgow, G12 8TA Glasgow, United Kingdom
| | - Christopher Linington
- Institute of Infection, Immunity and Inflammation, University of Glasgow, G12 8TA Glasgow, United Kingdom
| | - Katja Thümmler
- Institute of Infection, Immunity and Inflammation, University of Glasgow, G12 8TA Glasgow, United Kingdom.
| |
Collapse
|
27
|
Bierhansl L, Hartung HP, Aktas O, Ruck T, Roden M, Meuth SG. Thinking outside the box: non-canonical targets in multiple sclerosis. Nat Rev Drug Discov 2022; 21:578-600. [PMID: 35668103 PMCID: PMC9169033 DOI: 10.1038/s41573-022-00477-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2022] [Indexed: 12/11/2022]
Abstract
Multiple sclerosis (MS) is an immune-mediated disease of the central nervous system that causes demyelination, axonal degeneration and astrogliosis, resulting in progressive neurological disability. Fuelled by an evolving understanding of MS immunopathogenesis, the range of available immunotherapies for clinical use has expanded over the past two decades. However, MS remains an incurable disease and even targeted immunotherapies often fail to control insidious disease progression, indicating the need for new and exceptional therapeutic options beyond the established immunological landscape. In this Review, we highlight such non-canonical targets in preclinical MS research with a focus on five highly promising areas: oligodendrocytes; the blood-brain barrier; metabolites and cellular metabolism; the coagulation system; and tolerance induction. Recent findings in these areas may guide the field towards novel targets for future therapeutic approaches in MS.
Collapse
Affiliation(s)
- Laura Bierhansl
- Department of Neurology, Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Orhan Aktas
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Tobias Ruck
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Michael Roden
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Department of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
- German Center of Diabetes Research, Partner Düsseldorf, Neuherberg, Germany
| | - Sven G Meuth
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
28
|
Cerebral Iron Deposition in Neurodegeneration. Biomolecules 2022; 12:biom12050714. [PMID: 35625641 PMCID: PMC9138489 DOI: 10.3390/biom12050714] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 05/12/2022] [Accepted: 05/13/2022] [Indexed: 02/04/2023] Open
Abstract
Disruption of cerebral iron regulation appears to have a role in aging and in the pathogenesis of various neurodegenerative disorders. Possible unfavorable impacts of iron accumulation include reactive oxygen species generation, induction of ferroptosis, and acceleration of inflammatory changes. Whole-brain iron-sensitive magnetic resonance imaging (MRI) techniques allow the examination of macroscopic patterns of brain iron deposits in vivo, while modern analytical methods ex vivo enable the determination of metal-specific content inside individual cell-types, sometimes also within specific cellular compartments. The present review summarizes the whole brain, cellular, and subcellular patterns of iron accumulation in neurodegenerative diseases of genetic and sporadic origin. We also provide an update on mechanisms, biomarkers, and effects of brain iron accumulation in these disorders, focusing on recent publications. In Parkinson’s disease, Friedreich’s disease, and several disorders within the neurodegeneration with brain iron accumulation group, there is a focal siderosis, typically in regions with the most pronounced neuropathological changes. The second group of disorders including multiple sclerosis, Alzheimer’s disease, and amyotrophic lateral sclerosis shows iron accumulation in the globus pallidus, caudate, and putamen, and in specific cortical regions. Yet, other disorders such as aceruloplasminemia, neuroferritinopathy, or Wilson disease manifest with diffuse iron accumulation in the deep gray matter in a pattern comparable to or even more extensive than that observed during normal aging. On the microscopic level, brain iron deposits are present mostly in dystrophic microglia variably accompanied by iron-laden macrophages and in astrocytes, implicating a role of inflammatory changes and blood–brain barrier disturbance in iron accumulation. Options and potential benefits of iron reducing strategies in neurodegeneration are discussed. Future research investigating whether genetic predispositions play a role in brain Fe accumulation is necessary. If confirmed, the prevention of further brain Fe uptake in individuals at risk may be key for preventing neurodegenerative disorders.
Collapse
|
29
|
Proteomics in Multiple Sclerosis: The Perspective of the Clinician. Int J Mol Sci 2022; 23:ijms23095162. [PMID: 35563559 PMCID: PMC9100097 DOI: 10.3390/ijms23095162] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/26/2022] [Accepted: 05/02/2022] [Indexed: 02/08/2023] Open
Abstract
Multiple sclerosis (MS) is the inflammatory demyelinating and neurodegenerative disease of the central nervous system (CNS) that affects approximately 2.8 million people worldwide. In the last decade, a new era was heralded in by a new phenotypic classification, a new diagnostic protocol and the first ever therapeutic guideline, making personalized medicine the aim of MS management. However, despite this great evolution, there are still many aspects of the disease that are unknown and need to be further researched. A hallmark of these research are molecular biomarkers that could help in the diagnosis, differential diagnosis, therapy and prognosis of the disease. Proteomics, a rapidly evolving discipline of molecular biology may fulfill this dire need for the discovery of molecular biomarkers. In this review, we aimed to give a comprehensive summary on the utility of proteomics in the field of MS research. We reviewed the published results of the method in case of the pathogenesis of the disease and for biomarkers of diagnosis, differential diagnosis, conversion of disease courses, disease activity, progression and immunological therapy. We found proteomics to be a highly effective emerging tool that has been providing important findings in the research of MS.
Collapse
|
30
|
Elkjaer ML, Röttger R, Baumbach J, Illes Z. A Systematic Review of Tissue and Single Cell Transcriptome/Proteome Studies of the Brain in Multiple Sclerosis. Front Immunol 2022; 13:761225. [PMID: 35309325 PMCID: PMC8924618 DOI: 10.3389/fimmu.2022.761225] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 01/28/2022] [Indexed: 11/27/2022] Open
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating and degenerative disease of the central nervous system (CNS). Although inflammatory responses are efficiently treated, therapies for progression are scarce and suboptimal, and biomarkers to predict the disease course are insufficient. Cure or preventive measures for MS require knowledge of core pathological events at the site of the tissue damage. Novelties in systems biology have emerged and paved the way for a more fine-grained understanding of key pathological pathways within the CNS, but they have also raised questions still without answers. Here, we systemically review the power of tissue and single-cell/nucleus CNS omics and discuss major gaps of integration into the clinical practice. Systemic search identified 49 transcriptome and 11 proteome studies of the CNS from 1997 till October 2021. Pioneering molecular discoveries indicate that MS affects the whole brain and all resident cell types. Despite inconsistency of results, studies imply increase in transcripts/proteins of semaphorins, heat shock proteins, myelin proteins, apolipoproteins and HLAs. Different lesions are characterized by distinct astrocytic and microglial polarization, altered oligodendrogenesis, and changes in specific neuronal subtypes. In all white matter lesion types, CXCL12, SCD, CD163 are highly expressed, and STAT6- and TGFβ-signaling are increased. In the grey matter lesions, TNF-signaling seems to drive cell death, and especially CUX2-expressing neurons may be susceptible to neurodegeneration. The vast heterogeneity at both cellular and lesional levels may underlie the clinical heterogeneity of MS, and it may be more complex than the current disease phenotyping in the clinical practice. Systems biology has not solved the mystery of MS, but it has discovered multiple molecules and networks potentially contributing to the pathogenesis. However, these results are mostly descriptive; focused functional studies of the molecular changes may open up for a better interpretation. Guidelines for acceptable quality or awareness of results from low quality data, and standardized computational and biological pipelines may help to overcome limited tissue availability and the “snap shot” problem of omics. These may help in identifying core pathological events and point in directions for focus in clinical prevention.
Collapse
Affiliation(s)
- Maria L Elkjaer
- Department of Neurology, Odense University Hospital, Odense, Denmark.,Institute of Clinical Research, University of Southern Denmark, Odense, Denmark.,Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Richard Röttger
- Department of Mathematics and Computer Science, University of Southern Denmark, Odense, Denmark
| | - Jan Baumbach
- Chair of Computational Systems Biology, University of Hamburg, Hamburg, Germany
| | - Zsolt Illes
- Department of Neurology, Odense University Hospital, Odense, Denmark.,Institute of Clinical Research, University of Southern Denmark, Odense, Denmark.,Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
31
|
Schaller-Paule MA, Yalachkov Y, Steinmetz H, Friedauer L, Hattingen E, Miesbach W, Weber F, Kirchmayr K, Schaefer JH, Foerch C. Analysis of CSF D-Dimer to Identify Intrathecal Fibrin-Driven Autoimmunity in Patients With Multiple Sclerosis. NEUROLOGY - NEUROIMMUNOLOGY NEUROINFLAMMATION 2022; 9:9/3/e1150. [PMID: 35260469 PMCID: PMC8906189 DOI: 10.1212/nxi.0000000000001150] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 01/06/2022] [Indexed: 12/02/2022]
Abstract
Background and Objectives Proteins of the coagulation system contribute to autoimmune inflammation in patients with multiple sclerosis (MS). On blood-brain barrier (BBB) disruption, fibrinogen enters the CNS and is rapidly converted to fibrin, unfolding pleiotropic autoimmune mechanisms. Fibrin accumulation leads to subsequent proteolytic degradation that results in D-dimer generation. The primary objective of this study was to determine intrathecal levels of D-dimer in CSF as a measure of intrathecal coagulation cascade activation and to evaluate its diagnostic utility in patients with MS in contrast to healthy subjects. Key secondary objectives included analysis of CSF D-dimer in differential diagnoses of MS and its relation to routine clinical markers of disease activity. Methods Patients admitted for the assessment of suspected MS were prospectively recruited from October 2017 to December 2020. Blood plasma and citrated CSF samples were analyzed using a highly sensitive luminescent oxygen channeling immunoassay. Intrathecal generation of D-dimer was analyzed by adjusting for CSF/serum albumin (Qalb) and CSF/plasma D-dimer quotients (QD-dimer), and corresponding CSF fibrinogen levels were determined. Final diagnoses after full evaluation and clinical data were recorded. Results Of 187 patients, 113 patients received a diagnosis of MS or clinically/radiologically isolated syndrome. We found increased intrathecal CSF D-dimer generation levels (QD-dimer/Qalb-index) for patients with relapsing-remitting MS (RRMS; n = 71, median 4.7, interquartile range [IQR] 2.5–8.0) when compared with those for disease controls (n = 22, median 2.6, IQR 2.1–4.8, p = 0.031). Absolute CSF D-dimer values correlated with CSF fibrinogen levels (r = 0.463; p < 0 .001) and CSF leukocytes (r = 0.273; p = 0.003) and were elevated in MS patients with contrast enhancement (CE) compared with MS patients without CE on MRI (n = 48, median 6 ng/mL, and IQR 3–15.25 vs n = 41, median 4 ng/mL, and IQR 2–7; p = 0.026). Exploratory subgroup analyses indicated a correlation of intrathecal inflammatory activity and CSF D-dimer levels. Discussion D-dimer in CSF can be reliably determined and correlates with markers of CNS inflammation and CSF fibrinogen levels. Adjusted for BBB dysfunction, CSF D-dimer may allow the identification of intrathecal coagulation cascade activation in patients with MS. Classification of Evidence This study provides Class I evidence that CSF D-dimer levels are elevated in patients with RRMS.
Collapse
|
32
|
Abbadessa G, Miele G, Di Pietro A, Sparaco M, Palladino R, Armetta I, D'Elia G, Trojsi F, Signoriello E, Lus G, Lavorgna L, Bonavita S. Multiple sclerosis and genetic polymorphisms in fibrinogen-mediated hemostatic pathways: a case-control study. Neurol Sci 2022; 43:2601-2609. [PMID: 34561786 PMCID: PMC8918146 DOI: 10.1007/s10072-021-05608-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 09/10/2021] [Indexed: 11/01/2022]
Abstract
INTRODUCTION Blood coagulation constituents might exert immunomodulatory functions in the CNS and may trigger neuroinflammation and demyelination. We evaluated whether particular single-nucleotide polymorphisms (SNPs), thought to be involved in fibrinogen-mediated hemostatic pathways, are overrepresented in patients with MS compared with controls. METHODS The case-control study consisted of 119 MS patients recruited consecutively at our clinic, and 68 healthy controls. Afterwards, we created a cumulative genetic risk score (CGRS) which included the 5 selected hemostatic risk alleles (Beta-Fibrinogen 455G/A, Glycoprotein IIIa P1A2, Factor V Leiden, Factor V H2R, and Prothrombin 20210G/A). Multivariate ordinal logistic regression and multivariate multinomial logistic regression were applied to evaluate the effect of CGRS on MS susceptibility. RESULTS The FGB 455 G/A and Factor V H1299R variants might be associated with MS status, in the recessive and dominant model, respectively. A cumulative association of the five SNPs investigated with the disease was observed. DISCUSSION We found that MS patients carried more pro-hemostatic variants than healthy controls. An increasing number of unfavorable alleles might increase the likelihood of being in the MS group, in the cumulative analysis. Our findings encourage to evaluating these variants in a larger population-based cohort.
Collapse
Affiliation(s)
- Gianmarco Abbadessa
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy.
| | - Giuseppina Miele
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Andrea Di Pietro
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Maddalena Sparaco
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Raffaele Palladino
- Department of Public Health, University Federico II, Naples, Italy
- Department of Primary Care and Public Health, Imperial College London, London, UK
| | - Ignazio Armetta
- Clinical and Molecular Pathology, Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Giovanna D'Elia
- Clinical and Molecular Pathology, Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Francesca Trojsi
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Elisabetta Signoriello
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Giacomo Lus
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Luigi Lavorgna
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Simona Bonavita
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| |
Collapse
|
33
|
Ahmed SM, Fransen NL, Touil H, Michailidou I, Huitinga I, Gommerman JL, Bar-Or A, Ramaglia V. Accumulation of meningeal lymphocytes correlates with white matter lesion activity in progressive multiple sclerosis. JCI Insight 2022; 7:151683. [PMID: 35104246 PMCID: PMC8983127 DOI: 10.1172/jci.insight.151683] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 01/28/2022] [Indexed: 11/17/2022] Open
Abstract
Subpial cortical demyelination is an important component of multiple sclerosis (MS) pathology contributing to disease progression, yet mechanism(s) underlying its development remain unclear. Compartmentalized inflammation involving the meninges may drive this type of injury. Given recent findings identifying substantial white matter (WM) lesion activity in patients with progressive MS, elucidating whether and how WM lesional activity relates to meningeal inflammation and subpial cortical injury is of interest. Using postmortem FFPE tissue blocks (range, 5-72 blocks; median, 30 blocks) for each of 27 patients with progressive MS, we assessed the relationship between meningeal inflammation, the extent of subpial cortical demyelination, and the state of subcortical WM lesional activity. Meningeal accumulations of T cells and B cells, but not myeloid cells, were spatially adjacent to subpial cortical lesions, and greater immune cell accumulation was associated with larger subpial lesion areas. Patients with a higher extent of meningeal inflammation harbored a greater proportion of active and mixed active/inactive WM lesions and an overall lower proportion of inactive and remyelinated WM lesions. Our findings support the involvement of meningeal lymphocytes in subpial cortical injury and point to a potential link between inflammatory subpial cortical demyelination and pathological mechanisms occurring in the subcortical WM.
Collapse
Affiliation(s)
- Shanzeh M. Ahmed
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Nina L. Fransen
- Department of Neuroimmunology, Netherlands Institute for Neuroscience, Meibergdreef, Amsterdam, Netherlands.,Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Hanane Touil
- Department of Neurology and Center for Neuroinflammation and Neurotherapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Iliana Michailidou
- Department of Clinical Genetics, Leiden University Medical Center, Einthovenweg, Leiden, Netherlands
| | - Inge Huitinga
- Department of Neuroimmunology, Netherlands Institute for Neuroscience, Meibergdreef, Amsterdam, Netherlands.,Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands
| | | | - Amit Bar-Or
- Department of Neurology and Center for Neuroinflammation and Neurotherapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Valeria Ramaglia
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
34
|
Cooze BJ, Dickerson M, Loganathan R, Watkins LM, Grounds E, Pearson BR, Bevan RJ, Morgan BP, Magliozzi R, Reynolds R, Neal JW, Howell OW. The association between neurodegeneration and local complement activation in the thalamus to progressive multiple sclerosis outcome. Brain Pathol 2022; 32:e13054. [PMID: 35132719 PMCID: PMC9425007 DOI: 10.1111/bpa.13054] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 12/17/2021] [Accepted: 01/17/2022] [Indexed: 01/22/2023] Open
Abstract
The extent of grey matter demyelination and neurodegeneration in the progressive multiple sclerosis (PMS) brains at post‐mortem associates with more severe disease. Regional tissue atrophy, especially affecting the cortical and deep grey matter, including the thalamus, is prognostic for poor outcomes. Microglial and complement activation are important in the pathogenesis and contribute to damaging processes that underlie tissue atrophy in PMS. We investigated the extent of pathology and innate immune activation in the thalamus in comparison to cortical grey and white matter in blocks from 21 cases of PMS and 10 matched controls. Using a digital pathology workflow, we show that the thalamus is invariably affected by demyelination and had a far higher proportion of active inflammatory lesions than forebrain cortical tissue blocks from the same cases. Lesions were larger and more frequent in the medial nuclei near the ventricular margin, whilst neuronal loss was greatest in the lateral thalamic nuclei. The extent of thalamic neuron loss was not associated with thalamic demyelination but correlated with the burden of white matter pathology in other forebrain areas (Spearman r = 0.79, p < 0.0001). Only thalamic neuronal loss, and not that seen in other forebrain cortical areas, correlated with disease duration (Spearman r = −0.58, p = 0.009) and age of death (Spearman r = −0.47, p = 0.045). Immunoreactivity for the complement pattern recognition molecule C1q, and products of complement activation (C4d, Bb and C3b) were elevated in thalamic lesions with an active inflammatory pathology. Complement regulatory protein, C1 inhibitor, was unchanged in expression. We conclude that active inflammatory demyelination, neuronal loss and local complement synthesis and activation in the thalamus, are important to the pathological and clinical disease outcomes of PMS.
Collapse
Affiliation(s)
- Benjamin J Cooze
- Faculty of Medical, Health and Life Sciences, Swansea University, Swansea, UK
| | - Matthew Dickerson
- Faculty of Medical, Health and Life Sciences, Swansea University, Swansea, UK
| | | | - Lewis M Watkins
- Faculty of Medical, Health and Life Sciences, Swansea University, Swansea, UK
| | - Ethan Grounds
- Faculty of Medical, Health and Life Sciences, Swansea University, Swansea, UK
| | - Ben R Pearson
- Faculty of Medical, Health and Life Sciences, Swansea University, Swansea, UK
| | - Ryan Jack Bevan
- UK Dementia Research Institute at Cardiff University, Cardiff, UK
| | - B Paul Morgan
- UK Dementia Research Institute at Cardiff University, Cardiff, UK
| | - Roberta Magliozzi
- Department of Neurological and Movement Sciences, University of Verona, Italy
| | | | - James W Neal
- Faculty of Medical, Health and Life Sciences, Swansea University, Swansea, UK
| | - Owain W Howell
- Faculty of Medical, Health and Life Sciences, Swansea University, Swansea, UK
| |
Collapse
|
35
|
Age-related changes in multiple sclerosis and experimental autoimmune encephalomyelitis. Semin Immunol 2022; 59:101631. [PMID: 35752572 DOI: 10.1016/j.smim.2022.101631] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 06/03/2022] [Accepted: 06/13/2022] [Indexed: 01/15/2023]
Abstract
A better understanding of the pathological mechanisms that drive neurodegeneration in people living with multiple sclerosis (MS) is needed to design effective therapies to treat and/or prevent disease progression. We propose that CNS-intrinsic inflammation and re-modelling of the sub-arachnoid space of the leptomeninges sets the stage for neurodegeneration from the earliest stages of MS. While neurodegenerative processes are clinically silent early in disease, ageing results in neurodegenerative changes that become clinically manifest as progressive disability. Here we review pathological correlates of MS disease progression, highlight emerging mouse models that mimic key progressive changes in MS, and provide new perspectives on therapeutic approaches to protect against MS-associated neurodegeneration.
Collapse
|
36
|
Everest E, Ülgen E, Uygunoglu U, Tutuncu M, Saip S, Sezerman OU, Siva A, Tahir Turanli E. Investigation of multiple sclerosis-related pathways through the integration of genomic and proteomic data. PeerJ 2021; 9:e11922. [PMID: 39544199 PMCID: PMC11563213 DOI: 10.7717/peerj.11922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 07/16/2021] [Indexed: 11/17/2024] Open
Abstract
Background Multiple sclerosis (MS) has a complex pathophysiology, variable clinical presentation, and unpredictable prognosis; understanding the underlying mechanisms requires combinatorial approaches that warrant the integration of diverse molecular omics data. Methods Here, we combined genomic and proteomic data of the same individuals among a Turkish MS patient group to search for biologically important networks. We previously identified differentially-expressed proteins by cerebrospinal fluid proteome analysis of 179 MS patients and 42 non-MS controls. Among this study group, 11 unrelated MS patients and 60 independent, healthy controls were subjected to whole-genome SNP genotyping, and genome-wide associations were assessed. Pathway enrichment analyses of MS-associated SNPs and differentially-expressed proteins were conducted using the functional enrichment tool, PANOGA. Results Nine shared pathways were detected between the genomic and proteomic datasets after merging and clustering the enriched pathways. Complement and coagulation cascade was the most significantly associated pathway (hsa04610, P = 6.96 × 10-30). Other pathways involved in neurological or immunological mechanisms included adherens junctions (hsa04520, P = 6.64 × 10-25), pathogenic Escherichia coli infection (hsa05130, P = 9.03 × 10-14), prion diseases (hsa05020, P = 5.13 × 10-13). Conclusion We conclude that integrating multiple datasets of the same patients helps reducing false negative and positive results of genome-wide SNP associations and highlights the most prominent cellular players among the complex pathophysiological mechanisms.
Collapse
Affiliation(s)
- Elif Everest
- Department of Molecular Biology and Genetics, Faculty of Science and Letters, Istanbul Technical University, Istanbul, Turkey
| | - Ege Ülgen
- Department of Biostatistics and Medical Informatics, Faculty of Medicine, Acıbadem University, Istanbul, Turkey
| | - Ugur Uygunoglu
- Department of Neurology, Cerrahpaşa School of Medicine, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Melih Tutuncu
- Department of Neurology, Cerrahpaşa School of Medicine, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Sabahattin Saip
- Department of Neurology, Cerrahpaşa School of Medicine, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Osman Uğur Sezerman
- Department of Biostatistics and Medical Informatics, Faculty of Medicine, Acıbadem University, Istanbul, Turkey
| | - Aksel Siva
- Department of Neurology, Cerrahpaşa School of Medicine, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Eda Tahir Turanli
- Department of Molecular Biology and Genetics, Faculty of Science and Letters, Acıbadem University, Istanbul, Turkey
| |
Collapse
|
37
|
Petersen MA, Tognatta R, Meyer-Franke A, Bushong EA, Mendiola AS, Yan Z, Muthusamy A, Merlini M, Meza-Acevedo R, Cabriga B, Zhou Y, Thomas R, Ryu JK, Lassmann H, Ellisman MH, Akassoglou K. BMP receptor blockade overcomes extrinsic inhibition of remyelination and restores neurovascular homeostasis. Brain 2021; 144:2291-2301. [PMID: 34426831 PMCID: PMC8418337 DOI: 10.1093/brain/awab106] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 03/01/2021] [Accepted: 03/02/2021] [Indexed: 01/24/2023] Open
Abstract
Extrinsic inhibitors at sites of blood–brain barrier disruption and neurovascular damage contribute to remyelination failure in neurological diseases. However, therapies to overcome the extrinsic inhibition of remyelination are not widely available and the dynamics of glial progenitor niche remodelling at sites of neurovascular dysfunction are largely unknown. By integrating in vivo two-photon imaging co-registered with electron microscopy and transcriptomics in chronic neuroinflammatory lesions, we found that oligodendrocyte precursor cells clustered perivascularly at sites of limited remyelination with deposition of fibrinogen, a blood coagulation factor abundantly deposited in multiple sclerosis lesions. By developing a screen (OPC-X-screen) to identify compounds that promote remyelination in the presence of extrinsic inhibitors, we showed that known promyelinating drugs did not rescue the extrinsic inhibition of remyelination by fibrinogen. In contrast, bone morphogenetic protein type I receptor blockade rescued the inhibitory fibrinogen effects and restored a promyelinating progenitor niche by promoting myelinating oligodendrocytes, while suppressing astrocyte cell fate, with potent therapeutic effects in chronic models of multiple sclerosis. Thus, abortive oligodendrocyte precursor cell differentiation by fibrinogen is refractory to known promyelinating compounds, suggesting that blockade of the bone morphogenetic protein signalling pathway may enhance remyelinating efficacy by overcoming extrinsic inhibition in neuroinflammatory lesions with vascular damage.
Collapse
Affiliation(s)
- Mark A Petersen
- Gladstone UCSF Center for Neurovascular Brain Immunology, San Francisco, CA 94158, USA.,Gladstone Institutes, University of California, San Francisco, CA 94158, USA.,Department of Pediatrics, University of California, San Francisco, CA 94143, USA
| | - Reshmi Tognatta
- Gladstone UCSF Center for Neurovascular Brain Immunology, San Francisco, CA 94158, USA.,Gladstone Institutes, University of California, San Francisco, CA 94158, USA
| | - Anke Meyer-Franke
- Gladstone UCSF Center for Neurovascular Brain Immunology, San Francisco, CA 94158, USA.,Gladstone Institutes, University of California, San Francisco, CA 94158, USA
| | - Eric A Bushong
- National Center for Microscopy and Imaging Research, Center for Research on Biological Systems, University of California, San Diego, La Jolla, CA 92093, USA
| | - Andrew S Mendiola
- Gladstone UCSF Center for Neurovascular Brain Immunology, San Francisco, CA 94158, USA.,Gladstone Institutes, University of California, San Francisco, CA 94158, USA
| | - Zhaoqi Yan
- Gladstone UCSF Center for Neurovascular Brain Immunology, San Francisco, CA 94158, USA.,Gladstone Institutes, University of California, San Francisco, CA 94158, USA
| | - Abinaya Muthusamy
- Gladstone Institutes, University of California, San Francisco, CA 94158, USA
| | - Mario Merlini
- Gladstone Institutes, University of California, San Francisco, CA 94158, USA
| | - Rosa Meza-Acevedo
- Gladstone UCSF Center for Neurovascular Brain Immunology, San Francisco, CA 94158, USA.,Gladstone Institutes, University of California, San Francisco, CA 94158, USA
| | - Belinda Cabriga
- Gladstone UCSF Center for Neurovascular Brain Immunology, San Francisco, CA 94158, USA.,Gladstone Institutes, University of California, San Francisco, CA 94158, USA
| | - Yungui Zhou
- Gladstone UCSF Center for Neurovascular Brain Immunology, San Francisco, CA 94158, USA.,Gladstone Institutes, University of California, San Francisco, CA 94158, USA
| | - Reuben Thomas
- Gladstone Institutes, University of California, San Francisco, CA 94158, USA
| | - Jae Kyu Ryu
- Gladstone UCSF Center for Neurovascular Brain Immunology, San Francisco, CA 94158, USA.,Gladstone Institutes, University of California, San Francisco, CA 94158, USA.,Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA
| | - Hans Lassmann
- Center for Brain Research, Medical University of Vienna, A-1090 Vienna, Austria
| | - Mark H Ellisman
- National Center for Microscopy and Imaging Research, Center for Research on Biological Systems, University of California, San Diego, La Jolla, CA 92093, USA.,Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Katerina Akassoglou
- Gladstone UCSF Center for Neurovascular Brain Immunology, San Francisco, CA 94158, USA.,Gladstone Institutes, University of California, San Francisco, CA 94158, USA.,Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA
| |
Collapse
|
38
|
Kimura K, Lin Y, Yamaguchi H, Sato W, Takewaki D, Minote M, Doi Y, Okamoto T, Takahashi R, Kondo T, Yamamura T. Th1 - CD11c + B Cell Axis Associated with Response to Plasmapheresis in Multiple Sclerosis. Ann Neurol 2021; 90:595-611. [PMID: 34424567 PMCID: PMC9293420 DOI: 10.1002/ana.26202] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 07/21/2021] [Accepted: 08/15/2021] [Indexed: 12/29/2022]
Abstract
Objective Although plasmapheresis is a treatment option for patients with autoimmune neurological diseases, treatment response varies greatly among patients. The main objective of this study was to find out if biological/immune traits correlate with a beneficial response. Methods We thoroughly analyzed immune phenotypes in paired blood samples from a cohort of 31 patients with multiple sclerosis before and after plasmapheresis, in parallel with clinical evaluation of treatment response. Results The frequency of IFN‐γ+ Th1 cells was persistently higher in those who obtained benefit from plasmapheresis (responders) than nonresponders. The Th1 cell frequency before plasmapheresis provided a high predictive value for beneficial response, achieving area under the curve (AUC) of 0.902. Plasmapheresis treatment decreased inflammation‐related gene expressions in Th1 cells. Meanwhile, IFNG expression in Th1 cells positively correlated with the frequency of CD11c+ B cells, of which a pathogenic role has been suggested in several autoimmune diseases. In line with this, in vitro experiments showed that CD11c+ B cells would increase in response to exogenous IFN‐γ compared to IL‐4, and secrete high amounts of IgG. B cell receptor analysis indicated that clonal expansion of CD11c+ B cells takes place in patients with multiple sclerosis. Interestingly, CD11c+ B cells, which showed unique gene expression profile, decreased after plasmapheresis treatment along with all the immunoglobulin subsets in the circulation. Interpretation Taken together, we postulate that Th1 cell ‐ CD11c+ B cell axis is involved in treatment response to plasmapheresis, giving us clues to better understanding of complicated pathogenesis of autoimmune diseases, and getting closer to a personalized therapy. ANN NEUROL 2021;90:595–611
Collapse
Affiliation(s)
- Kimitoshi Kimura
- Department of Immunology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan.,Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Multiple Sclerosis Center, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Youwei Lin
- Department of Immunology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan.,Multiple Sclerosis Center, National Center of Neurology and Psychiatry, Tokyo, Japan.,Department of Neurology, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Hiromi Yamaguchi
- Department of Immunology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Wakiro Sato
- Department of Immunology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan.,Multiple Sclerosis Center, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Daiki Takewaki
- Department of Immunology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan.,Multiple Sclerosis Center, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Misako Minote
- Department of Immunology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan.,Multiple Sclerosis Center, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Yoshimitsu Doi
- Department of Immunology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan.,Multiple Sclerosis Center, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Tomoko Okamoto
- Multiple Sclerosis Center, National Center of Neurology and Psychiatry, Tokyo, Japan.,Department of Neurology, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Ryosuke Takahashi
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takayuki Kondo
- Department of Neurology, Kansai Medical University Medical Center, Osaka, Japan
| | - Takashi Yamamura
- Department of Immunology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan.,Multiple Sclerosis Center, National Center of Neurology and Psychiatry, Tokyo, Japan
| |
Collapse
|
39
|
Abbadessa G, Lavorgna L, Treaba CA, Bonavita S, Mainero C. Hemostatic factors in the pathogenesis of neuroinflammation in multiple sclerosis. Mult Scler 2021; 28:1834-1842. [PMID: 34410198 DOI: 10.1177/13524585211039111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND A growing body of evidence has shed light on the role of the hemostatic pathway and its components in the pathogenesis of multiple sclerosis (MS), particularly in enhancing and sustaining neuroinflammation. OBJECTIVE To review the clinical, experimental, and neuroimaging evidence supporting the role of different components of the hemostatic pathway in the pathogenesis of neuroinflammation in MS and discuss their translational potential as disease biomarkers and therapeutic targets. METHODS A literature search for most relevant articles from 1956 to 2020 was conducted in PubMed and Scopus. RESULTS Hemostasis components appear to be involved in different key events of neuroinflammation in MS including mononuclear cell diapedesis, microglia activation, and neuronal damage. CONCLUSION The findings on the interplay between hemostatic and thrombotic molecular pathways in the pathogenesis of neuroinflammation in MS open new opportunities for developing novel biomarkers for disease monitoring and prognosis, as well as novel therapeutic targets.
Collapse
Affiliation(s)
- Gianmarco Abbadessa
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Luigi Lavorgna
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Constantina Andrada Treaba
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA/Harvard Medical School, MA, USA
| | - Simona Bonavita
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Caterina Mainero
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA/Harvard Medical School, MA, USA
| |
Collapse
|
40
|
Bajrami A, Magliozzi R, Pisani AI, Pizzini FB, Crescenzo F, Marastoni D, Calabrese M. Volume changes of thalamus, hippocampus and cerebellum are associated with specific CSF profile in MS. Mult Scler 2021; 28:550-560. [PMID: 34378437 DOI: 10.1177/13524585211031786] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND The underlying pathogenesis of surface-in grey matter abnormalities in MS, demonstrated by both neuropathology and advanced MRI analyses, is under investigation and it might be related to CSF-mediated mechanism of inflammation and/or damage. OBJECTIVE To examine the link of CSF inflammatory profile with the damage of three regions early-involved in MS and bordering with CSF: thalamus, hippocampus and cerebellum. METHODS In this longitudinal, prospective study, we evaluated, in 109 relapsing-remitting MS patients, at diagnosis and after 2-year follow-up, the association between the baseline CSF level of 19 inflammatory mediators and the volume changes of thalamus, hippocampus, cerebellar cortex and control regions (globus pallidus, putamen). RESULTS The multivariable analysis showed that the CXCL13 and sCD163 CSF levels at baseline were independent predictors of thalamus (Rmodel2=0.80; p < 0.001) and hippocampus (Rmodel2=0.47; p < 0.001) volume change after 2-year follow-up. These molecules, plus CCL25, IFN-γ and fibrinogen, were independent predictors of the cerebellar cortex volume loss (Rmodel2=0.60; p < 0.001). No independent predictors of volume changes of the control regions were found. CONCLUSION Our results indicate an association between the CSF inflammatory profile and grey matter volume loss of regions anatomically close to CSF boundaries, thus supporting the hypothesis of a surface-in GM damage in MS.
Collapse
Affiliation(s)
- Albulena Bajrami
- Multiple Sclerosis Specialist Center, Neurology B, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Roberta Magliozzi
- Multiple Sclerosis Specialist Center, Neurology B, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Anna I Pisani
- Multiple Sclerosis Specialist Center, Neurology B, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Francesca B Pizzini
- Department of Diagnostic and Pathology, Integrated University Hospital of Verona, Neuroradiology & Radiology Units, Verona, Italy
| | - Francesco Crescenzo
- Multiple Sclerosis Specialist Center, Neurology B, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy/Neurology Unit, Mater Salutis Hospital, Legnago, Verona, Italy
| | - Damiano Marastoni
- Multiple Sclerosis Specialist Center, Neurology B, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Massimiliano Calabrese
- Multiple Sclerosis Specialist Center, Neurology B, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| |
Collapse
|
41
|
Magliozzi R, Pezzini F, Pucci M, Rossi S, Facchiano F, Marastoni D, Montagnana M, Lippi G, Reynolds R, Calabrese M. Changes in Cerebrospinal Fluid Balance of TNF and TNF Receptors in Naïve Multiple Sclerosis Patients: Early Involvement in Compartmentalised Intrathecal Inflammation. Cells 2021; 10:cells10071712. [PMID: 34359880 PMCID: PMC8303813 DOI: 10.3390/cells10071712] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 06/28/2021] [Accepted: 06/30/2021] [Indexed: 02/07/2023] Open
Abstract
An imbalance of TNF signalling in the inflammatory milieu generated by meningeal immune cell infiltrates in the subarachnoid space in multiple sclerosis (MS), and its animal model may lead to increased cortical pathology. In order to explore whether this feature may be present from the early stages of MS and may be associated with the clinical outcome, the protein levels of TNF, sTNF-R1 and sTNF-R2 were assayed in CSF collected from 122 treatment-naïve MS patients and 36 subjects with other neurological conditions at diagnosis. Potential correlations with other CSF cytokines/chemokines and with clinical and imaging parameters at diagnosis (T0) and after 2 years of follow-up (T24) were evaluated. Significantly increased levels of TNF (fold change: 7.739; p < 0.001), sTNF-R1 (fold change: 1.693; p < 0.001) and sTNF-R2 (fold change: 2.189; p < 0.001) were detected in CSF of MS patients compared to the control group at T0. Increased TNF levels in CSF were significantly (p < 0.01) associated with increased EDSS change (r = 0.43), relapses (r = 0.48) and the appearance of white matter lesions (r = 0.49). CSF levels of TNFR1 were associated with cortical lesion volume (r = 0.41) at T0, as well as with new cortical lesions (r = 0.56), whilst no correlation could be found between TNFR2 levels in CSF and clinical or MRI features. Combined correlation and pathway analysis (ingenuity) of the CSF protein pattern associated with TNF expression (encompassing elevated levels of BAFF, IFN-γ, IL-1β, IL-10, IL-8, IL-16, CCL21, haptoglobin and fibrinogen) showed a particular relationship to the interaction between innate and adaptive immune response. The CSF sTNF-R1-associated pattern (encompassing high levels of CXCL13, TWEAK, LIGHT, IL-35, osteopontin, pentraxin-3, sCD163 and chitinase-3-L1) was mainly related to altered T cell and B cell signalling. Finally, the CSF TNFR2-associated pattern (encompassing high CSF levels of IFN-β, IFN-λ2, sIL-6Rα) was linked to Th cell differentiation and regulatory cytokine signalling. In conclusion, dysregulation of TNF and TNF-R1/2 pathways associates with specific clinical/MRI profiles and can be identified at a very early stage in MS patients, at the time of diagnosis, contributing to the prediction of the disease outcome.
Collapse
MESH Headings
- Adaptive Immunity
- Adult
- Antigens, CD/cerebrospinal fluid
- Antigens, CD/genetics
- Antigens, CD/immunology
- Antigens, Differentiation, Myelomonocytic/cerebrospinal fluid
- Antigens, Differentiation, Myelomonocytic/genetics
- Antigens, Differentiation, Myelomonocytic/immunology
- B-Lymphocytes/immunology
- B-Lymphocytes/pathology
- C-Reactive Protein/cerebrospinal fluid
- C-Reactive Protein/genetics
- C-Reactive Protein/immunology
- Case-Control Studies
- Cerebral Cortex/diagnostic imaging
- Cerebral Cortex/immunology
- Cerebral Cortex/pathology
- Chemokine CXCL13/cerebrospinal fluid
- Chemokine CXCL13/genetics
- Chemokine CXCL13/immunology
- Chitinase-3-Like Protein 1/cerebrospinal fluid
- Chitinase-3-Like Protein 1/genetics
- Chitinase-3-Like Protein 1/immunology
- Cytokine TWEAK/cerebrospinal fluid
- Cytokine TWEAK/genetics
- Cytokine TWEAK/immunology
- Early Diagnosis
- Female
- Gene Expression Regulation
- Humans
- Immunity, Innate
- Interleukins/cerebrospinal fluid
- Interleukins/genetics
- Interleukins/immunology
- Magnetic Resonance Imaging
- Male
- Meninges/diagnostic imaging
- Meninges/immunology
- Meninges/pathology
- Multiple Sclerosis/cerebrospinal fluid
- Multiple Sclerosis/diagnostic imaging
- Multiple Sclerosis/genetics
- Multiple Sclerosis/pathology
- Osteopontin/cerebrospinal fluid
- Osteopontin/genetics
- Osteopontin/immunology
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/immunology
- Receptors, Tumor Necrosis Factor, Type I/cerebrospinal fluid
- Receptors, Tumor Necrosis Factor, Type I/genetics
- Receptors, Tumor Necrosis Factor, Type I/immunology
- Receptors, Tumor Necrosis Factor, Type II/cerebrospinal fluid
- Receptors, Tumor Necrosis Factor, Type II/genetics
- Receptors, Tumor Necrosis Factor, Type II/immunology
- Serum Amyloid P-Component/cerebrospinal fluid
- Serum Amyloid P-Component/genetics
- Serum Amyloid P-Component/immunology
- Signal Transduction
- T-Lymphocytes/immunology
- T-Lymphocytes/pathology
- Tumor Necrosis Factor Ligand Superfamily Member 14/cerebrospinal fluid
- Tumor Necrosis Factor Ligand Superfamily Member 14/genetics
- Tumor Necrosis Factor Ligand Superfamily Member 14/immunology
- Tumor Necrosis Factor-alpha/cerebrospinal fluid
- Tumor Necrosis Factor-alpha/genetics
- Tumor Necrosis Factor-alpha/immunology
- White Matter/diagnostic imaging
- White Matter/immunology
- White Matter/pathology
Collapse
Affiliation(s)
- Roberta Magliozzi
- Department of Neuroscience, Biomedicine and Movement Science, University of Verona, 37134 Verona, Italy; (F.P.); (M.P.); (D.M.); (M.M.); (G.L.); (M.C.)
- Department of Brain Sciences, Department of Medicine, Imperial College London, London W12 0NN, UK;
- Correspondence:
| | - Francesco Pezzini
- Department of Neuroscience, Biomedicine and Movement Science, University of Verona, 37134 Verona, Italy; (F.P.); (M.P.); (D.M.); (M.M.); (G.L.); (M.C.)
| | - Mairi Pucci
- Department of Neuroscience, Biomedicine and Movement Science, University of Verona, 37134 Verona, Italy; (F.P.); (M.P.); (D.M.); (M.M.); (G.L.); (M.C.)
| | - Stefania Rossi
- Department of Oncology and Molecular Medicine, Higher Institute of Health Care, 00161 Rome, Italy; (S.R.); (F.F.)
| | - Francesco Facchiano
- Department of Oncology and Molecular Medicine, Higher Institute of Health Care, 00161 Rome, Italy; (S.R.); (F.F.)
| | - Damiano Marastoni
- Department of Neuroscience, Biomedicine and Movement Science, University of Verona, 37134 Verona, Italy; (F.P.); (M.P.); (D.M.); (M.M.); (G.L.); (M.C.)
| | - Martina Montagnana
- Department of Neuroscience, Biomedicine and Movement Science, University of Verona, 37134 Verona, Italy; (F.P.); (M.P.); (D.M.); (M.M.); (G.L.); (M.C.)
| | - Giuseppe Lippi
- Department of Neuroscience, Biomedicine and Movement Science, University of Verona, 37134 Verona, Italy; (F.P.); (M.P.); (D.M.); (M.M.); (G.L.); (M.C.)
| | - Richard Reynolds
- Department of Brain Sciences, Department of Medicine, Imperial College London, London W12 0NN, UK;
- Centre for Molecular Neuropathology, Lee Kong Chian School of Medicine, Singapore 308232, Singapore
| | - Massimiliano Calabrese
- Department of Neuroscience, Biomedicine and Movement Science, University of Verona, 37134 Verona, Italy; (F.P.); (M.P.); (D.M.); (M.M.); (G.L.); (M.C.)
| |
Collapse
|
42
|
Bianchi L, Sframeli M, Vantaggiato L, Vita GL, Ciranni A, Polito F, Oteri R, Gitto E, Di Giuseppe F, Angelucci S, Versaci A, Messina S, Vita G, Bini L, Aguennouz M. Nusinersen Modulates Proteomics Profiles of Cerebrospinal Fluid in Spinal Muscular Atrophy Type 1 Patients. Int J Mol Sci 2021; 22:ijms22094329. [PMID: 33919289 PMCID: PMC8122268 DOI: 10.3390/ijms22094329] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 04/19/2021] [Indexed: 02/06/2023] Open
Abstract
Spinal muscular atrophy (SMA) type 1 is a severe infantile autosomal-recessive neuromuscular disorder caused by a survival motor neuron 1 gene (SMN1) mutation and characterized by progressive muscle weakness. Without supportive care, SMA type 1 is rapidly fatal. The antisense oligonucleotide nusinersen has recently improved the natural course of this disease. Here, we investigated, with a functional proteomic approach, cerebrospinal fluid (CSF) protein profiles from SMA type 1 patients who underwent nusinersen administration to clarify the biochemical response to the treatment and to monitor disease progression based on therapy. Six months after starting treatment (12 mg/5 mL × four doses of loading regimen administered at days 0, 14, 28, and 63), we observed a generalized reversion trend of the CSF protein pattern from our patient cohort to that of control donors. Notably, a marked up-regulation of apolipoprotein A1 and apolipoprotein E and a consistent variation in transthyretin proteoform occurrence were detected. Since these multifunctional proteins are critically active in biomolecular processes aberrant in SMA, i.e., synaptogenesis and neurite growth, neuronal survival and plasticity, inflammation, and oxidative stress control, their nusinersen induced modulation may support SMN improved-expression effects. Hence, these lipoproteins and transthyretin could represent valuable biomarkers to assess patient responsiveness and disease progression.
Collapse
Affiliation(s)
- Laura Bianchi
- Functional Proteomics Laboratory, Department of Life Sciences, University of Siena, 53100 Siena, Italy; (L.B.); (L.V.); (L.B.)
| | - Maria Sframeli
- Nemo Sud Clinical Centre, 98125 Messina, Italy; (M.S.); (G.L.V.)
| | - Lorenza Vantaggiato
- Functional Proteomics Laboratory, Department of Life Sciences, University of Siena, 53100 Siena, Italy; (L.B.); (L.V.); (L.B.)
| | - Gian Luca Vita
- Nemo Sud Clinical Centre, 98125 Messina, Italy; (M.S.); (G.L.V.)
| | - Annamaria Ciranni
- Unit of Neurology and Neuromuscular Diseases, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (A.C.); (F.P.); (R.O.); (S.M.); (M.A.)
| | - Francesca Polito
- Unit of Neurology and Neuromuscular Diseases, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (A.C.); (F.P.); (R.O.); (S.M.); (M.A.)
| | - Rosaria Oteri
- Unit of Neurology and Neuromuscular Diseases, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (A.C.); (F.P.); (R.O.); (S.M.); (M.A.)
| | - Eloisa Gitto
- Neonatal and Paediatric Intensive Care Unit, Department of Human Pathology in Adult and Developmental Age, University of Messina, 98125 Messina, Italy;
| | - Fabrizio Di Giuseppe
- Dentistry and Biotechnology, and Proteomics Unit, Centre of Advanced Studies and Technoloy, Department Medical, Oral & Biotechnological Sciences, “G. d’Annunzio”, University of Chieti-Pescara, 66100 Chieti, Italy; (F.D.G.); (S.A.)
| | - Stefania Angelucci
- Dentistry and Biotechnology, and Proteomics Unit, Centre of Advanced Studies and Technoloy, Department Medical, Oral & Biotechnological Sciences, “G. d’Annunzio”, University of Chieti-Pescara, 66100 Chieti, Italy; (F.D.G.); (S.A.)
| | - Antonio Versaci
- Intensive Care Unit, AOU Policlinico “G. Martino”, 98125 Messina, Italy;
| | - Sonia Messina
- Unit of Neurology and Neuromuscular Diseases, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (A.C.); (F.P.); (R.O.); (S.M.); (M.A.)
| | - Giuseppe Vita
- Nemo Sud Clinical Centre, 98125 Messina, Italy; (M.S.); (G.L.V.)
- Unit of Neurology and Neuromuscular Diseases, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (A.C.); (F.P.); (R.O.); (S.M.); (M.A.)
- Correspondence:
| | - Luca Bini
- Functional Proteomics Laboratory, Department of Life Sciences, University of Siena, 53100 Siena, Italy; (L.B.); (L.V.); (L.B.)
| | - M’hammed Aguennouz
- Unit of Neurology and Neuromuscular Diseases, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (A.C.); (F.P.); (R.O.); (S.M.); (M.A.)
| |
Collapse
|
43
|
Li Z, Liu Y, Jia A, Cui Y, Feng J. Cerebrospinal fluid cells immune landscape in multiple sclerosis. J Transl Med 2021; 19:125. [PMID: 33766068 PMCID: PMC7995713 DOI: 10.1186/s12967-021-02804-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 03/19/2021] [Indexed: 02/06/2023] Open
Abstract
Background Multiple Sclerosis (MS) is a potentially devastating autoimmune neurological disorder, which characteristically induces demyelination of white matter in the brain and spinal cord. Methods In this study, three characteristics of the central nervous system (CNS) immune microenvironment occurring during MS onset were explored; immune cell proportion alteration, differential gene expression profile, and related pathways. The raw data of two independent datasets were obtained from the ArrayExpress database; E-MTAB-69, which was used as a derivation cohort, and E-MTAB-2374 which was used as a validation cohort. Differentially expressed genes (DEGs) were identified by the false discovery rate (FDR) value of < 0.05 and |log2 (Fold Change)|> 1, for further analysis. Then, functional enrichment analyses were performed to explore the pathways associated with MS onset. The gene expression profiles were analyzed using CIBERSORT to identify the immune type alterations involved in MS disease. Results After verification, the proportion of five types of immune cells (plasma cells, monocytes, macrophage M2, neutrophils and eosinophils) in cerebrospinal fluid (CSF) were revealed to be significantly altered in MS cases compared to the control group. Thus, the complement and coagulation cascades and the systemic lupus erythematosus (SLE) pathways may play critical roles in MS. We identified NLRP3, LILRB2, C1QB, CD86, C1QA, CSF1R, IL1B and TLR2 as eight core genes correlated with MS. Conclusions Our study identified the change in the CNS immune microenvironment of MS cases by analysis of the in silico data using CIBERSORT. Our data may assist in providing directions for further research as to the molecular mechanisms of MS and provide future potential therapeutic targets in treatment. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-021-02804-7.
Collapse
Affiliation(s)
- Zijian Li
- Department of Neurology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, Liaoning, 110004, China
| | - Yongchao Liu
- Department of Neurology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, Liaoning, 110004, China
| | - Aili Jia
- Department of Neurology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, Liaoning, 110004, China
| | - Yueran Cui
- Department of Neurology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, Liaoning, 110004, China
| | - Juan Feng
- Department of Neurology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, Liaoning, 110004, China.
| |
Collapse
|
44
|
Peoples N, Strang C. Complement Activation in the Central Nervous System: A Biophysical Model for Immune Dysregulation in the Disease State. Front Mol Neurosci 2021; 14:620090. [PMID: 33746710 PMCID: PMC7969890 DOI: 10.3389/fnmol.2021.620090] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 01/12/2021] [Indexed: 01/08/2023] Open
Abstract
Complement, a feature of the innate immune system that targets pathogens for phagocytic clearance and promotes inflammation, is tightly regulated to prevent damage to host tissue. This regulation is paramount in the central nervous system (CNS) since complement proteins degrade neuronal synapses during development, homeostasis, and neurodegeneration. We propose that dysregulated complement, particularly C1 or C3b, may errantly target synapses for immune-mediated clearance, therefore highlighting regulatory failure as a major potential mediator of neurological disease. First, we explore the mechanics of molecular neuroimmune relationships for the regulatory proteins: Complement Receptor 1, C1-Inhibitor, Factor H, and the CUB-sushi multiple domain family. We propose that biophysical and chemical principles offer clues for understanding mechanisms of dysregulation. Second, we describe anticipated effects to CNS disease processes (particularly Alzheimer's Disease) and nest our ideas within existing basic science, clinical, and epidemiological findings. Finally, we illustrate how the concepts presented within this manuscript provoke new ways of approaching age-old neurodegenerative processes. Every component of this model is testable by straightforward experimentation and highlights the untapped potential of complement dysregulation as a driver of CNS disease. This includes a putative role for complement-based neurotherapeutic agents and companion biomarkers.
Collapse
|
45
|
Elkjaer ML, Nawrocki A, Kacprowski T, Lassen P, Simonsen AH, Marignier R, Sejbaek T, Nielsen HH, Wermuth L, Rashid AY, Høgh P, Sellebjerg F, Reynolds R, Baumbach J, Larsen MR, Illes Z. CSF proteome in multiple sclerosis subtypes related to brain lesion transcriptomes. Sci Rep 2021; 11:4132. [PMID: 33603109 PMCID: PMC7892884 DOI: 10.1038/s41598-021-83591-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 02/02/2021] [Indexed: 02/08/2023] Open
Abstract
To identify markers in the CSF of multiple sclerosis (MS) subtypes, we used a two-step proteomic approach: (i) Discovery proteomics compared 169 pooled CSF from MS subtypes and inflammatory/degenerative CNS diseases (NMO spectrum and Alzheimer disease) and healthy controls. (ii) Next, 299 proteins selected by comprehensive statistics were quantified in 170 individual CSF samples. (iii) Genes of the identified proteins were also screened among transcripts in 73 MS brain lesions compared to 25 control brains. F-test based feature selection resulted in 8 proteins differentiating the MS subtypes, and secondary progressive (SP)MS was the most different also from controls. Genes of 7 out these 8 proteins were present in MS brain lesions: GOLM was significantly differentially expressed in active, chronic active, inactive and remyelinating lesions, FRZB in active and chronic active lesions, and SELENBP1 in inactive lesions. Volcano maps of normalized proteins in the different disease groups also indicated the highest amount of altered proteins in SPMS. Apolipoprotein C-I, apolipoprotein A-II, augurin, receptor-type tyrosine-protein phosphatase gamma, and trypsin-1 were upregulated in the CSF of MS subtypes compared to controls. This CSF profile and associated brain lesion spectrum highlight non-inflammatory mechanisms in differentiating CNS diseases and MS subtypes and the uniqueness of SPMS.
Collapse
Affiliation(s)
- Maria L Elkjaer
- Department of Neurology, Odense University Hospital, J.B. Winslowsvej 4, 5000, Odense C, Denmark.,Institute of Clinical Research, University of Southern Denmark, Odense, Denmark.,Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Arkadiusz Nawrocki
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Tim Kacprowski
- Research Group Computational Systems Medicine, Chair of Experimental Bioinformatics, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Munich, Germany.,Division Data Science in Biomedicine, Peter L. Reichertz Institute for Medical Informatics of TU Braunschweig and Medical School Hannover, Brunswick, Germany
| | - Pernille Lassen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Anja Hviid Simonsen
- Danish Dementia Research Centre, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Romain Marignier
- Service de Neurologie, Sclérose en Plaques, Lyon Neuroscience Research Center, Lyon, France
| | - Tobias Sejbaek
- Department of Neurology, Odense University Hospital, J.B. Winslowsvej 4, 5000, Odense C, Denmark.,Department of Neurology, Hospital South West Jutland, University Hospital of Southern Denmark, Esbjerg, Denmark
| | - Helle H Nielsen
- Department of Neurology, Odense University Hospital, J.B. Winslowsvej 4, 5000, Odense C, Denmark.,Institute of Clinical Research, University of Southern Denmark, Odense, Denmark.,Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Lene Wermuth
- Department of Neurology, Odense University Hospital, J.B. Winslowsvej 4, 5000, Odense C, Denmark.,Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Alyaa Yakut Rashid
- Department of Neurology, Hospital South West Jutland, University Hospital of Southern Denmark, Esbjerg, Denmark
| | - Peter Høgh
- Regional Dementia Research Centre, Department of Neurology, Zealand University Hospital, Roskilde, Denmark.,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Finn Sellebjerg
- Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark., Copenhagen, Denmark
| | | | - Jan Baumbach
- Department of Mathematics and Computer Science, University of Southern Denmark, Odense, Denmark.,Chair of Experimental Bioinformatics, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Munich, Germany
| | - Martin R Larsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Zsolt Illes
- Department of Neurology, Odense University Hospital, J.B. Winslowsvej 4, 5000, Odense C, Denmark. .,Institute of Clinical Research, University of Southern Denmark, Odense, Denmark. .,Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
46
|
Silva BA, Miglietta E, Ferrari CC. Insights into the role of B cells in the cortical pathology of Multiple sclerosis: evidence from animal models and patients. Mult Scler Relat Disord 2021; 50:102845. [PMID: 33636613 DOI: 10.1016/j.msard.2021.102845] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 02/03/2021] [Accepted: 02/13/2021] [Indexed: 01/02/2023]
Abstract
Multiple sclerosis (MS) is a chronic, immune-mediated disease of the central nervous system (CNS) that affects both white and gray matter. Although it has been traditionally considered as a T cell mediated disease, the role of B cell in MS pathology has become a topic of great research interest. Cortical lesions, key feature of the progressive forms of MS, are involved in cognitive impairment and worsening of the patients' outcome. These lesions present pathognomonic hallmarks, such as: absence of blood-brain barrier (BBB) disruption, limited inflammatory events, reactive microglia, neurodegeneration, demyelination and meningeal inflammation. B cells located in the meninges, either as part of diffuse inflammation or as part of follicle-like structures, are strongly associated with cortical damage. The function of CD20-expressing B cells in MS is further highlighted by the success of specific therapies using anti-CD20 antibodies. The possible roles of B cells in pathology go beyond their ability to produce antibodies, as they also present antigens to T cells, secrete cytokines (both pathogenic and protective) within the CNS to modulate T and myeloid cell functions, and are involved in meningeal inflammation. Here, we will review the contributions of B cells to the pathogenesis of meningeal inflammation and cortical lesions in MS patients as well as in preclinical animal models.
Collapse
Affiliation(s)
- Berenice Anabel Silva
- Instituto de Medicina Traslacional e Ingeniería Biomédica (IMTIB), CONICET, Buenos Aires, Argentina; Leloir Institute Foundation, Institute for Biochemical Investigations, IIBBA, CONICET, Buenos Aires, Argentina; Centro Universitario de Esclerosis Múltiple, División Neurología, Hospital JM Ramos Mejía, Facultad de Medicina, Universidad de Buenos Aires, Argentina
| | - Esteban Miglietta
- Leloir Institute Foundation, Institute for Biochemical Investigations, IIBBA, CONICET, Buenos Aires, Argentina
| | - Carina Cintia Ferrari
- Instituto de Medicina Traslacional e Ingeniería Biomédica (IMTIB), CONICET, Buenos Aires, Argentina; Leloir Institute Foundation, Institute for Biochemical Investigations, IIBBA, CONICET, Buenos Aires, Argentina.
| |
Collapse
|
47
|
Ramaglia V, Rojas O, Naouar I, Gommerman JL. The Ins and Outs of Central Nervous System Inflammation-Lessons Learned from Multiple Sclerosis. Annu Rev Immunol 2021; 39:199-226. [PMID: 33524273 DOI: 10.1146/annurev-immunol-093019-124155] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Multiple sclerosis (MS) is a chronic disease that is characterized by the inappropriate invasion of lymphocytes and monocytes into the central nervous system (CNS), where they orchestrate the demyelination of axons, leading to physical and cognitive disability. There are many reasons immunologists should be interested in MS. Aside from the fact that there is still significant unmet need for patients living with the progressive form of the disease, MS is a case study for how immune cells cross CNS barriers and subsequently interact with specialized tissue parenchymal cells. In this review, we describe the types of immune cells that infiltrate the CNS and then describe interactions between immune cells and glial cells in different types of lesions. Lastly, we provide evidence for CNS-compartmentalized immune cells and speculate on how this impacts disease progression for MS patients.
Collapse
Affiliation(s)
- Valeria Ramaglia
- Department of Immunology, University of Toronto, Ontario M5S 1A8, Canada;
| | - Olga Rojas
- Department of Immunology, University of Toronto, Ontario M5S 1A8, Canada;
| | - Ikbel Naouar
- Department of Immunology, University of Toronto, Ontario M5S 1A8, Canada;
| | | |
Collapse
|
48
|
Abstract
PURPOSE OF REVIEW In multiple sclerosis, currently approved disease-modifying treatments are effective in modulating peripheral immunity, and coherently, in reducing clinical/radiological relapses, but still, they perform poorly in preventing disease progression and overall disability accrual. This review provides an up-to-date overview of the neuropathology of progressive multiple sclerosis, including a summary of the main mechanisms of disease progression. RECENT FINDINGS Clinical progression in multiple sclerosis is likely related to the accumulation of neuro-axonal loss in a lifelong inflammatory CNS environment (both adaptive and innate) and relative un-balance between damage, repair and brain functional reserve. A critical driver appears to be the T-cell and B-cell-mediated compartmentalized inflammation within the leptomeninges and within the parenchyma. Recent perspective highlighted also the role of the glial response to such lifelong inflammatory injury as the critical player for both pathological and clinical outcomes. SUMMARY The neuropathological and biological understanding of disease progression in multiple sclerosis have progressed in the last few years. As a consequence, new therapeutic approaches are emerging outside the modulation of T-cell activity and/or the depletion of B cells.
Collapse
|
49
|
Morgan BP, Gommerman JL, Ramaglia V. An "Outside-In" and "Inside-Out" Consideration of Complement in the Multiple Sclerosis Brain: Lessons From Development and Neurodegenerative Diseases. Front Cell Neurosci 2021; 14:600656. [PMID: 33488361 PMCID: PMC7817777 DOI: 10.3389/fncel.2020.600656] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 11/24/2020] [Indexed: 12/14/2022] Open
Abstract
The last 15 years have seen an explosion of new findings on the role of complement, a major arm of the immune system, in the central nervous system (CNS) compartment including contributions to cell migration, elimination of synapse during development, aberrant synapse pruning in neurologic disorders, damage to nerve cells in autoimmune diseases, and traumatic injury. Activation of the complement system in multiple sclerosis (MS) is typically thought to occur as part of a primary (auto)immune response from the periphery (the outside) against CNS antigens (the inside). However, evidence of local complement production from CNS-resident cells, intracellular complement functions, and the more recently discovered role of early complement components in shaping synaptic circuits in the absence of inflammation opens up the possibility that complement-related sequelae may start and finish within the brain itself. In this review, the complement system will be introduced, followed by evidence that implicates complement in shaping the developing, adult, and normal aging CNS as well as its contribution to pathology in neurodegenerative conditions. Discussion of data supporting "outside-in" vs. "inside-out" roles of complement in MS will be presented, concluded by thoughts on potential approaches to therapies targeting specific elements of the complement system.
Collapse
Affiliation(s)
- B. Paul Morgan
- UK Dementia Research Institute at Cardiff, Cardiff University, Cardiff, United Kingdom
| | | | - Valeria Ramaglia
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
50
|
Ramaglia V, Florescu A, Zuo M, Sheikh-Mohamed S, Gommerman JL. Stromal Cell–Mediated Coordination of Immune Cell Recruitment, Retention, and Function in Brain-Adjacent Regions. THE JOURNAL OF IMMUNOLOGY 2021; 206:282-291. [DOI: 10.4049/jimmunol.2000833] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 10/27/2020] [Indexed: 12/15/2022]
|