1
|
SoRelle ED, Luftig MA. Multiple sclerosis and infection: history, EBV, and the search for mechanism. Microbiol Mol Biol Rev 2025; 89:e0011923. [PMID: 39817754 PMCID: PMC11948499 DOI: 10.1128/mmbr.00119-23] [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] [Indexed: 01/18/2025] Open
Abstract
SUMMARYInfection has long been hypothesized as the cause of multiple sclerosis (MS), and recent evidence for Epstein-Barr virus (EBV) as the trigger of MS is clear and compelling. This clarity contrasts with yet uncertain viral mechanisms and their relation to MS neuroinflammation and demyelination. As long as this disparity persists, it will invigorate virologists, molecular biologists, immunologists, and clinicians to ascertain how EBV potentiates MS onset, and possibly the disease's chronic activity and progression. Such efforts should take advantage of the diverse body of basic and clinical research conducted over nearly two centuries since the first clinical descriptions of MS plaques. Defining the contribution of EBV to the complex and multifactorial pathology of MS will also require suitable experimental models and techniques. Such efforts will broaden our understanding of virus-driven neuroinflammation and specifically inform the development of EBV-targeted therapies for MS management and, ultimately, prevention.
Collapse
Affiliation(s)
- Elliott D. SoRelle
- Department of Molecular Genetics & Microbiology, Center for Virology, Duke University, Durham, North Carolina, USA
| | - Micah A. Luftig
- Department of Molecular Genetics & Microbiology, Center for Virology, Duke University, Durham, North Carolina, USA
| |
Collapse
|
2
|
Ansari A, Thibault PA, Salapa HE, Clarke JPWE, Levin MC. Mutations in hnRNP A1 drive neurodegeneration and alternative RNA splicing of neuronal gene targets. Neurobiol Dis 2025; 206:106814. [PMID: 39874994 DOI: 10.1016/j.nbd.2025.106814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 01/30/2025] Open
Abstract
RNA binding protein dysfunction is a pathogenic feature of multiple neurological diseases, including multiple sclerosis (MS). Neurodegeneration (the loss of, or damage to neurons and axons) is the primary driver of disease progression in MS. Herein, we utilized a novel, neuron-specific model of neurodegeneration by transducing primary mouse neurons with mutant forms of the RNA binding protein heterogeneous nuclear ribonucleoprotein A1 (hnRNP A1) identified from MS patients, including one within the M9-nuclear localization sequence of hnRNP A1 (A1(P275S)) and a second in the prion-like domain of hnRNP A1 (A1(F263S)) to test the hypothesis that neuronal hnRNP A1 dysfunction drives neurodegeneration in MS. Examination of hnRNP A1 localization in neurons revealed an increase in nucleocytoplasmic mislocalization in neurons transduced with A1(P275S), but not A1(F263S). Yet, both A1(F263S) and A1(P275S) induced neurodegeneration evidenced by significant reductions in total neurite length and complexity and an increase in FluoroJade-C neuronal cell body staining. RNA sequencing and differential alternative splicing analysis of mutant-expressing neurons revealed dramatic changes in alternative RNA splicing of transcripts critical to neuronal function. Further, amyloid precursor protein (APP), a marker for neurodegeneration in MS, showed differential splicing in mutant-expressing neurons, which was confirmed in MS brains with hnRNP A1 dysfunction. Overall, we have identified that hnRNP A1 plays a complex role in neuronal function and regulation by mediating the alternative splicing of neuron-specific transcripts. When neuronal hnRNP A1 function is impaired, as in disease, resultant dysfunction propagates through multiple pathways that may influence the progression of neurodegeneration in MS.
Collapse
Affiliation(s)
- Ansalna Ansari
- Office of the Saskatchewan Multiple Sclerosis Clinical Research Chair, University of Saskatchewan, Saskatoon, SK S7K 0M7, Canada; Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Patricia A Thibault
- Office of the Saskatchewan Multiple Sclerosis Clinical Research Chair, University of Saskatchewan, Saskatoon, SK S7K 0M7, Canada; Cameco MS Neuroscience Research Centre, College of Medicine, University of Saskatchewan, Saskatoon, SK S7K 0M7, Canada; Neurology Division, Department of Medicine, University of Saskatchewan, Saskatoon, SK S7N 0X8, Canada
| | - Hannah E Salapa
- Office of the Saskatchewan Multiple Sclerosis Clinical Research Chair, University of Saskatchewan, Saskatoon, SK S7K 0M7, Canada; Cameco MS Neuroscience Research Centre, College of Medicine, University of Saskatchewan, Saskatoon, SK S7K 0M7, Canada; Neurology Division, Department of Medicine, University of Saskatchewan, Saskatoon, SK S7N 0X8, Canada
| | - Joseph-Patrick W E Clarke
- Office of the Saskatchewan Multiple Sclerosis Clinical Research Chair, University of Saskatchewan, Saskatoon, SK S7K 0M7, Canada; Cameco MS Neuroscience Research Centre, College of Medicine, University of Saskatchewan, Saskatoon, SK S7K 0M7, Canada; Neurology Division, Department of Medicine, University of Saskatchewan, Saskatoon, SK S7N 0X8, Canada
| | - Michael C Levin
- Office of the Saskatchewan Multiple Sclerosis Clinical Research Chair, University of Saskatchewan, Saskatoon, SK S7K 0M7, Canada; Neurology Division, Department of Medicine, University of Saskatchewan, Saskatoon, SK S7N 0X8, Canada.
| |
Collapse
|
3
|
Rubino V, Cammarota M, Criscuolo C, Cianflone A, De Martino M, de Rosa V, Esposito F, Abbadessa G, Carriero F, Terrazzano G, Chieffi P, Bonavita S, Bresciamorra V, Annunziato L, Ruggiero G, Boscia F. Modulation of NCX1 expression in monocytes associates with multiple sclerosis progression. Heliyon 2025; 11:e42332. [PMID: 40041001 PMCID: PMC11876900 DOI: 10.1016/j.heliyon.2025.e42332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 01/16/2025] [Accepted: 01/28/2025] [Indexed: 03/06/2025] Open
Abstract
Ionic imbalance and functional heterogeneity of monocytes play key roles in multiple sclerosis (MS) progression. A better understanding of monocyte response in the context of ionic dysregulation during MS course may have relevant implications for understanding of disease pathogenesis and treatments. The sodium calcium exchanger NCX1 influences monocyte-derived macrophages reactivity under inflammation; however, little is known about its monocyte-specific expression during MS course. By means of RT-PCR, flow cytometry, and confocal analyses, we determined the expression profiling of NCX1 exchanger in monocytes of patients during relapsing-remitting MS (RRMS) and secondary progressive MS (SPMS) course. NCX1 expression was significantly upregulated in monocytes from transitional RRMS subjects. Conversely, it was significantly reduced in all monocyte subsets after RRMS conversion to SPMS. Interestingly, NCX1 levels in monocytes significantly correlated with the percentage and growth ability of the regulatory T cell (Treg) subset, whose derangement underlies MS progression. Perturbation of transcripts encoding the Ca2+-ATPase isoform 1 and 4, the Na+/K+-ATPase α1 subunit, and the long non-coding RNA SLC8A1-AS1 associated with NCX1 changes in peripheral blood mononuclear cells (PBMC) during MS. Our findings demonstrated a stage-specific dysregulation of NCX1 exchanger in monocytes during MS progression and suggested that ionic imbalance in monocytes may influence not only their functional response but also the immune regulatory network during MS course. These data may be relevant for the identification of novel biomarkers and/or therapeutic targets in MS.
Collapse
Affiliation(s)
- Valentina Rubino
- Department of Medical Translational Sciences, School of Medicine, “Federico II” University of Naples, 80131, Naples, Italy
| | - Mariarosaria Cammarota
- Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, “Federico II” University of Naples, 80131, Naples, Italy
| | - Chiara Criscuolo
- Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, “Federico II” University of Naples, 80131, Naples, Italy
- CDCD Neurology, “Federico II” University Hospital, Naples, Italy
| | - Alessandra Cianflone
- Clinical and Translational Research Unit, Santobono-Pausilipon Children's Hospital, 80129, Naples, Italy
| | - Marco De Martino
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138, Naples, Italy
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Naples, Italy
| | - Valeria de Rosa
- Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, “Federico II” University of Naples, 80131, Naples, Italy
| | - Francesco Esposito
- Institute of Experimental Endocrinology and Oncology (IEOS) “G. Salvatore”, National Research Council (CNR), 80131, Naples, Italy
| | - Gianmarco Abbadessa
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138, Naples, Italy
| | - Flavia Carriero
- Department of Health Science, University of Basilicata, 85100, Potenza, Italy
| | - Giuseppe Terrazzano
- Department of Health Science, University of Basilicata, 85100, Potenza, Italy
| | - Paolo Chieffi
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138, Naples, Italy
| | - Simona Bonavita
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138, Naples, Italy
| | - Vincenzo Bresciamorra
- Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, “Federico II” University of Naples, 80131, Naples, Italy
- Multiple Sclerosis Unit, Policlinico “Federico II” University Hospital, 80131, Naples, Naples, Italy
| | | | - Giuseppina Ruggiero
- Department of Medical Translational Sciences, School of Medicine, “Federico II” University of Naples, 80131, Naples, Italy
| | - Francesca Boscia
- Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, “Federico II” University of Naples, 80131, Naples, Italy
| |
Collapse
|
4
|
Goodin DS. The epidemiology, pathology and pathogenesis of MS: Therapeutic implications. Neurotherapeutics 2025:e00539. [PMID: 40021419 DOI: 10.1016/j.neurot.2025.e00539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 12/30/2024] [Accepted: 01/22/2025] [Indexed: 03/03/2025] Open
Abstract
Multiple sclerosis (MS) is a chronic, and potentially disabling, inflammatory disease of the central nervous system (CNS). MS is generally characterized by recurrent, and self-limited, episodes of neurological dysfunction, which occur unpredictably and often result in multifocal tissue injury within the CNS. Currently, women are affected two to three times as often as men although this may not have been the case during earlier Time-Periods. The pathogenesis of MS is known to involve both critical genetic and environmental mechanisms. Nevertheless, in addition to these two mechanisms, disease-pathogenesis also involves a "truly" random event. Indeed, it is this random mechanism, which is responsible for the currently-observed (and increasing) excess of women among patients with MS. This review summarizes the current state of knowledge regarding the pathogenesis of MS (includong its epidemiology, pathology, and genetics) and considers the therapeutic implications that these pathogenetic mechanisms have both for our currently available therapies as well as for the possible therapeutic approaches to the management of this potentially disabling condition in the future.
Collapse
Affiliation(s)
- Douglas S Goodin
- University of California, San Francisco and the San Francisco VA Medical Center, 675 Nelson Rising Lane, Suite #221D, San Francisco, CA 94158, USA.
| |
Collapse
|
5
|
Fang H, He J, Du D, Wang X, Xu X, Lu L, Zhou Y, Wen Y, He F, Li Y, Wen H, Zhou M. Deciphering the secret codes in N 7-methylguanosine modification: Context-dependent function of methyltransferase-like 1 in human diseases. Clin Transl Med 2025; 15:e70240. [PMID: 39979979 PMCID: PMC11842222 DOI: 10.1002/ctm2.70240] [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: 09/25/2024] [Revised: 02/05/2025] [Accepted: 02/12/2025] [Indexed: 02/22/2025] Open
Abstract
N7-methylguanosine (m7G) is one of the most prevalent post-transcriptional modifications of RNA and plays a critical role in RNA translation and stability. As a pivotal m7G regulator, methyltransferase-like 1 (METTL1) is responsible for methyl group transfer during the progression of m7G modification and contributes to the structure and functional regulation of RNA. Accumulating evidence in recent years has revealed that METTL1 plays key roles in various diseases depending on its m7G RNA methyltransferase activity. Elevated levels of METTL1 are typically associated with disease development and adverse consequences. In contrast, METTL1 may act as a disease suppressor in several disorders. While the roles of m7G modifications in disease have been extensively reviewed, the critical functions of METTL1 in various types of disease and the potential targeting of METTL1 for disease treatment have not yet been highlighted. This review describes the various biological functions of METTL1, summarises recent advances in understanding its pathogenic and disease-suppressive functions and discusses the underlying molecular mechanisms. Given that METTL1 can promote or inhibit disease processes, the possibility of applying METTL1 inhibitors and agonists is further discussed, with the goal of providing novel insights for future disease diagnosis and potential intervention targets. KEY POINTS: METTL1-mediated m7G modification is crucial for various biological processes, including RNA stability, maturation and translation. METTL1 has emerged as a critical epigenetic modulator in human illnesses, with its dysregulated expression correlating with multiple diseases progression and presenting opportunities for both diagnostic biomarker development and molecular-targeted therapy. Enormous knowledge gaps persist regarding context-dependent regulatory networks of METTL1 and dynamic m7G modification patterns, necessitating mechanistic interrogation to bridge basic research with clinical translation in precision medicine.
Collapse
Affiliation(s)
- Huan Fang
- Department of GastroenterologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Jing He
- Department of Breast SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Dan Du
- Department of Medical LaboratoryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Xue Wang
- Department of Medical LaboratoryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Xinyu Xu
- Department of Medical LaboratoryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Linping Lu
- Department of GastroenterologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Yefan Zhou
- Department of Medical LaboratoryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Yangyang Wen
- Department of GastroenterologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Fucheng He
- Department of Medical LaboratoryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Yingxia Li
- Department of GastroenterologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Hongtao Wen
- Department of GastroenterologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Mingxia Zhou
- Department of GastroenterologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| |
Collapse
|
6
|
Caranova M, Soares JF, Pereira DJ, Lima AC, Sousa L, Batista S, Castelo-Branco M, Duarte JV. Longitudinal Identification of Pre-Lesional Tissue in Multiple Sclerosis With Advanced Diffusion MRI. J Neuroimaging 2025; 35:e70022. [PMID: 39937068 DOI: 10.1111/jon.70022] [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: 12/11/2024] [Revised: 01/24/2025] [Accepted: 01/27/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND AND PURPOSE Structural MRI (sMRI) is used in monitoring multiple sclerosis (MS) but lacks sensitivity in detecting clinically relevant damage to normal-appearing white matter (NAWM), that is, pre-lesional tissue, and specificity for identifying the underlying substrate of injury. In this longitudinal study, we identified pre-lesional tissue in MS patients and investigated its microstructure by modeling diffusion-weighted imaging (DWI) data using diffusion tensor imaging and neurite orientation dispersion and density imaging (NODDI). METHODS We enrolled 18 patients with relapsing-remitting MS (10 females, 31.92 ± 8.09 years, disease duration 0.91 ± 1.81 years) and 18 healthy controls (10 females, 31.89 ± 8.15 years). Participants underwent two sMRI and DWI sessions (baseline and follow-up) with the same protocols. Average apparent diffusion coefficient (ADC), fractional anisotropy (FA), orientation dispersion index (ODI), and neurite density index (NDI) were estimated in data-driven regions of interest: nonpersistent lesional tissue (lesional tissue at baseline, resolved at follow-up), lesions that only existed at follow-up (pre-lesional tissue at baseline, lesions at follow-up), persistent lesional tissue (lesions at baseline and follow-up), and NAWM. RESULTS Compared to NAWM, pre-lesional tissue showed lower ODI, and resolved lesional tissue showed higher FA and ADC and lower ODI and NDI. Over time, persistent lesional tissue showed a decrease in FA and ODI and an increase in NDI. Compared to nonpersistent lesional tissue, persistent lesional tissue showed higher ADC and lower NDI. CONCLUSIONS DWI and, more particularly, NODDI, can reveal the unique microstructure of persistent, resolved, and pre-lesional tissue in MS.
Collapse
Affiliation(s)
- Maria Caranova
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Institute for Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal
| | - Júlia F Soares
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Institute for Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal
| | - Daniela Jardim Pereira
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Institute for Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal
- Neuroradiology Functional Unit, Imaging Service, Coimbra Local Health Unit, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Ana Cláudia Lima
- Neurology Department, Coimbra Hospital and University Centre, Coimbra, Portugal
| | - Lívia Sousa
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Neurology Department, Coimbra Hospital and University Centre, Coimbra, Portugal
| | - Sónia Batista
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Neurology Department, Coimbra Hospital and University Centre, Coimbra, Portugal
| | - Miguel Castelo-Branco
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Institute for Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - João V Duarte
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Institute for Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal
- Champalimaud Research, Champalimaud Foundation, Lisbon, Portugal
| |
Collapse
|
7
|
de Mol CL, Lamballais S, Muetzel R, Duijts L, Smolders J, White T, Neuteboom RF. Environmental multiple sclerosis (MS) risk factors, genetic MS risk, and brain development in a general paediatric population. J Neurol Neurosurg Psychiatry 2024:jnnp-2024-335053. [PMID: 39658135 DOI: 10.1136/jnnp-2024-335053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 11/03/2024] [Indexed: 12/12/2024]
Abstract
BACKGROUND Neuroaxonal loss occurs in the early stages of multiple sclerosis (MS), but whether it results from early inflammatory brain damage or an ongoing neurodegenerative process remains unclear. We hypothesise that genetic and childhood environmental risk factors for MS may already have an impact on neurodevelopment before the typical age of onset for MS in the general population. METHODS We examined associations and interactions of genetic and environmental risk factors for MS with brain MRI outcomes, including volumetric (n=5350) and diffusion data (n=5649), at ages 9 and 13 years in a large, population-based childhood cohort without MS diagnoses. Polygenic risk scores (PRSs) were used to assess genetic burden, with rs10191329 as a marker of MS severity. Environmental factors at age 5 included Epstein-Barr virus (EBV) serology, vitamin D status, body mass index, duration of outdoor activities, and household parental smoking. RESULTS Genetic data were available for 2817 and 2970 participants with volumetric and diffusion data, respectively. The MS-PRS was positively associated with EBV viral capsid antigen titres among EBV-positive children (β=0.15, p=2.98×10-6). A negative association was observed between the MS-PRS and subcortical grey matter volume (β=-0.03, p=0.014). Interaction between the MS-PRS and household parental smoking was negatively linked to total brain (β=-0.21, p=0.025) and thalamic volumes (β=-0.22, p=0.003), where a higher MS-PRS and household smoking were associated with lower volumes. No associations were observed for rs10191329 with brain outcomes. CONCLUSIONS Genetic and environmental risk factors for MS interact to influence brain volumes in childhood, suggesting a potential window for preventing MS in genetically susceptible individuals by reducing exposure to household smoking.
Collapse
Affiliation(s)
- Casper Louk de Mol
- Neurology, MS Center ErasMS, Erasmus Medical Center, Rotterdam, Zuid-Holland, Netherlands
| | - Sander Lamballais
- Clinical Genetics, Erasmus Medical Center, Rotterdam, Zuid-Holland, Netherlands
| | - Ryan Muetzel
- Child and Adolescent Psychiatry, Erasmus Medical Center - Sophia Children's Hospital, Rotterdam, Zuid-Holland, Netherlands
| | - Liesbeth Duijts
- Pediatrics, Erasmus Medical Center, Rotterdam, Zuid-Holland, Netherlands
| | - Joost Smolders
- Neurology, MS Center ErasMS, Erasmus Medical Center, Rotterdam, Zuid-Holland, Netherlands
- Neuroimmunology Researchgroup, Netherlands Institute for Neuroscience, Amsterdam, Noord-Holland, Netherlands
| | - Tonya White
- Section on Social and Cognitive Developmental Neuroscience, NIH, Bethesda, Maryland, USA
| | | |
Collapse
|
8
|
Shiri H, Javan M. Sox2-mediated transdifferentiation of hAT-MSCs into induced neural progenitor-like cells for remyelination therapies. Tissue Cell 2024; 91:102553. [PMID: 39255744 DOI: 10.1016/j.tice.2024.102553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/03/2024] [Accepted: 09/03/2024] [Indexed: 09/12/2024]
Abstract
Mesenchymal stem cells (MSCs) are converted to neural cells using growth factors and chemicals. Although these neural cells are effective at modulating disease symptoms, they are less effective at replacing lost neural cells. Direct transdifferentiation seems to be a promising method for generating the required cells for regenerative medicine applications. Sox2 is a key transcription factor in neural progenitor (NP) fate determination and has been frequently used for transdifferentiating different cell types to NPs. Here, we demonstrated that the overexpression of a single transcription factor, Sox2, in human adipose tissue-derived mesenchymal stem cells (hAT-MSCs) led to the generation of induced NPs-like cells that were clonogenic, proliferative and passageable, and showed the potential to differentiate into three neural lineages. NPs are known as progenitors with the potential to differentiate into oligodendrocytes. In vivo, following transplantation into demyelinated adult mouse brains, they survived, differentiated and integrated into the adult brain while participating in the remyelination process and behavioral improvement. This report introduces a beneficial, low-cost and effective approach for generating NPs from an accessible adult source for autologous applications in treating neurodegenerative diseases, including remyelination therapies for multiple sclerosis and other demyelinating diseases.
Collapse
Affiliation(s)
- Hamed Shiri
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Javan
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; Institute for Brain and Cognition, Tarbiat Modares University, Tehran, Iran; Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
9
|
Clarelli F, Corona A, Pääkkönen K, Sorosina M, Zollo A, Piehl F, Olsson T, Stridh P, Jagodic M, Hemmer B, Gasperi C, Harroud A, Shchetynsky K, Mingione A, Mascia E, Misra K, Giordano A, Mazzieri MLT, Priori A, Saarela J, Kockum I, Filippi M, Esposito F, Boneschi FGM. Pharmacogenomics of clinical response to Natalizumab in multiple sclerosis: a genome-wide multi-centric association study. J Neurol 2024; 271:7250-7263. [PMID: 39264442 PMCID: PMC11561017 DOI: 10.1007/s00415-024-12608-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 09/13/2024]
Abstract
BACKGROUND Inter-individual differences in treatment response are marked in multiple sclerosis (MS). This is true for Natalizumab (NTZ), to which a subset of patients displays sub-optimal treatment response. We conducted a multi-centric genome-wide association study (GWAS), with additional pathway and network analysis to identify genetic predictors of response to NTZ. METHODS MS patients from three different centers were included. Response to NTZ was dichotomized, nominating responders (R) relapse-free patients and non-responders (NR) all the others, over a follow-up of 4 years. Association analysis on ~ 4.7 M imputed autosomal common single-nucleotide polymorphisms (SNPs) was performed fitting logistic regression models, adjusted for baseline covariates, followed by meta-analysis at SNP and gene level. Finally, these signals were projected onto STRING interactome, to elicit modules and hub genes linked to response. RESULTS Overall, 1834 patients were included: 119 from Italy (R = 94, NR = 25), 81 from Germany (R = 61, NR = 20), and 1634 from Sweden (R = 1349, NR = 285). The top-associated variant was rs11132400T (p = 1.33 × 10-6, OR = 0.58), affecting expression of several genes in the locus, like KLKB1. The interactome analysis implicated a module of 135 genes, with over-representation of terms like canonical WNT signaling pathway (padjust = 7.08 × 10-6). Response-associated genes like GRB2 and LRP6, already implicated in MS pathogenesis, were topologically prioritized within the module. CONCLUSION This GWAS, the largest pharmacogenomic study of response to NTZ, suggested MS-implicated genes and Wnt/β-catenin signaling pathway, an essential component for blood-brain barrier formation and maintenance, to be related to treatment response.
Collapse
Affiliation(s)
- Ferdinando Clarelli
- Laboratory of Human Genetics of Neurological Disorders, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, Milan, Italy
| | - Andrea Corona
- Laboratory of Precision Medicine of Neurological Diseases, Department of Health Science, University of Milan, Milan, Italy
- CRC "Aldo Ravelli" for Experimental Brain Therapeutics, Department of Health Science, University of Milan, Milan, Italy
| | - Kimmo Pääkkönen
- Institute for Molecular Medicine Finland (FIMM), University of FI Helsinki, Helsinki, Finland
| | - Melissa Sorosina
- Laboratory of Human Genetics of Neurological Disorders, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, Milan, Italy
| | - Alen Zollo
- Laboratory of Precision Medicine of Neurological Diseases, Department of Health Science, University of Milan, Milan, Italy
- CRC "Aldo Ravelli" for Experimental Brain Therapeutics, Department of Health Science, University of Milan, Milan, Italy
| | - Fredrik Piehl
- The Karolinska Neuroimmunology & Multiple Sclerosis Centre, Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Visionsgatan 18, 171 76, Stockholm, Sweden
| | - Tomas Olsson
- The Karolinska Neuroimmunology & Multiple Sclerosis Centre, Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Visionsgatan 18, 171 76, Stockholm, Sweden
| | - Pernilla Stridh
- The Karolinska Neuroimmunology & Multiple Sclerosis Centre, Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Visionsgatan 18, 171 76, Stockholm, Sweden
| | - Maja Jagodic
- The Karolinska Neuroimmunology & Multiple Sclerosis Centre, Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Visionsgatan 18, 171 76, Stockholm, Sweden
| | - Bernhard Hemmer
- Department of Neurology, School of Medicine, Technical University of Munich, Klinikum Rechts Der Isar, Ismaninger Str. 22, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Christiane Gasperi
- Department of Neurology, School of Medicine, Technical University of Munich, Klinikum Rechts Der Isar, Ismaninger Str. 22, Munich, Germany
| | - Adil Harroud
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
| | - Klementy Shchetynsky
- The Karolinska Neuroimmunology & Multiple Sclerosis Centre, Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Visionsgatan 18, 171 76, Stockholm, Sweden
| | - Alessandra Mingione
- Laboratory of Precision Medicine of Neurological Diseases, Department of Health Science, University of Milan, Milan, Italy
- CRC "Aldo Ravelli" for Experimental Brain Therapeutics, Department of Health Science, University of Milan, Milan, Italy
| | - Elisabetta Mascia
- Laboratory of Human Genetics of Neurological Disorders, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, Milan, Italy
| | - Kaalindi Misra
- Laboratory of Human Genetics of Neurological Disorders, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, Milan, Italy
| | - Antonino Giordano
- Laboratory of Human Genetics of Neurological Disorders, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Maria Laura Terzi Mazzieri
- Laboratory of Precision Medicine of Neurological Diseases, Department of Health Science, University of Milan, Milan, Italy
- CRC "Aldo Ravelli" for Experimental Brain Therapeutics, Department of Health Science, University of Milan, Milan, Italy
| | - Alberto Priori
- CRC "Aldo Ravelli" for Experimental Brain Therapeutics, Department of Health Science, University of Milan, Milan, Italy
- Clinical Neurology Unit, Azienda Socio-Sanitaria Territoriale Santi Paolo E Carlo and Department of Health Sciences, University of Milan, Milan, Italy
| | - Janna Saarela
- Institute for Molecular Medicine Finland (FIMM), University of FI Helsinki, Helsinki, Finland
| | - Ingrid Kockum
- The Karolinska Neuroimmunology & Multiple Sclerosis Centre, Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Visionsgatan 18, 171 76, Stockholm, Sweden
| | - Massimo Filippi
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
- Neurorehabilitation Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 48, Milan, Italy
- Neurophysiology Service, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, Milan, Italy
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, Milan, Italy
- Vita-Salute San Raffaele University, Via Olgettina, 60, Milan, Italy
| | - Federica Esposito
- Laboratory of Human Genetics of Neurological Disorders, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, Milan, Italy.
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy.
| | - Filippo Giovanni Martinelli Boneschi
- Laboratory of Precision Medicine of Neurological Diseases, Department of Health Science, University of Milan, Milan, Italy.
- CRC "Aldo Ravelli" for Experimental Brain Therapeutics, Department of Health Science, University of Milan, Milan, Italy.
- Clinical Neurology Unit, Azienda Socio-Sanitaria Territoriale Santi Paolo E Carlo and Department of Health Sciences, University of Milan, Milan, Italy.
| |
Collapse
|
10
|
Geloso MC, Zupo L, Corvino V. Crosstalk between peripheral inflammation and brain: Focus on the responses of microglia and astrocytes to peripheral challenge. Neurochem Int 2024; 180:105872. [PMID: 39362496 DOI: 10.1016/j.neuint.2024.105872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/18/2024] [Accepted: 09/30/2024] [Indexed: 10/05/2024]
Abstract
A growing body of evidence supports the link between peripheral inflammation and impairment of neurologic functions, including mood and cognitive abilities. The pathogenic event connecting peripheral inflammation and brain dysfunction is represented by neuroinflammation, a pathogenic phenomenon that provides an important contribution to neurodegeneration and cognitive decline also in Alzheimer's, Parkinson's, Huntington's diseases, as well as in Multiple Sclerosis. It is driven by resident brain immune cells, microglia and astrocytes, that acquire an activated phenotype in response to proinflammatory molecules moving from the periphery to the brain parenchyma. Although a huge progress has been made in clarifying cellular and molecular mechanisms bridging peripheral and central inflammation, a clear picture has not been achieved so far. Therefore, experimental models are of crucial relevance to clarify knowledge gaps in this regard. Many findings demonstrate that systemic inflammation induced by pathogen-associated molecular patterns, such as lipopolysaccharide (LPS), is able to trigger neuroinflammation. Therefore, LPS-administration is widely considered a useful tool to study this phenomenon. On this basis, the present review will focus on in vivo studies based on acute and subacute effects of systemic administration of LPS, with special attention on the state of art of microglia and astrocyte response to peripheral challenge.
Collapse
Affiliation(s)
- Maria Concetta Geloso
- Department of Neuroscience, Section of Human Anatomy, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy; Gemelli Science and Technology Park (GSTeP)-Organoids Research Core Facility, Fondazione Policlinico Agostino Gemelli IRCCS, Rome, Italy.
| | - Luca Zupo
- Department of Neuroscience, Section of Human Anatomy, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy
| | - Valentina Corvino
- Department of Neuroscience, Section of Human Anatomy, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy
| |
Collapse
|
11
|
Bilek F, Ercan Z, Deniz G, Ozgul S, Demir CF. High-intensity intermittent exercise increases serum levels of chitinase 3-like protein-1 and matrix metalloproteinase-9 in persons with multiple sclerosis. J Neuroimmunol 2024; 395:578434. [PMID: 39178495 DOI: 10.1016/j.jneuroim.2024.578434] [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: 03/27/2024] [Revised: 08/07/2024] [Accepted: 08/18/2024] [Indexed: 08/26/2024]
Abstract
The study aimed to evaluate the effect of high-intensity intermittent exercise (HIIE) on serum levels of MMP-9 and CHI3L1 in multiple sclerosis. Study group received HIIE twice a week for 12 weeks, while control group received no treatment. In intra-group comparison, study group showed a significant increase in MMP-9 and CHI3L1 levels, while control group showed no significant difference. In intergroup comparison, a significant difference was found only in CHI3L1 levels after treatment. The increase in MMP-9 and CH3L-1 concentrations in study group suggests that these biomarkers may play a role in regulating specific skeletal muscle adaptations due to HIIE.
Collapse
Affiliation(s)
- Furkan Bilek
- Muğla Sıtkı Koçman University, Fethiye Faculty of Health Sciences, Department of Gerontology, Muğla, Türkiye.
| | - Zubeyde Ercan
- Fırat University, Faculty of Health Sciences, Department of Physiotherapy and Rehabilitation, Elazığ, Türkiye.
| | - Gulnihal Deniz
- Erzurum Technical University, Faculty of Health Sciences, Department of Physiotherapy and Rehabilitation, Erzurum, Türkiye.
| | - Sinem Ozgul
- Fırat University Hospital, Physiotherapy and Rehabilitation Clinic, Elazığ, Türkiye
| | - Caner Feyzi Demir
- Fırat University, School of Medicine, Department of Neurology, Elazığ, Türkiye.
| |
Collapse
|
12
|
Smoot K, Gervasi-Follmar T, Marginean H, Chen C, Cohan S. Impact of oral melatonin supplementation on urine and serum melatonin concentrations and quality-of-life measures in persons with relapsing multiple sclerosis. Mult Scler Relat Disord 2024; 90:105799. [PMID: 39126937 DOI: 10.1016/j.msard.2024.105799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/05/2024] [Accepted: 08/02/2024] [Indexed: 08/12/2024]
Abstract
INTRODUCTION Melatonin is an antioxidant and anti-inflammatory agent that modulates the immune system by scavenging free radicals, reducing the upregulation of pro-inflammatory cytokines, and reducing transendothelial cell migration. Therefore, melatonin may play a role in regulating multiple sclerosis (MS) disease activity. However, little is known about how melatonin supplementation effects individuals with MS. OBJECTIVE Determine if there was a dose-dependent elevation in urine and serum melatonin concentrations. Determine if melatonin supplementation had an impact on patient reported outcomes. METHODS This was a randomized, dose-blinded exploratory study. Adults (age 18-65) with relapsing forms of multiple sclerosis (RMS) treated with a stable dose of oral disease modifying therapy for at least 6 months were randomized into melatonin 3 mg or 5 mg daily. Urinary and serum melatonin levels and modified fatigue impact scale (MFIS), multiple sclerosis impact scale (MSIS-29), and Pittsburgh sleep quality index (PSQI), patient determined disease steps (PDDS) and performance scales (PS) were measured at baseline, 3, 6, and 12 months. Urinary and serum melatonin analyses was performed to estimate mean concentrations and their differences between treatment arms over time by a repeated measures linear mixed model. The model included treatment, assessment time, and treatment × time interaction. RESULTS Thirty patients, randomized 1:1, were analyzed in an intent to treat population. Twenty-three completed the study. The repeated measures linear mixed model analysis of all timepoints revealed higher melatonin concentrations in patients on 5 mg compared to 3 mg melatonin for both urinary 6-SMT (p = 0.03) and serum melatonin (p = 0.04). MFIS, MSIS-29, PSQI, and PDSS-PS scores did not significantly change from baseline to month 12. No significant differences in these measures were seen between the two doses. Five patients stopped melatonin (three on 5 mg and two on 3 mg) due to adverse events, including one patient who developed focal spongiotic dermatitis. One patient experienced three consecutive serious adverse events that were unrelated to melatonin supplementation. CONCLUSIONS The 5 mg melatonin supplementation group had higher concentrations of urinary 6-SMT and serum melatonin compared to the 3 mg group over 12 months of treatment. There was a correlation between 6-SMT and serum melatonin concentrations. This suggests that measuring serum melatonin is a reliable alternative to measuring urinary 6-SMT. However, no differences in clinical benefit between the two dosage groups were demonstrated in the patient reported outcomes. TRIAL REGISTRATION NUMBER NCT03498131.
Collapse
Affiliation(s)
- Kyle Smoot
- Providence Brain and Spine Institute, Providence Saint Vincent Medical Center, Portland, OR, United States.
| | - Tiffany Gervasi-Follmar
- Providence Brain and Spine Institute, Providence Saint Vincent Medical Center, Portland, OR, United States
| | - Horia Marginean
- Providence Brain and Spine Institute, Providence Saint Vincent Medical Center, Portland, OR, United States
| | - Chiayi Chen
- Providence Brain and Spine Institute, Providence Saint Vincent Medical Center, Portland, OR, United States
| | - Stanley Cohan
- Providence Brain and Spine Institute, Providence Saint Vincent Medical Center, Portland, OR, United States
| |
Collapse
|
13
|
Neili NE, AbdelKafi-Koubaa Z, Jebali J, Kaidi K, Sahraoui G, Ahmed MB, Srairi-Abid N, Marrakchi N, Doghri R, ELBini I. Modulation of αv integrins by lebecetin, a viper venom-derived molecule, in experimental neuroinflammation and demyelination models. Sci Rep 2024; 14:22398. [PMID: 39333683 PMCID: PMC11436777 DOI: 10.1038/s41598-024-73259-1] [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: 05/06/2024] [Accepted: 09/16/2024] [Indexed: 09/29/2024] Open
Abstract
Several neurodegenerative diseases, such as multiple sclerosis and Parkinson's disease, are linked to alterations in myelin content or structure. Transmembrane receptors such as integrins could be involved in these alterations. In the present study, we investigated the role of αv-integrins in experimental models of neuroinflammation and demyelination with the use of lebecetin (LCT), a C-lectin protein purified from Macrovipera lebetina viper venom, as an αv-integrin modulator. In a model of neuroinflammation, LCT inhibited the upregulation of αv, β3, β5, α5, and β1 integrins, as well as the associated release of pro-inflammatory factor IL-6 and chemokine CXCL-10, and decreased the expression of phosphorylated NfκB. The subsequent "indirect culture" between reactive astrocytes and oligodendrocytes showed a down-regulation of αv and β3 integrins versus upregulation of β1 one, accompanied by a reduced expression of myelin basic protein (MBP). Treatment of oligodendrocytes with LCT rectified the changes in integrin and MBP expression. Through Western blot quantification, LCT was shown to upregulate the expression levels of PI3K and p-mTOR while downregulating expression levels of p-AKT in oligodendrocytes, suggesting the neuroprotective and pro-myelinating effects of LCT may be related to the PI3K/mTor/AKT pathway. Concomitantly, we found that LCT promoted remyelination by tracking the increased expression of MBP in the brains of cuprizone-intoxicated mice. These results point to an involvement of integrins in not only neuroinflammation but demyelination as well. Thus, targeting αv integrins could offer potential therapeutic avenues for the treatment of demyelinating diseases.
Collapse
Affiliation(s)
- Nour-Elhouda Neili
- Laboratory of Biomolecules, Venoms and Theranostic Applications (LR20IPT01), Pasteur Institute of Tunis, University of Tunis, El Manar, Tunis, Tunisia
| | - Zaineb AbdelKafi-Koubaa
- Laboratory of Biomolecules, Venoms and Theranostic Applications (LR20IPT01), Pasteur Institute of Tunis, University of Tunis, El Manar, Tunis, Tunisia
- Research Laboratory of Precision Medicine/Personalized Medicine and Oncology Investigation (LR21SP01), Saleh Azaiez Institute, Tunis, Tunisia
| | - Jed Jebali
- Laboratory of Biomolecules, Venoms and Theranostic Applications (LR20IPT01), Pasteur Institute of Tunis, University of Tunis, El Manar, Tunis, Tunisia
| | - Khouloud Kaidi
- Laboratory of Biomolecules, Venoms and Theranostic Applications (LR20IPT01), Pasteur Institute of Tunis, University of Tunis, El Manar, Tunis, Tunisia
| | - Ghada Sahraoui
- Research Laboratory of Precision Medicine/Personalized Medicine and Oncology Investigation (LR21SP01), Saleh Azaiez Institute, Tunis, Tunisia
- Faculty of Medicine of Tunis, University of Tunis, El Manar, Tunis, Tunisia
| | - Melika Ben Ahmed
- Faculty of Medicine of Tunis, University of Tunis, El Manar, Tunis, Tunisia
- Laboratory of Transmission, Control and Immunobiology of Infections (LR16IPT02), Pasteur Institute of Tunis, University of Tunis, El Manar, Tunis, Tunisia
| | - Najet Srairi-Abid
- Laboratory of Biomolecules, Venoms and Theranostic Applications (LR20IPT01), Pasteur Institute of Tunis, University of Tunis, El Manar, Tunis, Tunisia
| | - Naziha Marrakchi
- Laboratory of Biomolecules, Venoms and Theranostic Applications (LR20IPT01), Pasteur Institute of Tunis, University of Tunis, El Manar, Tunis, Tunisia
| | - Raoudha Doghri
- Research Laboratory of Precision Medicine/Personalized Medicine and Oncology Investigation (LR21SP01), Saleh Azaiez Institute, Tunis, Tunisia
- Faculty of Medicine of Tunis, University of Tunis, El Manar, Tunis, Tunisia
| | - Ines ELBini
- Laboratory of Biomolecules, Venoms and Theranostic Applications (LR20IPT01), Pasteur Institute of Tunis, University of Tunis, El Manar, Tunis, Tunisia.
| |
Collapse
|
14
|
Singh V, Zheng Y, Ontaneda D, Mahajan KR, Holloman J, Fox RJ, Nakamura K, Trapp BD. Disability independent of cerebral white matter demyelination in progressive multiple sclerosis. Acta Neuropathol 2024; 148:34. [PMID: 39217272 PMCID: PMC11365858 DOI: 10.1007/s00401-024-02796-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/20/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
The pathogenic mechanisms contributing to neurological disability in progressive multiple sclerosis (PMS) are poorly understood. Cortical neuronal loss independent of cerebral white matter (WM) demyelination in myelocortical MS (MCMS) and identification of MS patients with widespread cortical atrophy and disability progression independent of relapse activity (PIRA) support pathogenic mechanisms other than cerebral WM demyelination. The three-dimensional distribution and underlying pathology of myelinated T2 lesions were investigated in postmortem MCMS brains. Postmortem brain slices from previously characterized MCMS (10 cases) and typical MS (TMS) cases (12 cases) were co-registered with in situ postmortem T2 hyperintensities and T1 hypointensities. T1 intensity thresholds were used to establish a classifier that differentiates MCMS from TMS. The classifier was validated in 36 uncharacterized postmortem brains and applied to baseline MRIs from 255 living PMS participants enrolled in SPRINT-MS. Myelinated T2 hyperintensities in postmortem MCMS brains have a contiguous periventricular distribution that expands at the occipital poles of the lateral ventricles where a surface-in gradient of myelinated axonal degeneration was observed. The MRI classifier distinguished pathologically confirmed postmortem MCMS and TMS cases with an accuracy of 94%. For SPRINT-MS patients, the MRI classifier identified 78% as TMS, 10% as MCMS, and 12% with a paucity of cerebral T1 and T2 intensities. In SPRINT-MS, expanded disability status scale and brain atrophy measures were similar in MCMS and TMS cohorts. A paucity of cerebral WM demyelination in 22% of living PMS patients raises questions regarding a primary role for cerebral WM demyelination in disability progression in all MS patients and has implications for clinical management of MS patients and clinical trial outcomes in PMS. Periventricular myelinated fiber degeneration provides additional support for surface-in gradients of neurodegeneration in MS.
Collapse
Affiliation(s)
- Vikas Singh
- Department of Neurosciences, NC30, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
| | - Yufan Zheng
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Daniel Ontaneda
- Mellen Center for Treatment and Research in MS, Cleveland Clinic, Cleveland, OH, USA
| | - Kedar R Mahajan
- Department of Neurosciences, NC30, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
- Mellen Center for Treatment and Research in MS, Cleveland Clinic, Cleveland, OH, USA
| | - Jameson Holloman
- Department of Neurosciences, NC30, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
- Mellen Center for Treatment and Research in MS, Cleveland Clinic, Cleveland, OH, USA
| | - Robert J Fox
- Mellen Center for Treatment and Research in MS, Cleveland Clinic, Cleveland, OH, USA
| | - Kunio Nakamura
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Bruce D Trapp
- Department of Neurosciences, NC30, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA.
| |
Collapse
|
15
|
Swain HN, Boyce PD, Bromet BA, Barozinksy K, Hance L, Shields D, Olbricht GR, Semon JA. Mesenchymal stem cells in autoimmune disease: A systematic review and meta-analysis of pre-clinical studies. Biochimie 2024; 223:54-73. [PMID: 38657832 DOI: 10.1016/j.biochi.2024.04.009] [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: 01/30/2024] [Revised: 04/08/2024] [Accepted: 04/20/2024] [Indexed: 04/26/2024]
Abstract
Mesenchymal Stem Cells (MSCs) are of interest in the clinic because of their immunomodulation capabilities, capacity to act upstream of inflammation, and ability to sense metabolic environments. In standard physiologic conditions, they play a role in maintaining the homeostasis of tissues and organs; however, there is evidence that they can contribute to some autoimmune diseases. Gaining a deeper understanding of the factors that transition MSCs from their physiological function to a pathological role in their native environment, and elucidating mechanisms that reduce their therapeutic relevance in regenerative medicine, is essential. We conducted a Systematic Review and Meta-Analysis of human MSCs in preclinical studies of autoimmune disease, evaluating 60 studies that included 845 patient samples and 571 control samples. MSCs from any tissue source were included, and the study was limited to four autoimmune diseases: multiple sclerosis, rheumatoid arthritis, systemic sclerosis, and lupus. We developed a novel Risk of Bias tool to determine study quality for in vitro studies. Using the International Society for Cell & Gene Therapy's criteria to define an MSC, most studies reported no difference in morphology, adhesion, cell surface markers, or differentiation into bone, fat, or cartilage when comparing control and autoimmune MSCs. However, there were reported differences in proliferation. Additionally, 308 biomolecules were differentially expressed, and the abilities to migrate, invade, and form capillaries were decreased. The findings from this study could help to explain the pathogenic mechanisms of autoimmune disease and potentially lead to improved MSC-based therapeutic applications.
Collapse
Affiliation(s)
- Hailey N Swain
- Department of Biological Sciences, Missouri University of Science and Technology, USA
| | - Parker D Boyce
- Department of Biological Sciences, Missouri University of Science and Technology, USA
| | - Bradley A Bromet
- Department of Biological Sciences, Missouri University of Science and Technology, USA
| | - Kaiden Barozinksy
- Department of Biological Sciences, Missouri University of Science and Technology, USA
| | - Lacy Hance
- Department of Biological Sciences, Missouri University of Science and Technology, USA
| | - Dakota Shields
- Department of Mathematics and Statistics, Missouri University of Science and Technology, USA
| | - Gayla R Olbricht
- Department of Mathematics and Statistics, Missouri University of Science and Technology, USA
| | - Julie A Semon
- Department of Biological Sciences, Missouri University of Science and Technology, USA.
| |
Collapse
|
16
|
Bulondo F, Babensee JE. Optimization of Interleukin-10 incorporation for dendritic cells embedded in Poly(ethylene glycol) hydrogels. J Biomed Mater Res A 2024; 112:1317-1336. [PMID: 38562052 DOI: 10.1002/jbm.a.37714] [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: 09/18/2023] [Revised: 03/19/2024] [Accepted: 03/22/2024] [Indexed: 04/04/2024]
Abstract
Translational research in biomaterials and immunoengineering is leading to the development of novel advanced therapeutics to treat diseases such as cancer, autoimmunity, and viral infections. Dendritic cells (DCs) are at the center of these therapeutics given that they bridge innate and adaptive immunity. The biomaterial system developed herein uses a hydrogel carrier to deliver immunomodulatory DCs for amelioration of autoimmunity. This biomaterial vehicle is comprised of a poly (ethylene glycol)-4 arm maleimide (PEG-4MAL) hydrogels, conjugated with the immunosuppressive cytokine, interleukin-10, IL-10, and cross-linked with a collagenase-degradable peptide sequence for the injectable delivery of immunosuppressive DCs to an anatomical disease-relevant site of the cervical lymph nodes, for intended application to treat multiple sclerosis. The amount of IL-10 incorporated in the hydrogel was optimized to be 500 ng in vitro, based on immunological endpoints. At this concentration, DCs exhibited the best viability, most immunosuppressive phenotype, and protection against proinflammatory insult as compared with hydrogel-incorporated DCs with lower IL-10 loading amounts. Additionally, the effect of the degradability of the PEG-4MAL hydrogel on the release rate of incorporated IL-10 was assessed by varying the ratio of degradable peptides: VPM (degradable) and DTT (nondegradable) and measuring the IL-10 release rates. This IL-10-conjugated hydrogel delivery system for immunosuppressive DCs is set to be assessed for in vivo functionality as the immunosuppressive cytokine provides a tolerogenic environment that keeps DCs in their immature phenotype, which consequently enhances cell viability and optimizes the system's immunomodulatory functionality.
Collapse
Affiliation(s)
- Fredrick Bulondo
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA
- Department of Biomedical Sciences and Engineering, Mbarara University of Science and Technology, Mbarara, Uganda
| | - Julia E Babensee
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA
| |
Collapse
|
17
|
Ashrafpour S, Nasr-Taherabadi MJ, Sabouri-Rad A, Hosseinzadeh S, Pourabdolhossein F. Arbutin intervention ameliorates memory impairment in a rat model of lysolecethin induced demyelination: Neuroprotective and anti-inflammatory effects. Behav Brain Res 2024; 469:115041. [PMID: 38723674 DOI: 10.1016/j.bbr.2024.115041] [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: 03/06/2024] [Revised: 04/17/2024] [Accepted: 05/03/2024] [Indexed: 05/23/2024]
Abstract
Cognitive impairment (CI) and memory deficit are prevalent manifestations of multiple sclerosis (MS). This study explores the therapeutic potential of arbutin on memory deficits using a rat hippocampal demyelination model induced by lysophosphatidylcholine (LPC). Demyelination was induced by bilateral injection of 1% LPC into the CA1 area of the hippocampus, and the treated group received daily arbutin injections (50 mg/kg, i.p) for two weeks. Arbutin significantly improved memory impairment 14 days post-demyelination as assessed by Morris water maze test. Histological and immunohistochemical analyses demonstrated that arbutin reduced demyelination suppressed pro-inflammatory markers (IL-1β, TNF-α) and increased anti-inflammatory cytokine IL-10. Arbutin also diminished astrocyte activation, decreased iNOS, enhanced anti-oxidative factors (Nrf2, HO-1), and exhibited neuroprotective effects by elevating myelin markers (MBP) and brain derived neurotrophic factor (BDNF). These findings propose arbutin as a potential therapeutic candidate for multiple sclerosis-associated memory deficits, warranting further clinical exploration.
Collapse
Affiliation(s)
- Sahand Ashrafpour
- Mobility Impairment Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran; Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | | | - Alie Sabouri-Rad
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Soheila Hosseinzadeh
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Fereshteh Pourabdolhossein
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran; Department of Physiology, School of Medicine, Babol University of Medical Sciences, Babol, Iran.
| |
Collapse
|
18
|
Kornfeld SF, Cummings SE, Yaworski R, De Repentigny Y, Gagnon S, Zandee S, Fathi S, Prat A, Kothary R. Loss of miR-145 promotes remyelination and functional recovery in a model of chronic central demyelination. Commun Biol 2024; 7:813. [PMID: 38965401 PMCID: PMC11224363 DOI: 10.1038/s42003-024-06513-x] [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: 09/23/2020] [Accepted: 06/27/2024] [Indexed: 07/06/2024] Open
Abstract
Strategies for treating progressive multiple sclerosis (MS) remain limited. Here, we found that miR-145-5p is overabundant uniquely in chronic lesion tissues from secondary progressive MS patients. We induced both acute and chronic demyelination in miR-145 knockout mice to determine its contributions to remyelination failure. Following acute demyelination, no advantage to miR-145 loss could be detected. However, after chronic demyelination, animals with miR-145 loss demonstrated increased remyelination and functional recovery, coincident with altered presence of astrocytes and microglia within the corpus callosum relative to wild-type animals. This improved response in miR-145 knockout animals coincided with a pathological upregulation of miR-145-5p in wild-type animals with chronic cuprizone exposure, paralleling human chronic lesions. Furthermore, miR-145 overexpression specifically in oligodendrocytes (OLs) severely stunted differentiation and negatively impacted survival. RNAseq analysis showed altered transcriptome in these cells with downregulated major pathways involved in myelination. Our data suggest that pathological accumulation of miR-145-5p is a distinctive feature of chronic demyelination and is strongly implicated in the failure of remyelination, possibly due to the inhibition of OL differentiation together with alterations in other glial cells. This is mirrored in chronic MS lesions, and thus miR-145-5p serves as a potential relevant therapeutic target in progressive forms of MS.
Collapse
Affiliation(s)
- Samantha F Kornfeld
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Sarah E Cummings
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Rebecca Yaworski
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Yves De Repentigny
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
| | - Sabrina Gagnon
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
| | - Stephanie Zandee
- Neuroimmunology Unit and Multiple Sclerosis Clinic, The Research Center of the Centre Hospitalier de l'Université de Montréal (CRCHUM), Department of Neuroscience, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Samaneh Fathi
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
| | - Alexandre Prat
- Neuroimmunology Unit and Multiple Sclerosis Clinic, The Research Center of the Centre Hospitalier de l'Université de Montréal (CRCHUM), Department of Neuroscience, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Rashmi Kothary
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada.
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada.
- Department of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada.
- Centre for Neuromuscular Disease, University of Ottawa, Ottawa, ON, K1H 8M5, Canada.
| |
Collapse
|
19
|
Elendu C, Amaechi DC, Elendu TC, Ozigis MO, Adegbola MO, Adebayo MA, Afolabi OG. Renal disease associated with multiple sclerosis: A narrative review. Medicine (Baltimore) 2024; 103:e38222. [PMID: 38758897 PMCID: PMC11098179 DOI: 10.1097/md.0000000000038222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 03/14/2024] [Indexed: 05/19/2024] Open
Abstract
Multiple sclerosis (MS) is a chronic autoimmune neurological disorder characterized by central nervous system demyelination, leading to various neurological impairments. While the primary focus of research and clinical management has centered on the neurological aspects of MS, emerging evidence suggests a complex interplay between MS and renal disease. This narrative review endeavors to elucidate the intriguing association between MS and renal disease, providing a comprehensive overview of the current knowledge on this topic. Our review begins by outlining the pathophysiology of MS and the diverse mechanisms contributing to its progression. We then delve into renal disease, categorizing the various types and their clinical presentations. This review focuses on exploring the intricate relationship between these seemingly distinct conditions. We analyze existing literature to uncover shared risk factors, potential pathophysiological links, and the impact of MS on renal function. Furthermore, we discuss the clinical presentation and diagnostic challenges in identifying renal disease in MS patients. Importantly, we examine available treatment options and their efficacy in managing renal complications in this unique patient population. The consequences of renal disease on the overall quality of life (QOL) for individuals living with MS are also examined, shedding light on the multifaceted burden of these coexisting conditions.
Collapse
|
20
|
Zamecnik CR, Sowa GM, Abdelhak A, Dandekar R, Bair RD, Wade KJ, Bartley CM, Kizer K, Augusto DG, Tubati A, Gomez R, Fouassier C, Gerungan C, Caspar CM, Alexander J, Wapniarski AE, Loudermilk RP, Eggers EL, Zorn KC, Ananth K, Jabassini N, Mann SA, Ragan NR, Santaniello A, Henry RG, Baranzini SE, Zamvil SS, Sabatino JJ, Bove RM, Guo CY, Gelfand JM, Cuneo R, von Büdingen HC, Oksenberg JR, Cree BAC, Hollenbach JA, Green AJ, Hauser SL, Wallin MT, DeRisi JL, Wilson MR. An autoantibody signature predictive for multiple sclerosis. Nat Med 2024; 30:1300-1308. [PMID: 38641750 PMCID: PMC11980355 DOI: 10.1038/s41591-024-02938-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 03/21/2024] [Indexed: 04/21/2024]
Abstract
Although B cells are implicated in multiple sclerosis (MS) pathophysiology, a predictive or diagnostic autoantibody remains elusive. In this study, the Department of Defense Serum Repository (DoDSR), a cohort of over 10 million individuals, was used to generate whole-proteome autoantibody profiles of hundreds of patients with MS (PwMS) years before and subsequently after MS onset. This analysis defines a unique cluster in approximately 10% of PwMS who share an autoantibody signature against a common motif that has similarity with many human pathogens. These patients exhibit antibody reactivity years before developing MS symptoms and have higher levels of serum neurofilament light (sNfL) compared to other PwMS. Furthermore, this profile is preserved over time, providing molecular evidence for an immunologically active preclinical period years before clinical onset. This autoantibody reactivity was validated in samples from a separate incident MS cohort in both cerebrospinal fluid and serum, where it is highly specific for patients eventually diagnosed with MS. This signature is a starting point for further immunological characterization of this MS patient subset and may be clinically useful as an antigen-specific biomarker for high-risk patients with clinically or radiologically isolated neuroinflammatory syndromes.
Collapse
Affiliation(s)
- Colin R Zamecnik
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Gavin M Sowa
- University of California, San Francisco School of Medicine, San Francisco, CA, USA
- Department of Medicine, McGaw Medical Center of Northwestern University, Chicago, IL, USA
| | - Ahmed Abdelhak
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Ravi Dandekar
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Rebecca D Bair
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Kristen J Wade
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Christopher M Bartley
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Kerry Kizer
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Danillo G Augusto
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Asritha Tubati
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Refujia Gomez
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Camille Fouassier
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Chloe Gerungan
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Colette M Caspar
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Jessica Alexander
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Anne E Wapniarski
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Rita P Loudermilk
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Erica L Eggers
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Kelsey C Zorn
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
| | - Kirtana Ananth
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Nora Jabassini
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Sabrina A Mann
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
- Chan Zuckerberg Biohub San Francisco, San Francisco, CA, USA
| | - Nicholas R Ragan
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Adam Santaniello
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Roland G Henry
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Sergio E Baranzini
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Scott S Zamvil
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Joseph J Sabatino
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Riley M Bove
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Chu-Yueh Guo
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Jeffrey M Gelfand
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Richard Cuneo
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - H-Christian von Büdingen
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Jorge R Oksenberg
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Bruce A C Cree
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Jill A Hollenbach
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
| | - Ari J Green
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Stephen L Hauser
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Mitchell T Wallin
- Department of Veterans Affairs, Multiple Sclerosis Center of Excellence, Washington, DC, USA
- University of Maryland School of Medicine, Baltimore, MD, USA
| | - Joseph L DeRisi
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
- Chan Zuckerberg Biohub San Francisco, San Francisco, CA, USA
| | - Michael R Wilson
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
21
|
Hou B, Yin J, Liu S, Guo J, Zhang B, Zhang Z, Yang L, Tan X, Long Y, Feng S, Zhou J, Wu Y, Wang X, Han S, Wang Z, He X. Inhibiting the NLRP3 Inflammasome with MCC950 Alleviates Neurological Impairment in the Brain of EAE Mice. Mol Neurobiol 2024; 61:1318-1330. [PMID: 37702910 PMCID: PMC10896958 DOI: 10.1007/s12035-023-03618-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 08/27/2023] [Indexed: 09/14/2023]
Abstract
Multiple sclerosis (MS) is a chronic disease that is characterized by demyelination and neuronal damage. Experimental autoimmune encephalomyelitis (EAE) mice are used to model the disease progression of MS and mirror MS-like pathology. Previous researches have confirmed that inhibition of NLRP3 inflammasome significantly alleviated the severity of EAE mice and the demyelination of spinal cord, but its effect on neuronal damage and oligodendrocyte loss in the brain remains unclear. In this study, female C57BL/6 mice were immunized with MOG35-55 and PTX to establish experimental autoimmune encephalomyelitis (EAE) model. MCC950, a selective NLRP3 inflammasome inhibitor, was used to investigate the effect of NLRP3 inflammasome on the pathological changes and glial cell activation in the brain of EAE mice by immunohistochemistry. Our results demonstrated that MCC950 ameliorated the neuronal damage, demyelination, and oligodendrocyte loss in the brain of EAE mice. This protective effect of MCC950 may be attributed to its ability to suppress the activation of glial cells and prevents microglia polarization to M1 phenotype. Our work indicates that inhibition of NLRP3 inflammasome has the therapeutic effects of neuroprotection through immunomodulation and is a promising therapeutic strategy for MS.
Collapse
Affiliation(s)
- Baohua Hou
- College of Medicine, Henan Polytechnic University, Jiaozuo, 454000, China
- Department of Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430000, China
- Central Laboratory, The First Affiliated Hospital of Henan Polytechnic University (Jiaozuo Second People's Hospital), Jiaozuo, China
| | - Jun Yin
- Department of Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430000, China
| | - Shuyan Liu
- Department of Endocrinology, The First Affiliated Hospital of Henan Polytechnic University (Jiaozuo Second People's Hospital), Jiaozuo, 454000, China
| | - Jincheng Guo
- Department of Thoracic Surgery, The First Affiliated Hospital of Henan Polytechnic University (Jiaozuo Second People's Hospital), Jiaozuo, 454000, China
| | - Baobao Zhang
- College of Medicine, Henan Polytechnic University, Jiaozuo, 454000, China
| | - Zhenzhen Zhang
- College of Medicine, Henan Polytechnic University, Jiaozuo, 454000, China
| | - Lanping Yang
- College of Medicine, Henan Polytechnic University, Jiaozuo, 454000, China
| | - Xiying Tan
- College of Medicine, Henan Polytechnic University, Jiaozuo, 454000, China
| | - Yijiao Long
- College of Medicine, Henan Polytechnic University, Jiaozuo, 454000, China
| | - Sijie Feng
- College of Medicine, Henan Polytechnic University, Jiaozuo, 454000, China
| | - Jingchun Zhou
- Beijing Bencaoyuan Pharmaceutical Co, Ltd, Beijing, 102629, China
| | - Yifan Wu
- Department of Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430000, China
| | - Xueyang Wang
- Department of Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430000, China
| | - Song Han
- Department of Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430000, China
| | - Zhenhui Wang
- College of Medicine, Henan Polytechnic University, Jiaozuo, 454000, China.
| | - Xiaohua He
- Department of Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430000, China.
| |
Collapse
|
22
|
Raineri D, Abreu H, Vilardo B, Kustrimovic N, Venegoni C, Cappellano G, Chiocchetti A. Deep Flow Cytometry Unveils Distinct Immune Cell Subsets in Inducible T Cell Co-Stimulator Ligand (ICOSL)- and ICOS-Knockout Mice during Experimental Autoimmune Encephalomyelitis. Int J Mol Sci 2024; 25:2509. [PMID: 38473756 DOI: 10.3390/ijms25052509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 02/10/2024] [Accepted: 02/16/2024] [Indexed: 03/14/2024] Open
Abstract
The inducible T cell co-stimulator ligand (ICOSL), expressed by antigen presenting cells, binds to the inducible T cell co-stimulator (ICOS) on activated T cells. Improper function of the ICOS/ICOSL pathway has been implicated in several autoimmune diseases, including multiple sclerosis (MS). Previous studies showed that ICOS-knockout (KO) mice exhibit severe experimental autoimmune encephalomyelitis (EAE), the animal model of MS, but data on ICOSL deficiency are not available. In our study, we explored the impact of both ICOS and ICOSL deficiencies on MOG35-55 -induced EAE and its associated immune cell dynamics by employing ICOSL-KO and ICOS-KO mice with a C57BL/6J background. During EAE resolution, MOG-driven cytokine levels and the immunophenotype of splenocytes were evaluated by ELISA and multiparametric flow cytometry, respectively. We found that both KO mice exhibited an overlapping and more severe EAE compared to C57BL/6J mice, corroborated by a reduction in memory/regulatory T cell subsets and interleukin (IL-)17 levels. It is noteworthy that an unsupervised analysis showed that ICOSL deficiency modifies the immune response in an original way, by affecting T central and effector memory (TCM, TEM), long-lived CD4+ TEM cells, and macrophages, compared to ICOS-KO and C57BL/6J mice, suggesting a role for other binding partners to ICOSL in EAE development, which deserves further study.
Collapse
Affiliation(s)
- Davide Raineri
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, University of Eastern Piedmont, 28100 Novara, Italy
- Center for Translational Research on Autoimmune and Allergic Disease-CAAD, University of Eastern Piedmont, 28100 Novara, Italy
| | - Hugo Abreu
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, University of Eastern Piedmont, 28100 Novara, Italy
- Center for Translational Research on Autoimmune and Allergic Disease-CAAD, University of Eastern Piedmont, 28100 Novara, Italy
| | - Beatrice Vilardo
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, University of Eastern Piedmont, 28100 Novara, Italy
- Center for Translational Research on Autoimmune and Allergic Disease-CAAD, University of Eastern Piedmont, 28100 Novara, Italy
| | - Natasa Kustrimovic
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, University of Eastern Piedmont, 28100 Novara, Italy
- Center for Translational Research on Autoimmune and Allergic Disease-CAAD, University of Eastern Piedmont, 28100 Novara, Italy
| | - Chiara Venegoni
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, University of Eastern Piedmont, 28100 Novara, Italy
- Center for Translational Research on Autoimmune and Allergic Disease-CAAD, University of Eastern Piedmont, 28100 Novara, Italy
| | - Giuseppe Cappellano
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, University of Eastern Piedmont, 28100 Novara, Italy
- Center for Translational Research on Autoimmune and Allergic Disease-CAAD, University of Eastern Piedmont, 28100 Novara, Italy
| | - Annalisa Chiocchetti
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, University of Eastern Piedmont, 28100 Novara, Italy
- Center for Translational Research on Autoimmune and Allergic Disease-CAAD, University of Eastern Piedmont, 28100 Novara, Italy
| |
Collapse
|
23
|
Mendes O. Inflammation and neurodegeneration in multiple sclerosis. A REVIEW ON DIVERSE NEUROLOGICAL DISORDERS 2024:321-345. [DOI: 10.1016/b978-0-323-95735-9.00023-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
24
|
Lam M, Lee D, Kosater I, Khairallah A, Taga M, Zhang Y, Fujita M, Nag S, Bennett DA, De Jager P, Menon V. Human disease-specific cell signatures in non-lesional tissue in Multiple Sclerosis detected by single-cell and spatial transcriptomics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.20.572491. [PMID: 38187779 PMCID: PMC10769298 DOI: 10.1101/2023.12.20.572491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Recent investigations of cell type changes in Multiple Sclerosis (MS) using single-cell profiling methods have focused on active lesional and peri-lesional brain tissue, and have implicated a number of peripheral and central nervous system cell types. However, an important question is the extent to which so-called "normal-appearing" non-lesional tissue in individuals with MS accumulate changes over the lifespan. Here, we compared post-mortem non-lesional brain tissue from donors with a pathological or clinical diagnosis of MS from the Religious Orders Study or Rush Memory and Aging Project (ROSMAP) cohorts to age- and sex-matched brains from persons without MS (controls). We profiled three brain regions using single-nucleus RNA-seq: dorsolateral prefrontal cortex (DLPFC), normal appearing white matter (NAWM) and the pulvinar in thalamus (PULV), from 15 control individuals, 8 individuals with MS, and 5 individuals with other detrimental pathologies accompanied by demyelination, resulting in a total of 78 samples. We identified region- and cell type-specific differences in non-lesional samples from individuals diagnosed with MS and/or exhibiting secondary demyelination with other neurological conditions, as compared to control donors. These differences included lower proportions of oligodendrocytes with expression of myelination related genes MOBP, MBP, PLP1, as well as higher proportions of CRYAB+ oligodendrocytes in all three brain regions. Among microglial signatures, we identified subgroups that were higher in both demyelination (TMEM163+/ERC2+), as well as those that were specifically higher in MS donors (HIF1A+/SPP1+) and specifically in donors with secondary demyelination (SOCS6+/MYO1E+), in both white and grey matter. To validate our findings, we generated Visium spatial transcriptomics data on matched tissue from 13 donors, and recapitulated our observations of gene expression differences in oligodendrocytes and microglia. Finally, we show that some of the differences observed between control and MS donors in NAWM mirror those previously reported between control WM and active lesions in MS donors. Overall, our investigation sheds additional light on cell type- and disease-specific differences present even in non-lesional white and grey matter tissue, highlighting widespread cellular signatures that may be associated with downstream pathological changes.
Collapse
Affiliation(s)
- Matti Lam
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center
| | - Dylan Lee
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center
| | - Ivy Kosater
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center
| | - Anthony Khairallah
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center
| | - Mariko Taga
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center
| | - Ya Zhang
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center
| | - Masashi Fujita
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center
| | - Sukriti Nag
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL
| | - Philip De Jager
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center
| | - Vilas Menon
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center
| |
Collapse
|
25
|
Caranova M, Soares JF, Batista S, Castelo-Branco M, Duarte JV. A systematic review of microstructural abnormalities in multiple sclerosis detected with NODDI and DTI models of diffusion-weighted magnetic resonance imaging. Magn Reson Imaging 2023; 104:61-71. [PMID: 37775062 DOI: 10.1016/j.mri.2023.09.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/31/2023] [Accepted: 09/25/2023] [Indexed: 10/01/2023]
Abstract
Multiple sclerosis (MS), namely the phenotype of the relapsing-remitting form, is the most common white matter disease and is mostly characterized by demyelination and inflammation, which lead to neurodegeneration and cognitive decline. Its diagnosis and monitoring are performed through conventional structural MRI, in which T2-hyperintense lesions can be identified, but this technique lacks sensitivity and specificity, mainly in detecting damage to normal appearing tissues. Models of diffusion-weighted MRI such as diffusion-tensor imaging (DTI) and neurite orientation dispersion and density imaging (NODDI) allow to uncover microstructural abnormalities that occur in MS, mainly in normal appearing tissues such as the normal appearing white matter (NAWM), which allows to overcome limitations of conventional MRI. DTI is the standard method used for modelling this kind of data, but it has limitations, which can be tackled by using more complex diffusion models, such as NODDI, which provides additional information on morphological properties of tissues. Although there are several studies in MS using both diffusion models, there is no formal assessment that summarizes the findings of both methods in lesioned and normal appearing tissues, and whether one is more advantageous than the other. Hence, this systematic review aims to identify what microstructural abnormalities are seen in lesions and/or NAWM in relapsing-remitting MS while using two different approaches to modelling diffusion data, namely DTI and NODDI, and if one of them is more appropriate than the other or if they are complementary to each other. The search was performed using PubMed, which was last searched on November 2022, and aimed at finding studies that either utilized both DTI and NODDI in the same dataset, or only one of the methods. Eleven articles were included in this review, which included cohorts with a relatively low sample size (total number of patients = 254, total number of healthy controls = 240), and patients with a moderate disease duration, all with relapsing-remitting MS. Overall, studies found decreased fractional anisotropy (FA), neurite density index (NDI) and orientation dispersion index (ODI), and increased mean, axial and radial diffusivities (MD, AD and RD, respectively) in lesions, when compared to contralateral NAWM and healthy controls' white matter. Compared to healthy controls' white matter, NAWM showed lower FA and NDI and higher MD, AD, RD, and ODI. Results from the included articles confirm that there is active demyelination and inflammation in both lesions and NAWM, as well as loss in neurites, and that structural damage is not confined to focal lesions, which is in concordance with histological findings and results from other imaging techniques. Furthermore, NODDI is suggested to have higher sensitivity and specificity, as seen by inspecting imaging results, compared to DTI, while still being clinically feasible. The use of biomarkers derived from such advanced diffusion models in clinical practice could imply a better understanding of treatment efficacy and disease progression, without relying on the manifestation of clinical symptoms, such as relapses.
Collapse
Affiliation(s)
- Maria Caranova
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Institute for Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal.
| | - Júlia F Soares
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Institute for Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal
| | - Sónia Batista
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal; Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Miguel Castelo-Branco
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Institute for Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal; Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - João Valente Duarte
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Institute for Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal; Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
26
|
Corsi F, Deidda Tarquini G, Urbani M, Bejarano I, Traversa E, Ghibelli L. The Impressive Anti-Inflammatory Activity of Cerium Oxide Nanoparticles: More than Redox? NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:2803. [PMID: 37887953 PMCID: PMC10609664 DOI: 10.3390/nano13202803] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/04/2023] [Accepted: 10/19/2023] [Indexed: 10/28/2023]
Abstract
Cerium oxide nanoparticles (CNPs) are biocompatible nanozymes exerting multifunctional biomimetic activities, including superoxide dismutase (SOD), catalase, glutathione peroxidase, photolyase, and phosphatase. SOD- and catalase-mimesis depend on Ce3+/Ce4+ redox switch on nanoparticle surface, which allows scavenging the most noxious reactive oxygen species in a self-regenerating, energy-free manner. As oxidative stress plays pivotal roles in the pathogenesis of inflammatory disorders, CNPs have recently attracted attention as potential anti-inflammatory agents. A careful survey of the literature reveals that CNPs, alone or as constituents of implants and scaffolds, strongly contrast chronic inflammation (including neurodegenerative and autoimmune diseases, liver steatosis, gastrointestinal disorders), infections, and trauma, thereby ameliorating/restoring organ function. By general consensus, CNPs inhibit inflammation cues while boosting the pro-resolving anti-inflammatory signaling pathways. The mechanism of CNPs' anti-inflammatory effects has hardly been investigated, being rather deductively attributed to CNP-induced ROS scavenging. However, CNPs are multi-functional nanozymes that exert additional bioactivities independent from the Ce3+/Ce4+ redox switch, such as phosphatase activity, which could conceivably mediate some of the anti-inflammatory effects reported, suggesting that CNPs fight inflammation via pleiotropic actions. Since CNP anti-inflammatory activity is potentially a pharmacological breakthrough, it is important to precisely attribute the described effects to one or another of their nanozyme functions, thus achieving therapeutic credibility.
Collapse
Affiliation(s)
- Francesca Corsi
- Department of Chemical Science and Technologies, University of Rome “Tor Vergata”, 00133 Rome, Italy; (G.D.T.); (M.U.); (E.T.)
- Department of Biology, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | - Greta Deidda Tarquini
- Department of Chemical Science and Technologies, University of Rome “Tor Vergata”, 00133 Rome, Italy; (G.D.T.); (M.U.); (E.T.)
- Department of Biology, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | - Marta Urbani
- Department of Chemical Science and Technologies, University of Rome “Tor Vergata”, 00133 Rome, Italy; (G.D.T.); (M.U.); (E.T.)
- Department of Biology, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | - Ignacio Bejarano
- Institute of Biomedicine of Seville (IBiS), University of Seville, HUVR, Junta de Andalucía, CSIC, 41013 Seville, Spain;
- Department of Medical Biochemistry, Molecular Biology and Immunology, University of Seville, 41004 Seville, Spain
| | - Enrico Traversa
- Department of Chemical Science and Technologies, University of Rome “Tor Vergata”, 00133 Rome, Italy; (G.D.T.); (M.U.); (E.T.)
| | - Lina Ghibelli
- Department of Biology, University of Rome “Tor Vergata”, 00133 Rome, Italy
| |
Collapse
|
27
|
de Villers-Sidani É, Voss P, Bastien N, Cisneros-Franco JM, Hussein S, Mayo NE, Koch NA, Drouin-Picaro A, Blanchette F, Guitton D, Giacomini PS. Oculomotor analysis to assess brain health: preliminary findings from a longitudinal study of multiple sclerosis using novel tablet-based eye-tracking software. Front Neurol 2023; 14:1243594. [PMID: 37745656 PMCID: PMC10516298 DOI: 10.3389/fneur.2023.1243594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 08/07/2023] [Indexed: 09/26/2023] Open
Abstract
A growing body of evidence supports the link between eye movement anomalies and brain health. Indeed, the oculomotor system is composed of a diverse network of cortical and subcortical structures and circuits that are susceptible to a variety of degenerative processes. Here we show preliminary findings from the baseline measurements of an ongoing longitudinal cohort study in MS participants, designed to determine if disease and cognitive status can be estimated and tracked with high accuracy based on eye movement parameters alone. Using a novel gaze-tracking technology that can reliably and accurately track eye movements with good precision without the need for infrared cameras, using only an iPad Pro embedded camera, we show in this cross-sectional study that several eye movement parameters significantly correlated with clinical outcome measures of interest. Eye movement parameters were extracted from fixation, pro-saccade, anti-saccade, and smooth pursuit visual tasks, whereas the clinical outcome measures were the scores of several disease assessment tools and standard cognitive tests such as the Expanded Disability Status Scale (EDSS), Brief International Cognitive Assessment for MS (BICAMS), the Multiple Sclerosis Functional Composite (MSFC) and the Symbol Digit Modalities Test (SDMT). Furthermore, partial least squares regression analyses show that a small set of oculomotor parameters can explain up to 84% of the variance of the clinical outcome measures. Taken together, these findings not only replicate previously known associations between eye movement parameters and clinical scores, this time using a novel mobile-based technology, but also the notion that interrogating the oculomotor system with a novel eye-tracking technology can inform us of disease severity, as well as the cognitive status of MS participants.
Collapse
Affiliation(s)
- Étienne de Villers-Sidani
- Innodem Neurosciences, Montreal, QC, Canada
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Patrice Voss
- Innodem Neurosciences, Montreal, QC, Canada
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | | | - J. Miguel Cisneros-Franco
- Innodem Neurosciences, Montreal, QC, Canada
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | | | - Nancy E. Mayo
- Faculty of Medicine, School of Physical and Occupational Therapy, McGill University, Montreal, QC, Canada
| | - Nils A. Koch
- Innodem Neurosciences, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | | | | | - Daniel Guitton
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Paul S. Giacomini
- Innodem Neurosciences, Montreal, QC, Canada
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| |
Collapse
|
28
|
Klehmet J, Begus-Nahrmann Y, Taipale K, Niemczyk G, Rehberg-Weber K. Impact of interferon beta exposure on birth outcome and child development - Results from the post-authorisation safety study PRIMA. Mult Scler Relat Disord 2023; 77:104844. [PMID: 37393802 DOI: 10.1016/j.msard.2023.104844] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/26/2023] [Accepted: 06/19/2023] [Indexed: 07/04/2023]
Abstract
BACKGROUND Interferon beta therapies are well-established disease-modifying treatments for patients with relapsing multiple sclerosis (MS). Based on clinical evidence from two large cohort studies, both, the EMA and FDA updated the labels of the interferon beta class in terms of pregnancy and breastfeeding in 2019 and 2020, respectively. To complement pregnancy label updates with patient-reported real-world data, this study examined German pregnancy and outcome reports including available data on child development from women with MS treated with peginterferon beta-1a or intramuscular (IM) interferon beta-1a. METHODS The post-authorisation safety study PRIMA included adult women diagnosed with relapsing-remitting MS or clinically isolated syndrome, who were treated with peginterferon beta-1a or IM interferon beta-1a before or during pregnancy and registered in the marketing authorisation holder's MS Service center patient support program. In the prospective part of the study, conducted from April to October 2021, data on developmental milestones of the newborns were collected via telephone interview from mothers reporting live births. RESULTS In total, 426 women were enrolled, reporting 542 pregnancies that resulted in 466 live births. A total of 162 women completed the questionnaire for 192 live births (53.1% male). Newborns had Apgar scores indicative of healthy infants. Weight, length and head circumference at birth and physical growth curves up to 48 months lay within the expected range of the German general population. Most newborn screenings and examinations during check-ups were inconspicuous over the study period of 48 months. Out of 158 breastfed infants, 112 (70.9%) were breastfed exclusively until month 5. CONCLUSION Study results confirmed former reports indicating that exposure to interferon beta therapies during pregnancy or lactation had no adverse effects on intrauterine growth and child development over the study period, which covered the first 4 years of life. These real-world data obtained within the scope of a patient support program for peginterferon beta-1a or IM interferon beta-1a corroborate German and Scandinavian registry data and support the label update of all interferon beta therapies. REGISTRATION NCT04655222, EUPAS38347.
Collapse
Affiliation(s)
- Juliane Klehmet
- Department of Neurology, Jüdisches Krankenhaus Berlin, Heinz-Galinski-Strasse 1, Berlin-Mitte 13347, Federal Republic of Germany.
| | | | - Kirsi Taipale
- Biogen GmbH, Riedenburger Straße 7, München 81677, Federal Republic of Germany
| | - Gabriele Niemczyk
- Biogen GmbH, Riedenburger Straße 7, München 81677, Federal Republic of Germany
| | - Karin Rehberg-Weber
- Biogen GmbH, Riedenburger Straße 7, München 81677, Federal Republic of Germany
| |
Collapse
|
29
|
Ahmed A, Saleem MA, Saeed F, Afzaal M, Imran A, Akram S, Hussain M, Khan A, Al Jbawi E. A comprehensive review on the impact of calcium and vitamin D insufficiency and allied metabolic disorders in females. Food Sci Nutr 2023; 11:5004-5027. [PMID: 37701195 PMCID: PMC10494632 DOI: 10.1002/fsn3.3519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 06/01/2023] [Accepted: 06/01/2023] [Indexed: 09/14/2023] Open
Abstract
Calcium is imperative in maintaining a quality life, particularly during later ages. Its deficiency results in a wide range of metabolic disorders such as dental changes, cataracts, alterations in brain function, and osteoporosis. These deficiencies are more pronounced in females due to increased calcium turnover throughout their life cycle, especially during pregnancy and lactation. Vitamin D perform a central role in the metabolism of calcium. Recent scientific interventions have linked calcium with an array of metabolic disorders in females including hypertension, obesity, premenstrual dysphoric disorder, polycystic ovary syndrome (PCOS), multiple sclerosis, and breast cancer. This review encompasses these female metabolic disorders with special reference to calcium and vitamin D deficiency. This review article aims to present and elaborate on available data regarding the worldwide occurrence of insufficient calcium consumption in females and allied health risks, to provide a basis for formulating strategies and population-level scientific studies to adequately boost calcium intake and position where required.
Collapse
Affiliation(s)
- Aftab Ahmed
- Department of Nutritional SciencesGovernment College University FaisalabadFaisalabadPakistan
| | - Muhammad Awais Saleem
- Department of Nutritional SciencesGovernment College University FaisalabadFaisalabadPakistan
- Department of Human Nutrition and DieteticsMirpur University of Science and TechnologyMirpurPakistan
| | - Farhan Saeed
- Department of Food SciencesGovernment College University FaisalabadFaisalabadPakistan
| | - Muhammad Afzaal
- Department of Food SciencesGovernment College University FaisalabadFaisalabadPakistan
| | - Ali Imran
- Department of Food SciencesGovernment College University FaisalabadFaisalabadPakistan
| | - Sidra Akram
- Department of Nutritional SciencesGovernment College University FaisalabadFaisalabadPakistan
| | - Muzzamal Hussain
- Department of Food SciencesGovernment College University FaisalabadFaisalabadPakistan
| | - Aqsa Khan
- Department of Nutritional SciencesGovernment College University FaisalabadFaisalabadPakistan
| | | |
Collapse
|
30
|
Balraj A, Clarkson-Paredes C, Miller RH. Loss of optic nerve oligodendrocytes during maturation alters retinal organization. Exp Eye Res 2023:109540. [PMID: 37364629 DOI: 10.1016/j.exer.2023.109540] [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: 02/28/2023] [Revised: 06/01/2023] [Accepted: 06/12/2023] [Indexed: 06/28/2023]
Abstract
The myelin sheath facilitates signal conduction along axons in white matter tracts, and when disrupted, can result in significant functional deficits. Demyelination, observed in diseases like multiple sclerosis and optic neuritis, are associated with neural degeneration, however the extent of this damage on upstream circuitry is not well understood. Here we use the MBP-iCP9 mouse model to induce selective oligodendrocyte ablation in the optic nerve at P14 via a chemical inducer of dimerization (CID), resulting in partial demyelination of retinal ganglion cell (RGC) axons with minimal inflammation after two weeks. Oligodendrocyte loss reduced axon diameter and altered compound action potential waveforms, blocking conduction in the slowest-conducting axon populations. Demyelination resulted in disruptions to the normal composition of the retina, including reduced density of RBPMS+, Brn3a+, and OFF-transient RGCs, thinning of the IPL, and reduced density of displaced amacrine cells. The INL and ONL were unaffected by oligodendrocyte loss, suggesting that demyelination-induced deficits in this model are specific to the IPL and GCL. These results show that a partial demyelination of a subpopulation of RGC axons disrupts optic nerve function and affects the organization of the retinal network. This study highlights the significance of myelination in maintaining upstream neural connectivity and provides support for targeting neuronal degeneration in treatments of demyelinating diseases.
Collapse
Affiliation(s)
- Annika Balraj
- Department of Anatomy and Cell Biology, The George Washington University, School of Medicine and Health Sciences, Washington, D.C., USA
| | - Cheryl Clarkson-Paredes
- Department of Anatomy and Cell Biology, The George Washington University, School of Medicine and Health Sciences, Washington, D.C., USA
| | - Robert H Miller
- Department of Anatomy and Cell Biology, The George Washington University, School of Medicine and Health Sciences, Washington, D.C., USA.
| |
Collapse
|
31
|
Zamecnik CR, Sowa GM, Abdelhak A, Dandekar R, Bair RD, Wade KJ, Bartley CM, Tubati A, Gomez R, Fouassier C, Gerungan C, Alexander J, Wapniarski AE, Loudermilk RP, Eggers EL, Zorn KC, Ananth K, Jabassini N, Mann SA, Ragan NR, Santaniello A, Henry RG, Baranzini SE, Zamvil SS, Bove RM, Guo CY, Gelfand JM, Cuneo R, von Büdingen HC, Oksenberg JR, Cree BAC, Hollenbach JA, Green AJ, Hauser SL, Wallin MT, DeRisi JL, Wilson MR. A Predictive Autoantibody Signature in Multiple Sclerosis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.05.01.23288943. [PMID: 37205595 PMCID: PMC10187343 DOI: 10.1101/2023.05.01.23288943] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Although B cells are implicated in multiple sclerosis (MS) pathophysiology, a predictive or diagnostic autoantibody remains elusive. Here, the Department of Defense Serum Repository (DoDSR), a cohort of over 10 million individuals, was used to generate whole-proteome autoantibody profiles of hundreds of patients with MS (PwMS) years before and subsequently after MS onset. This analysis defines a unique cluster of PwMS that share an autoantibody signature against a common motif that has similarity with many human pathogens. These patients exhibit antibody reactivity years before developing MS symptoms and have higher levels of serum neurofilament light (sNfL) compared to other PwMS. Furthermore, this profile is preserved over time, providing molecular evidence for an immunologically active prodromal period years before clinical onset. This autoantibody reactivity was validated in samples from a separate incident MS cohort in both cerebrospinal fluid (CSF) and serum, where it is highly specific for patients eventually diagnosed with MS. This signature is a starting point for further immunological characterization of this MS patient subset and may be clinically useful as an antigen-specific biomarker for high-risk patients with clinically- or radiologically-isolated neuroinflammatory syndromes.
Collapse
Affiliation(s)
- Colin R. Zamecnik
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Gavin M. Sowa
- Department of Medicine, McGaw Medical Center of Northwestern University, Chicago, IL, USA
| | - Ahmed Abdelhak
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Ravi Dandekar
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Rebecca D. Bair
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Kristen J. Wade
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Christopher M. Bartley
- UCSF Weill Institute for Neurosciences, Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA, USA
| | - Asritha Tubati
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Refujia Gomez
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Camille Fouassier
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Chloe Gerungan
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Jessica Alexander
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Anne E. Wapniarski
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Rita P. Loudermilk
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Erica L. Eggers
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Kelsey C. Zorn
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
| | - Kirtana Ananth
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Nora Jabassini
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Sabrina A. Mann
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
| | - Nicholas R. Ragan
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Adam Santaniello
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Roland G. Henry
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Sergio E. Baranzini
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Scott S. Zamvil
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Riley M. Bove
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Chu-Yueh Guo
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Jeffrey M. Gelfand
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Richard Cuneo
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - H.-Christian von Büdingen
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Jorge R. Oksenberg
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Bruce AC Cree
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Jill A. Hollenbach
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA USA
| | - Ari J. Green
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Stephen L. Hauser
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Mitchell T. Wallin
- Veterans Affairs, Multiple Sclerosis Center of Excellence, Washington, DC and University of Maryland School of Medicine, Baltimore, MD, USA
| | - Joseph L. DeRisi
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Michael R. Wilson
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| |
Collapse
|
32
|
Sulimai N, Brown J, Lominadze D. Vascular Effects on Cerebrovascular Permeability and Neurodegeneration. Biomolecules 2023; 13:648. [PMID: 37189395 PMCID: PMC10136045 DOI: 10.3390/biom13040648] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 03/29/2023] [Accepted: 04/02/2023] [Indexed: 05/17/2023] Open
Abstract
Neurons and glial cells in the brain are protected by the blood brain barrier (BBB). The local regulation of blood flow is determined by neurons and signal conducting cells called astrocytes. Although alterations in neurons and glial cells affect the function of neurons, the majority of effects are coming from other cells and organs of the body. Although it seems obvious that effects beginning in brain vasculature would play an important role in the development of various neuroinflammatory and neurodegenerative pathologies, significant interest has only been directed to the possible mechanisms involved in the development of vascular cognitive impairment and dementia (VCID) for the last decade. Presently, the National Institute of Neurological Disorders and Stroke applies considerable attention toward research related to VCID and vascular impairments during Alzheimer's disease. Thus, any changes in cerebral vessels, such as in blood flow, thrombogenesis, permeability, or others, which affect the proper vasculo-neuronal connection and interaction and result in neuronal degeneration that leads to memory decline should be considered as a subject of investigation under the VCID category. Out of several vascular effects that can trigger neurodegeneration, changes in cerebrovascular permeability seem to result in the most devastating effects. The present review emphasizes the importance of changes in the BBB and possible mechanisms primarily involving fibrinogen in the development and/or progression of neuroinflammatory and neurodegenerative diseases resulting in memory decline.
Collapse
Affiliation(s)
- Nurul Sulimai
- Department of Surgery, College of Medicine, University of South Florida Morsani, Tampa, FL 33612, USA; (N.S.); (J.B.)
| | - Jason Brown
- Department of Surgery, College of Medicine, University of South Florida Morsani, Tampa, FL 33612, USA; (N.S.); (J.B.)
| | - David Lominadze
- Department of Surgery, College of Medicine, University of South Florida Morsani, Tampa, FL 33612, USA; (N.S.); (J.B.)
- Department of Molecular Pharmacology and Physiology, College of Medicine, University of South Florida Morsani, Tampa, FL 33612, USA
| |
Collapse
|
33
|
Bissonnette JN, Pimer L, Francis AM, Hull KM, Leckey J, MacGillivray M, Berrigan LI, Fisher DJ. An investigation of auditory processing in relapsing-remitting multiple sclerosis. Exp Brain Res 2023; 241:1319-1327. [PMID: 37004533 DOI: 10.1007/s00221-023-06594-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 03/05/2023] [Indexed: 04/04/2023]
Abstract
Multiple sclerosis (MS) is one of the most common neurological diseases in North America and it is frequently associated with sensory processing difficulties, cognitive deficits, and psychiatric illness. While many studies have examined cognitive deficits in MS measured by behavioural responses and neuroimaging techniques, only a few studies have examined neurophysiological measures of auditory functioning in MS, such as the mismatch negativity (MMN). The MMN is an event-related potential that indicates automatic auditory change detection. This study examined whether MMN endpoints measured by electroencephalography (EEG) differ in individuals with relapsing-remitting MS compared to healthy controls and whether the symptomatology of MS, including symptoms of depression and fatigue, are related to MMN measures. A multi-feature MMN paradigm, which includes five distinct deviant tones, was used to assess auditory cortex function in MS. There were no significant differences in MMN amplitudes or latencies between the MS and control group (p < 0.05) and corresponding effect sizes were small. However, there was a correlation between reduced MMN amplitudes in response to an intensity deviant and physician-reported disability. The intensity MMN may be more sensitive to deterioration in this population. Ultimately, this study provides a comprehensive profile of early auditory processing abilities in MS and suggests that a reduction in the MMN response may be representative of disease severity in MS.
Collapse
Affiliation(s)
- Jenna N Bissonnette
- Department of Psychology, Mount Saint Vincent University, 166 Bedford Hwy, Halifax, NS, B3M 2J6, Canada
- Department of Psychiatry, Dalhousie University, Halifax, NS, Canada
| | - Laura Pimer
- Department of Psychology, Mount Saint Vincent University, 166 Bedford Hwy, Halifax, NS, B3M 2J6, Canada
| | - Ashley M Francis
- Department of Psychiatry, Dalhousie University, Halifax, NS, Canada
- Department of Psychology, St. Francis Xavier University, Antigonish, NS, Canada
| | - Krista M Hull
- Department of Psychology, Mount Saint Vincent University, 166 Bedford Hwy, Halifax, NS, B3M 2J6, Canada
| | - Jennifer Leckey
- Department of Psychology, Mount Saint Vincent University, 166 Bedford Hwy, Halifax, NS, B3M 2J6, Canada
| | - Megan MacGillivray
- Department of Psychology, St. Francis Xavier University, Antigonish, NS, Canada
| | - Lindsay I Berrigan
- Department of Psychiatry, Dalhousie University, Halifax, NS, Canada
- Department of Psychology, St. Francis Xavier University, Antigonish, NS, Canada
| | - Derek J Fisher
- Department of Psychology, Mount Saint Vincent University, 166 Bedford Hwy, Halifax, NS, B3M 2J6, Canada.
- Department of Psychiatry, Dalhousie University, Halifax, NS, Canada.
- Nova Scotia Health Authority, Halifax, NS, Canada.
| |
Collapse
|
34
|
Predictors of hypogammaglobulinemia and serious infections among patients receiving ocrelizumab or rituximab for treatment of MS and NMOSD. J Neuroimmunol 2023; 377:578066. [PMID: 36917920 DOI: 10.1016/j.jneuroim.2023.578066] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/01/2023] [Accepted: 03/05/2023] [Indexed: 03/14/2023]
Abstract
BACKGROUND AND OBJECTIVES Ocrelizumab and rituximab are monoclonal antibodies targeting the CD20 marker on B lymphocytes. The enhanced efficacy of B lymphocyte depleting therapies poses a greater risk of decreased immunoglobulin (Ig) levels. The rate and risk factors of hypogammaglobulinemia in MS and NMOSD patients treated with anti-CD20 therapies are unknown. METHODS A retrospective study was conducted among patients who received anti-CD20 therapy for the treatment of MS, NMOSD, and other related neurological disorders. The goal was to determine the incidence and risk factors of hypogammaglobulinemia and serious infections in patients receiving ocrelizumab versus rituximab. The secondary goals were to determine the rates of lymphopenia, neutropenia, and early B cell repopulation among patients on anti-CD20 therapy. RESULTS Overall, 184 patients (mean age 48.4 ± 13.7, 66.8% female) met inclusion criteria; 152 patients received ocrelizumab and 32 patients received rituximab. A total of 22 patients (12%) developed hypogammaglobulinemia. Patients who developed hypogammaglobulinemia were more likely to have been ≥50 years of age (p = .0275) with lower baseline IgG (p = .001) and IgA (p = .0038) levels. Serious infections were observed in 21 patients (11%) and seen more commonly in those that developed total lymphopenia (<1.0 × 109/L) and had longer duration of B-cell therapy. Multivariate analysis identified age ≥ 50 years, white race, and rituximab as independent predictors of hypogammaglobulinemia, and absolute lymphopenia as an independent risk factor for serious infections. DISCUSSION Among patients receiving anti-CD20 therapy, 12% of patients experienced hypogammaglobulinemia which was seen more commonly in white patients, at least 50 years old, with lower baseline IgG and IgA levels and in those treated with rituximab. Serious infections were seen more commonly in patients with total lymphopenia and longer exposure to anti-CD20 therapy.
Collapse
|
35
|
Wang L, Tan H, Yu J, ZhangBao J, Huang W, Chang X, Zhou L, Lu C, Xiao Y, Lu J, Zhao C, Wang M, Wu X, Wu M, Dong Q, Ngew KY, Quan C. Baseline retinal nerve fiber layer thickness as a predictor of multiple sclerosis progression: New insights from the FREEDOMS II study. Eur J Neurol 2023; 30:443-452. [PMID: 36286605 DOI: 10.1111/ene.15612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 10/13/2022] [Accepted: 10/20/2022] [Indexed: 01/07/2023]
Abstract
BACKGROUND AND PURPOSE The aim was to evaluate the potential of retinal nerve fiber layer thickness (RNFLT) measured with optical coherence tomography in predicting disease progression in relapsing-remitting multiple sclerosis (RRMS). METHODS Analyses were conducted post hoc of this 24-month, phase III, double-blind study, in which RRMS patients were randomized (1:1:1) to once daily oral fingolimod 0.5 mg, 1.25 mg or placebo. The key outcomes were the association between baseline RNFLT and baseline clinical characteristics and clinical/imaging outcomes up to 24 months. Change of RNFLT with fingolimod versus placebo within 24 months and time to retinal nerve fiber layer (RNFL) thinning were evaluated. RESULTS Altogether 885 patients were included. At baseline, lower RNFLT was correlated with higher Expanded Disability Status Scale score (r = -1.085, p = 0.018), lower brain volume (r = 0.025, p = 0.006) and deep gray matter volume (r = 0.731, p < 0.0001), worse visual acuity (r = -19.846, p < 0.0001) and longer duration since diagnosis (r = -0.258, p = 0.018). At month 12, low baseline RNFLT (<86 μm) versus high baseline RNFLT (≥99 μm) was associated with a greater brain volume loss (percentage change -0.605% vs. -0.315%, p = 0.035) in patients without optic neuritis history. At month 24, low baseline RNFLT versus high baseline RNFLT was associated with a higher number of new or newly enlarged T2 lesions (mean number 4.0 vs. 2.8, p = 0.014) and a higher risk of subsequent RNFL thinning (hazard ratio 2.55; 95% confidence interval 1.84-3.53; p < 0.001). The atrophy of the RNFL in the inferior quadrant was alleviated with fingolimod 0.5 mg versus placebo at month 24 (Δ(least squares mean) = 1.8, p = 0.047). CONCLUSION Retinal nerve fiber layer thickness could predict disease progression in RRMS. TRIAL REGISTRATION Clinicaltrials.gov identifier: NCT00355134, https://clinicaltrials.gov/ct2/show/NCT00355134.
Collapse
Affiliation(s)
- Liang Wang
- Department of Neurology and Rare Disease Center, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
| | - Hongmei Tan
- Department of Neurology and Rare Disease Center, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
| | - Jian Yu
- Department of Ophthalmology and Vision Science, Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jingzi ZhangBao
- Department of Neurology and Rare Disease Center, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
| | - Wenjuan Huang
- Department of Neurology and Rare Disease Center, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
| | - Xuechun Chang
- Department of Neurology and Rare Disease Center, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
| | - Lei Zhou
- Department of Neurology and Rare Disease Center, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
| | - Chuanzhen Lu
- Department of Neurology and Rare Disease Center, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yiqin Xiao
- Department of Ophthalmology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jiahong Lu
- Department of Neurology and Rare Disease Center, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chongbo Zhao
- Department of Neurology and Rare Disease Center, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Min Wang
- Department of Ophthalmology and Vision Science, Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xue Wu
- Novartis Pharmaceuticals, Shanghai, China
| | - Mengyun Wu
- Novartis Pharmaceuticals, Shanghai, China
| | - Qiang Dong
- Department of Neurology and Rare Disease Center, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Kok Yew Ngew
- Novartis Corporation (Malaysia) Sdn. Bhd., Petaling Jaya, Malaysia
| | - Chao Quan
- Department of Neurology and Rare Disease Center, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
36
|
Kalluri HV, Rosebraugh MR, Misko TP, Ziemann A, Liu W, Cree BAC. Phase 1 Evaluation of Elezanumab (Anti-Repulsive Guidance Molecule A Monoclonal Antibody) in Healthy and Multiple Sclerosis Participants. Ann Neurol 2023; 93:285-296. [PMID: 36093738 PMCID: PMC10100020 DOI: 10.1002/ana.26503] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 08/01/2022] [Accepted: 09/06/2022] [Indexed: 01/31/2023]
Abstract
OBJECTIVE This study was undertaken to describe the safety, tolerability, pharmacokinetics, and immunogenicity of elezanumab (ABT-555), a fully human monoclonal antibody (mAb) directed against repulsive guidance molecule A (RGMa), in healthy and multiple sclerosis (MS) study participants. METHODS The single-center, first-in-human, single ascending dose (SAD) study evaluated elezanumab (50-1,600mg intravenous [IV] and 150mg subcutaneous) in 47 healthy men and women. The multicenter multiple ascending dose (MAD; NCT02601885) study evaluated elezanumab (150mg, 600mg, and 1,800mg) in 20 adult men and women with MS, receiving either maintenance or no immunomodulatory treatment. RESULTS No pattern of study drug-related adverse events was identified for either the SAD or MAD elezanumab regimens. Across both studies, the Tmax occurred within 4 hours of elezanumab IV infusion, and the harmonic mean of t1/2 ranged between 18.6 and 67.7 days. Following multiple dosing, elezanumab Cmax , area under the curve, and Ctrough increased dose-proportionally and resulted in dose-dependent increases in elezanumab cerebrospinal fluid (CSF) concentrations. Elezanumab CSF penetration was 0.1% to 0.4% across both studies, with CSF levels of free RGMa decreased by >40%. Changes in CSF interleukin-10 (IL-10) and free RGMa demonstrated dose/exposure-dependence. INTERPRETATION The elezanumab pharmacokinetic profile supports monthly, or bimonthly, administration of up to 1,800mg with the option of a loading dose of 3,600mg. Elezanumab partitioning into CSF is within the range expected for mAbs. Reduced CSF levels of free RGMa demonstrate central nervous system target binding of elezanumab with an apparent maximal effect at 1,800mg IV. Exposure-associated increases in CSF IL-10, an anti-inflammatory cytokine with neuroprotective/neurorestorative properties, support potential pathway modulation in MS participants. ANN NEUROL 2023;93:285-296.
Collapse
Affiliation(s)
- Hari V Kalluri
- Clinical Pharmacology and Pharmacometrics, AbbVie, North Chicago, IL
| | | | | | | | - Wei Liu
- Clinical Pharmacology and Pharmacometrics, AbbVie, North Chicago, IL
| | - Bruce A C Cree
- Weill Institute of Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
37
|
de Mol CL, van Luijn MM, Kreft KL, Looman KIM, van Zelm MC, White T, Moll HA, Smolders J, Neuteboom RF. Multiple sclerosis risk variants influence the peripheral B-cell compartment early in life in the general population. Eur J Neurol 2023; 30:434-442. [PMID: 36169606 PMCID: PMC10092523 DOI: 10.1111/ene.15582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 07/09/2022] [Accepted: 09/23/2022] [Indexed: 01/07/2023]
Abstract
BACKGROUND AND PURPOSE Multiple sclerosis (MS) is associated with abnormal B-cell function, and MS genetic risk alleles affect multiple genes that are expressed in B cells. However, how these genetic variants impact the B-cell compartment in early childhood is unclear. In the current study, we aim to assess whether polygenic risk scores (PRSs) for MS are associated with changes in the blood B-cell compartment in children from the general population. METHODS Six-year-old children from the population-based Generation R Study were included. Genotype data were used to calculate MS-PRSs and B-cell subset-enriched MS-PRSs, established by designating risk loci based on expression and function. Analyses of variance were performed to examine the effect of MS-PRSs on total B-cell numbers (n = 1261) as well as naive and memory subsets (n = 675). RESULTS After correction for multiple testing, no significant associations were observed between MS-PRSs and total B-cell numbers and frequencies of subsets therein. A naive B-cell-MS-PRS (n = 26 variants) was significantly associated with lower relative, but not absolute, naive B-cell numbers (p = 1.03 × 10-4 and p = 0.82, respectively), and higher frequencies and absolute numbers of CD27+ memory B cells (p = 8.83 × 10-4 and p = 4.89 × 10-3 , respectively). These associations remained significant after adjustment for Epstein-Barr virus seropositivity and the HLA-DRB1*15:01 genotype. CONCLUSIONS The composition of the blood B-cell compartment is associated with specific naive B-cell-associated MS risk variants during childhood, possibly contributing to MS pathophysiology later in life. Cell subset-specific PRSs may offer a more sensitive tool to define the impact of genetic risk on the immune system in diseases such as MS.
Collapse
Affiliation(s)
- Casper L de Mol
- Department of Neurology, Erasmus University Medical Center Rotterdam, Rotterdam, the Netherlands
- Generation R Study Group, Erasmus University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Marvin M van Luijn
- Department of Immunology, Erasmus University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Karim L Kreft
- Department of Neurology, Erasmus University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Kirsten I M Looman
- Generation R Study Group, Erasmus University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Pediatrics, Erasmus University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Menno C van Zelm
- Department of Immunology and Pathology, Central Clinical School, Monash University and Alfred Hospital, Melbourne, Victoria, Australia
| | - Tonya White
- Department of Child and Adolescent Psychiatry, Erasmus University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Henriette A Moll
- Generation R Study Group, Erasmus University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Immunology, Erasmus University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Joost Smolders
- Department of Neurology, Erasmus University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Immunology, Erasmus University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Rinze F Neuteboom
- Department of Neurology, Erasmus University Medical Center Rotterdam, Rotterdam, the Netherlands
| |
Collapse
|
38
|
Xu D, Li C, Xu Y, Huang M, Cui D, Xie J. Myeloid-derived suppressor cell: A crucial player in autoimmune diseases. Front Immunol 2022; 13:1021612. [PMID: 36569895 PMCID: PMC9780445 DOI: 10.3389/fimmu.2022.1021612] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 11/24/2022] [Indexed: 12/13/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are identified as a highly heterogeneous group of immature cells derived from bone marrow and play critical immunosuppressive functions in autoimmune diseases. Accumulating evidence indicates that the pathophysiology of autoimmune diseases was closely related to genetic mutations and epigenetic modifications, with the latter more common. Epigenetic modifications, which involve DNA methylation, covalent histone modification, and non-coding RNA-mediated regulation, refer to inheritable and potentially reversible changes in DNA and chromatin that regulate gene expression without altering the DNA sequence. Recently, numerous reports have shown that epigenetic modifications in MDSCs play important roles in the differentiation and development of MDSCs and their suppressive functions. The molecular mechanisms of differentiation and development of MDSCs and their regulatory roles in the initiation and progression of autoimmune diseases have been extensively studied, but the exact function of MDSCs remains controversial. Therefore, the biological and epigenetic regulation of MDSCs in autoimmune diseases still needs to be further characterized. This review provides a detailed summary of the current research on the regulatory roles of DNA methylation, histone modifications, and non-coding RNAs in the development and immunosuppressive activity of MDSCs, and further summarizes the distinct role of MDSCs in the pathogenesis of autoimmune diseases, in order to provide help for the diagnosis and treatment of diseases from the perspective of epigenetic regulation of MDSCs.
Collapse
Affiliation(s)
- Dandan Xu
- Department of Blood Transfusion, The First Affiliated Hospital, School of Medicine, Hangzhou, Zhejiang University, China
| | - Cheng Li
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Yushan Xu
- Department of Blood Transfusion, The First Affiliated Hospital, School of Medicine, Hangzhou, Zhejiang University, China
| | - Mingyue Huang
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Dawei Cui
- Department of Blood Transfusion, The First Affiliated Hospital, School of Medicine, Hangzhou, Zhejiang University, China,*Correspondence: Dawei Cui, ; Jue Xie,
| | - Jue Xie
- Department of Blood Transfusion, The First Affiliated Hospital, School of Medicine, Hangzhou, Zhejiang University, China,*Correspondence: Dawei Cui, ; Jue Xie,
| |
Collapse
|
39
|
Govindarajulu M, Ramesh S, Shankar T, Kora MK, Moore T, Dhanasekaran M. Role of Neddylation in Neurodegenerative Diseases. NEUROSCI 2022; 3:533-545. [PMID: 39483771 PMCID: PMC11523694 DOI: 10.3390/neurosci3040038] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 09/26/2022] [Indexed: 11/03/2024] Open
Abstract
Neurodegenerative diseases are characterized by progressive loss of neurons in specific regions of the brain. Neuronal death is often associated with the accumulation of misfolded proteins due to genetic mutations or abnormal protein homeostasis. An essential mechanism for regulating the clearance of misfolded proteins is neddylation, a post-translational modification closely related to ubiquitination. Neddylation is brought about by conjugating neural precursor cell-expressed developmentally downregulated protein 8 (NEDD8) to target substrates through a cascade of cellular events. Neddylation is crucial for many biological processes, and dysfunctional neddylation is implicated in several neurodegenerative diseases. This review discusses the current understanding of the role of neddylation pathways in neurodegenerative disorders and the emergence of neddylation signaling as a potential target for drug discovery and development in neurodegenerative diseases.
Collapse
Affiliation(s)
- Manoj Govindarajulu
- Department of Drug Discovery and Development, Auburn University Harrison School of Pharmacy, Auburn, AL 36849, USA
| | - Sindhu Ramesh
- Department of Drug Discovery and Development, Auburn University Harrison School of Pharmacy, Auburn, AL 36849, USA
| | - Tharanth Shankar
- Department of Internal Medicine, Ramaiah Medical College and Hospital, Bengaluru 560054, Karnataka, India
| | | | - Timothy Moore
- Department of Drug Discovery and Development, Auburn University Harrison School of Pharmacy, Auburn, AL 36849, USA
| | - Muralikrishnan Dhanasekaran
- Department of Drug Discovery and Development, Auburn University Harrison School of Pharmacy, Auburn, AL 36849, USA
| |
Collapse
|
40
|
Mesenchymal stem cell therapy: A review of clinical trials for multiple sclerosis. Regen Ther 2022; 21:201-209. [PMID: 36092509 PMCID: PMC9420954 DOI: 10.1016/j.reth.2022.07.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/02/2022] [Accepted: 07/15/2022] [Indexed: 11/22/2022] Open
Abstract
Multiple sclerosis (MS) is a disease of the central nervous system (CNS) that is the result of the body's own immune cells being auto-reactive to the myelin regions of the body as if these regions were foreign antigens. This demyelination process is damaging to the electrical conductivity of neurons. The current medicines are only capable of fighting off the symptoms of the disease, but not the disease itself. Specialized stem cells, known as mesenchymal stem cells (MSCs), seem to be the candidate therapy to get rid of MS. MSCs can be isolated from multiple sources of the person's body, and even from the umbilical cord (UC) and placenta of a donor. These cells have anti-inflammatory effects so they can target the overactivity and self-antigen attacks by T cells and macrophages; this immune system overactivity is characteristic of MS. MSCs show the ability to locate into brain lesions when injected and thus can compensate for the loss of the brain function by differentiating into neuronal precursor cells and glial cells. The author has listed tables of clinical trials that have utilized MSCs from different sources, along with the years and the phase of study completed for each trial. The consensus is that these cells work on inhibiting CD4+ and CD8+ T cell activation, T regulatory cells (Tregs), and macrophage switch into the auto-immune phenotype. The best source of MSCs seems to be the UC due to the easiness of extraction, the noninvasive method of collection, their higher expansion ability and more powerful immune-modulating properties compared to other locations in the body. Studies showed there was a significant decline of mRNA expression of several cytokines after the administration of MSCs derived from the UC (UCMSCs). Other researchers were able to repair the defects of Tregs in MS patients by co-culturing Tregs from these patients with UCMSCs, which decreased the production of the pro-inflammatory cytokine IFN γ, and also suggested a strong link between Tregs lack of functionality in MS patients with the pathogenesis of the disease.
Collapse
|
41
|
Kim J, Bollaert RE, Cerna J, Adamson BC, Robbs CM, Khan NA, Motl RW. Moderate-to-Vigorous Physical Activity is Related With Retinal Neuronal and Axonal Integrity in Persons With Multiple Sclerosis. Neurorehabil Neural Repair 2022; 36:810-815. [PMID: 36317869 DOI: 10.1177/15459683221131787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Moderate-to-vigorous physical activity (MVPA) may confer benefits for axonal and/or neuronal integrity in adults with multiple sclerosis (MS). PURPOSE Examine the association between device-measured MVPA with optical coherence tomography (OCT) metrics of retinal nerve fiber layer (RNFL) thickness and total macular volume (TMV) in persons with and without MS. METHODS Adults with MS (N = 41), along with sex-matched healthy control (HC) participants (N = 79), underwent measurements of retinal morphology via OCT and wore an accelerometer for a period of 7 days as a measure of MVPA. RESULTS Persons with MS had significantly lower MVPA, RNFL thickness, and TMV compared with HCs. MVPA was correlated with RNFL (r = .38, P < .01) thickness and TMV (r = .49, P < .01). Hierarchical linear regression analyses indicated that addition of MVPA attenuated the Group effect on RNFL and TMV. MVPA accounted for 8% and 3% of the variance in TMV (β = .343, P < .01) and RNFL thickness (β = .217, P = .03), respectively. CONCLUSION MVPA was positively associated with axonal and neuronal integrity assessed by OCT and partially explained group differences in those metrics. These results present possible future targets for MS management by increasing MVPA.
Collapse
Affiliation(s)
- Jeongwoon Kim
- Department of Kinesiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Rachel E Bollaert
- Program in Exercise Science, Marquette University, Milwaukee, WI, USA
| | - Jonathan Cerna
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Brynn C Adamson
- Department of Health Sciences, University of Colorado, Colorado Springs, CO, USA
| | | | - Naiman A Khan
- Department of Kinesiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA.,Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA.,Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Robert W Motl
- Department of Kinesiology and Nutrition, University of Illinois Chicago, Chicago, IL, USA
| |
Collapse
|
42
|
Berrigan LI, LeFevre JA, Rees LM, Berard JA, Francis A, Freedman MS, Walker LAS. The symbol digit modalities test and the paced auditory serial addition test involve more than processing speed. Mult Scler Relat Disord 2022; 68:104229. [PMID: 36279599 DOI: 10.1016/j.msard.2022.104229] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 09/17/2022] [Accepted: 10/06/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND Slowed processing speed is the most frequently reported cognitive deficit for people with multiple sclerosis (MS). However, measures used to assess processing speed may also recruit other cognitive abilities. The present objective was to determine the contributions of different cognitive functions to performance on two commonly used processing speed measures: the Symbol Digit Modalities Test (SDMT) and the Paced Auditory Serial Addition Test (PASAT). METHODS Adults with relapsing-remitting MS (n = 70) and controls (n = 72) completed the SDMT, PASAT, and multiple measures assessing processing speed, working memory, and learning. Hierarchical regression analyses were used to examine the contributions of MS, processing speed, working memory, learning, and all possible interactions among factors to SDMT and PASAT scores. RESULTS Processing speed and working memory generally contributed to performance on the SDMT and PASAT, with learning additionally contributing to SDMT performance. However, significant interactions revealed processing speed did not influence PASAT performance for individuals with high working memory ability whereas processing speed became increasingly more important as working memory declined to average and low levels. Further, processing speed was associated with SDMT performance for patients with MS but not controls. CONCLUSIONS These findings support a multifactorial interpretation of the SDMT and PASAT, which facilitates their usefulness as screening measures for cognitive decline but prevents them from identifying which specific cognitive functions are affected.
Collapse
Affiliation(s)
- Lindsay I Berrigan
- Departments of Psychology and Biology, St. Francis Xavier University, Antigonish, NS, Canada; Department of Psychiatry, Dalhousie University, Halifax, NS, Canada.
| | - Jo-Anne LeFevre
- Departments of Cognitive Science and Psychology, Carleton University, Ottawa, ON, Canada
| | - Laura M Rees
- School of Psychology, University of Ottawa, ON, Canada; Neuropsychology Service, The Ottawa Hospital, Ottawa, ON, Canada; The Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Jason A Berard
- The Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Ashley Francis
- Department of Psychiatry, Dalhousie University, Halifax, NS, Canada
| | - Mark S Freedman
- The Ottawa Hospital Research Institute, Ottawa, ON, Canada; Department of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Lisa A S Walker
- School of Psychology, University of Ottawa, ON, Canada; Neuropsychology Service, The Ottawa Hospital, Ottawa, ON, Canada; The Ottawa Hospital Research Institute, Ottawa, ON, Canada; Department of Medicine, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
43
|
Montañés‐Masias B, Bort‐Roig J, Pascual JC, Soler J, Briones‐Buixassa L. Online psychological interventions to improve symptoms in multiple sclerosis: A systematic review: Online psychological interventions in Multiple Sclerosis. Acta Neurol Scand 2022; 146:448-464. [PMID: 36121184 PMCID: PMC9825977 DOI: 10.1111/ane.13709] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 08/22/2022] [Indexed: 01/11/2023]
Abstract
The aim of this systematic review was to assess the effectiveness of Internet-based psychological interventions in the treatment of physical, socio-affective and cognitive symptoms and quality of life (QoL) in people with multiple sclerosis (pwMS) to provide currently available evidence. Systematic searches for eligible studies were carried out in four databases (August 2021) using key words. Studies were screened, data extracted, quality appraised and analysed by three independent reviewers, using predefined criteria and following the PRISMA rules. Study quality was assessed using Standard Quality Assessment Criteria for Evaluating Primary Research Papers from a Variety of Fields QUALSYST tool. Physical, socio-affective and cognitive symptoms and QoL were the primary outcomes. Thirteen studies were included. Two principal approaches were reported: Cognitive behavioural therapy (CBT) and mindfulness-based interventions (MBI). Interventions varied from tailored versions to videoconference by a clinician, duration mean 8 weeks, delivered via individually and groups, all online. The review found that iCBT interventions were effective for improve depression, anxiety, fatigue and QoL, and slightly in cognitive functioning in pwMS, whereas MBI interventions reported benefits in depression, anxiety, stress and QoL, and less evidence in fatigue. Generally, study quality was acceptable in most studies; eleven of the studies scored a low risk of bias on all items in the Qualsyst Tool, whereas only two studies were considered unacceptable. Psychological online interventions may improve physical, socio-affective and cognitive symptoms as well as QoL in pwMS, overcoming the face-to-face barriers (i.e. disability). Contact with the therapist and groups sessions have been identified as enablers of the online interventions. Nevertheless, the limited number of studies and the heterogeneity of health outcomes reported made difficult to afford robust conclusions on psychological intervention effects in pwMS.
Collapse
Affiliation(s)
- Brenda Montañés‐Masias
- Research group in Mental Health and Social Innovation (SaMIS), Centre for Health and Social Care ResearchUniversitat de Vic – Universitat Central de CatalunyaVicCataloniaSpain
| | - Judit Bort‐Roig
- Sport and Physical Activity Research Group, Centre for Health and Social Care ResearchUniversitat de Vic – Universitat Central de CatalunyaVicCataloniaSpain
| | - Juan Carlos Pascual
- Department of PsychiatryHospital de la Santa Creu i Sant PauBarcelonaSpain,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM)Institut de Recerca Biomèdica Sant Pau (IIB‐Sant Pau)BarcelonaSpain,Department of Psychiatry and Forensic Medicine & Institute of NeurosciencesUniversitat Autònoma de BarcelonaBellaterraSpain
| | - Joaquim Soler
- Department of PsychiatryHospital de la Santa Creu i Sant PauBarcelonaSpain,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM)Institut de Recerca Biomèdica Sant Pau (IIB‐Sant Pau)BarcelonaSpain,Department of Psychiatry and Forensic Medicine & Institute of NeurosciencesUniversitat Autònoma de BarcelonaBellaterraSpain
| | - Laia Briones‐Buixassa
- Research group in Mental Health and Social Innovation (SaMIS), Centre for Health and Social Care ResearchUniversitat de Vic – Universitat Central de CatalunyaVicCataloniaSpain
| |
Collapse
|
44
|
Arpan I, Shah VV, McNames J, Harker G, Carlson-Kuhta P, Spain R, El-Gohary M, Mancini M, Horak FB. Fall Prediction Based on Instrumented Measures of Gait and Turning in Daily Life in People with Multiple Sclerosis. SENSORS (BASEL, SWITZERLAND) 2022; 22:5940. [PMID: 36015700 PMCID: PMC9415310 DOI: 10.3390/s22165940] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 08/02/2022] [Accepted: 08/04/2022] [Indexed: 06/15/2023]
Abstract
This study investigates the potential of passive monitoring of gait and turning in daily life in people with multiple sclerosis (PwMS) to identify those at future risk of falls. Seven days of passive monitoring of gait and turning were carried out in a pilot study of 26 PwMS in home settings using wearable inertial sensors. The retrospective fall history was collected at the baseline. After gait and turning data collection in daily life, PwMS were followed biweekly for a year and were classified as fallers if they experienced >1 fall. The ability of short-term passive monitoring of gait and turning, as well as retrospective fall history to predict future falls were compared using receiver operator curves and regression analysis. The history of retrospective falls was not identified as a significant predictor of future falls in this cohort (AUC = 0.62, p = 0.32). Among quantitative monitoring measures of gait and turning, the pitch at toe-off was the best predictor of falls (AUC = 0.86, p < 0.01). Fallers had a smaller pitch of their feet at toe-off, reflecting less plantarflexion during the push-off phase of walking, which can impact forward propulsion and swing initiation and can result in poor foot clearance and an increased metabolic cost of walking. In conclusion, our cohort of PwMS showed that objective monitoring of gait and turning in daily life can identify those at future risk of falls, and the pitch at toe-off was the single most influential predictor of future falls. Therefore, interventions aimed at improving the strength of plantarflexion muscles, range of motion, and increased proprioceptive input may benefit PwMS at future fall risk.
Collapse
Affiliation(s)
- Ishu Arpan
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA
- Advanced Imaging Research Center, Oregon Health & Science University Portland, OR 97239, USA
| | - Vrutangkumar V. Shah
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA
- APDM Wearable Technologies-A Clario Company, 2828 S Corbett Ave, Ste 135, Portland, OR 97201, USA
| | - James McNames
- APDM Wearable Technologies-A Clario Company, 2828 S Corbett Ave, Ste 135, Portland, OR 97201, USA
- Department of Electrical and Computer Engineering, Portland State University, 1825 SW Broadway, Portland, OR 97201, USA
| | - Graham Harker
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA
| | | | - Rebecca Spain
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Mahmoud El-Gohary
- APDM Wearable Technologies-A Clario Company, 2828 S Corbett Ave, Ste 135, Portland, OR 97201, USA
| | - Martina Mancini
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Fay B. Horak
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA
- APDM Wearable Technologies-A Clario Company, 2828 S Corbett Ave, Ste 135, Portland, OR 97201, USA
| |
Collapse
|
45
|
Mehdi-alamdarlou S, Ahmadi F, Azadi A, Shahbazi MA, Heidari R, Ashrafi H. A cell-mimicking platelet-based drug delivery system as a potential carrier of dimethyl fumarate for multiple sclerosis. Int J Pharm 2022; 625:122084. [DOI: 10.1016/j.ijpharm.2022.122084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/20/2022] [Accepted: 08/03/2022] [Indexed: 11/25/2022]
|
46
|
Sulimai NH, Brown J, Lominadze D. Fibrinogen, Fibrinogen-like 1 and Fibrinogen-like 2 Proteins, and Their Effects. Biomedicines 2022; 10:1712. [PMID: 35885017 PMCID: PMC9313381 DOI: 10.3390/biomedicines10071712] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/11/2022] [Accepted: 07/13/2022] [Indexed: 12/05/2022] Open
Abstract
Fibrinogen (Fg) and its derivatives play a considerable role in many diseases. For example, increased levels of Fg have been found in many inflammatory diseases, such as Alzheimer's disease, multiple sclerosis, traumatic brain injury, rheumatoid arthritis, systemic lupus erythematosus, and cancer. Although associations of Fg, Fg chains, and its derivatives with various diseases have been established, their specific effects and the mechanisms of actions involved are still unclear. The present review is the first attempt to discuss the role of Fg, Fg chains, its derivatives, and other members of Fg family proteins, such as Fg-like protein 1 and 2, in inflammatory diseases and their effects in immunomodulation.
Collapse
Affiliation(s)
- Nurul H. Sulimai
- Departments of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA; (N.H.S.); (J.B.)
| | - Jason Brown
- Departments of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA; (N.H.S.); (J.B.)
| | - David Lominadze
- Departments of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA; (N.H.S.); (J.B.)
- Departments of Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA
| |
Collapse
|
47
|
Bashirzade AA, Zabegalov KN, Volgin AD, Belova AS, Demin KA, de Abreu MS, Babchenko VY, Bashirzade KA, Yenkoyan KB, Tikhonova MA, Amstislavskaya TG, Kalueff AV. Modeling neurodegenerative disorders in zebrafish. Neurosci Biobehav Rev 2022; 138:104679. [PMID: 35490912 DOI: 10.1016/j.neubiorev.2022.104679] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 04/11/2022] [Accepted: 04/24/2022] [Indexed: 12/15/2022]
Abstract
Neurodegeneration is a major cause of Alzheimer's, Parkinson's, Huntington's, multiple and amyotrophic lateral sclerosis, pontocerebellar hypoplasia, dementia and other related brain disorders. Their complex pathogenesis commonly includes genetic and neurochemical deficits, misfolded protein toxicity, demyelination, apoptosis and mitochondrial dysfunctions. Albeit differing in specific underlying mechanisms, neurodegenerative disorders typically display evolutionarily conserved mechanisms across taxa. Here, we review the role of zebrafish models in recapitulating major human and rodent neurodegenerative conditions, demonstrating this species as a highly relevant experimental model for research on neurodegenerative diseases, and discussing how these fish models can further clarify the underlying genetic, neurochemical, neuroanatomical and behavioral pathogenic mechanisms.
Collapse
Affiliation(s)
- Alim A Bashirzade
- Novosibirsk State University, Institute of Medicine and Psychology, Novosibirsk, Russia; Scientific Research Institute of Neuroscience and Medicine, Novosibirsk, Russia
| | | | - Andrey D Volgin
- Novosibirsk State University, Institute of Medicine and Psychology, Novosibirsk, Russia; Scientific Research Institute of Neuroscience and Medicine, Novosibirsk, Russia
| | - Alisa S Belova
- Novosibirsk State University, Institute of Medicine and Psychology, Novosibirsk, Russia; Scientific Research Institute of Neuroscience and Medicine, Novosibirsk, Russia
| | - Konstantin A Demin
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia; Granov Scientific Research Center of Radiology and Surgical Technologies, St. Petersburg, Russia; Almazov Medical Research Center, St. Petersburg, Russia
| | | | - Vladislav Ya Babchenko
- Novosibirsk State University, Institute of Medicine and Psychology, Novosibirsk, Russia; Scientific Research Institute of Neuroscience and Medicine, Novosibirsk, Russia
| | - Kseniya A Bashirzade
- Novosibirsk State University, Institute of Medicine and Psychology, Novosibirsk, Russia
| | - Konstantin B Yenkoyan
- Neuroscience Laboratory, COBRAIN Center, M Heratsi Yerevan State Medical University, Yerevan, Armenia; COBRAIN Center - Scientific Educational Center for Fundamental Brain Research, Yerevan, Armenia
| | - Maria A Tikhonova
- Novosibirsk State University, Institute of Medicine and Psychology, Novosibirsk, Russia; Scientific Research Institute of Neuroscience and Medicine, Novosibirsk, Russia
| | - Tamara G Amstislavskaya
- Novosibirsk State University, Institute of Medicine and Psychology, Novosibirsk, Russia; Scientific Research Institute of Neuroscience and Medicine, Novosibirsk, Russia
| | - Allan V Kalueff
- The Russian Academy of Sciences, Moscow, Russia; Ural Federal University, Yekaterinburg, Russia; COBRAIN Center - Scientific Educational Center for Fundamental Brain Research, Yerevan, Armenia.
| |
Collapse
|
48
|
de Mol CL, Neuteboom RF, Jansen PR, White T. White matter microstructural differences in children and genetic risk for multiple sclerosis: A population-based study. Mult Scler 2022; 28:730-741. [PMID: 34379023 PMCID: PMC8978478 DOI: 10.1177/13524585211034826] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 06/17/2021] [Accepted: 07/06/2021] [Indexed: 11/25/2022]
Abstract
BACKGROUND MS patients show abnormalities in white matter (WM) on brain imaging, with heterogeneity in the location of WM lesions. The "pothole" method can be applied to diffusion-weighted images to identify spatially distinct clusters of divergent brain WM microstructure. OBJECTIVE To investigate the association between genetic risk for MS and spatially independent clusters of decreased or increased fractional anisotropy (FA) in the brain. In addition, we studied sex- and age-related differences. METHODS 3 Tesla diffusion tensor imaging (DTI) data were collected in 8- to 12-year-old children from a population-based study. Global and tract-based potholes (lower FA clusters) and molehills (higher FA clusters) were quantified in 3047 participants with usable DTI data. A polygenic risk score (PRS) for MS was calculated in genotyped individuals (n = 1087) and linear regression analyses assessed the relationship between the PRS and the number of potholes and molehills, correcting for multiple testing using the False Discovery Rate. RESULTS The number of molehills increased with age, potholes decreased with age, and fewer potholes were observed in girls during typical development. The MS-PRS was positively associated with the number of molehills (β = 0.9, SE = 0.29, p = 0.002). Molehills were found more often in the corpus callosum (β = 0.3, SE = 0.09, p = 0.0003). CONCLUSION Genetic risk for MS is associated with spatially distinct clusters of increased FA during childhood brain development.
Collapse
Affiliation(s)
- C Louk de Mol
- Department of Neurology, MS Center ErasMS,
Erasmus MC University Medical Center Rotterdam, Rotterdam, The
Netherlands/The Generation R Study Group, Erasmus MC University Medical
Center Rotterdam, Rotterdam, The Netherlands
| | - Rinze F Neuteboom
- Department of Neurology, MS Center ErasMS,
Erasmus MC University Medical Center Rotterdam, Rotterdam, The
Netherlands
| | - Philip R Jansen
- The Generation R Study Group, Erasmus MC
University Medical Center Rotterdam, Rotterdam, The Netherlands/Department
of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research,
Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands/Department
of Clinical Genetics, Amsterdam UMC, Vrije Universiteit Amsterdam,
Amsterdam, The Netherlands
| | - Tonya White
- Department of Child and Adolescent Psychiatry,
Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands;
Department of Radiology and Nuclear Medicine, Erasmus MC University Medical
Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
49
|
Differences in MS clinical and epidemiological characteristics between Ashkenazi and non-Ashkenazi Jewish patients in Israel: a retrospective single center study. Sci Rep 2022; 12:4555. [PMID: 35296756 PMCID: PMC8927451 DOI: 10.1038/s41598-022-08565-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 03/08/2022] [Indexed: 11/08/2022] Open
Abstract
The prevalence and severity of Multiple Sclerosis (MS) varies across different ethnicities, with a tendency to a more severe phenotype in non-Caucasian populations. Our objective was to evaluate the differences in disease phenotype between Ashkenazi Jewish and Non-Ashkenazi Jewish patients in Israel. We conducted a single center retrospective cohort study in which subjects were assigned to Ashkenazi or Non-Ashkenazi groups according to self-reported ancestry and disease severity was assessed using the expanded disability status (EDSS), MS severity score (MSSS), progression index (PI) and MRI metrics. 330 Ashkenazi Jewish (AJ) and 207 Non-Ashkenazi Jewish patients (Non-AJ) were included. Non-AJ had a younger age of disease onset (32.7 years vs. 35.7 years, p = 0.05), with a lower proportion of females (62.3% vs. 73.3%, p = 0.01). These differences were maintained within the subgroup of Israeli native patients. Ethnicity was a significant predictor of MSSS (β = 0.601, p = 0.003), with a higher estimate than that of other epidemiological factors. To conclude, Non-AJ patients had an earlier age of onset and a more disabling disease as well as having a more balanced female to male ratio compared to AJ patients. These findings demonstrate variability of disease phenotype within Caucasian patient's dependent on their ethnicity despite equivalent access to healthcare services.
Collapse
|
50
|
Garlı M, Kurna SA, Alış A, Akın Çakır E, Yükselen NP, Açıkalın B. Evaluation of peripapillary and subfoveal choroidal vascularity index in patients with multiple sclerosis. Photodiagnosis Photodyn Ther 2022; 38:102810. [PMID: 35304309 DOI: 10.1016/j.pdpdt.2022.102810] [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: 12/13/2021] [Revised: 02/02/2022] [Accepted: 03/14/2022] [Indexed: 11/18/2022]
Abstract
PURPOSE To evaluate the changes in the peripapillary choroidal vascularity index (PCVI) and subfoveal choroidal vascularity index (SFCVI) in multiple sclerosis (MS) patients and healthy subjects METHODS: A total of 145 eyes of 73 patients were investigated in this cross-sectional study. 78 eyes of 39 MS patients (Group 1) and 67 eyes of 34 healthy subjects (Group 2) were evaluated. MS patients with a history of optic neuritis (ON) constituted Group 1a, those without a history of ON constituted Group 1b. RESULTS The mean PCVI was significantly lower in Group 1 than Group 2 (61,39±3,00 % vs 64,49±2,29 % respectively, p<0.001). The mean SFCVI scores of Group 1 was significantly lower than Group 2 (64,01±2,66 % vs. 66,87±2,14 % respectively, p<0.001). The mean PCVI of Group 1a (59,26±2,85 %) was significantly lower compared to Group 1b (62,87±2,08 %) and Group 2 (64,49±2,29 %, p1<0.001, p2<0.001). The mean SFCVI of Group 1a was significantly lower than Group 2 (64.26±2.75 % vs. 66.87±2.14 % respectively, p<0.001). CONCLUSION PCVI and SFCVI scores were significantly lower in MS patients compared to healthy controls. PCVI scores of MS patients who had a history of ON were significantly lower than those of patients without a previous ON attack, as were SFCVI scores. We consider that evaluation of PCVI and SFCVI might be useful for monitoring ocular involvement in patients with MS.
Collapse
Affiliation(s)
- Murat Garlı
- Department of Ophthalmology, Fatih Sultan Mehmet Training and Research Hospital, University of Health Sciences, Istanbul 34752, Turkey.
| | - Sevda Aydın Kurna
- Department of Ophthalmology, Fatih Sultan Mehmet Training and Research Hospital, University of Health Sciences, Istanbul 34752, Turkey
| | - Abdülkadir Alış
- Department of Ophthalmology, Fatih Sultan Mehmet Training and Research Hospital, University of Health Sciences, Istanbul 34752, Turkey
| | - Esra Akın Çakır
- Department of Ophthalmology, Fatih Sultan Mehmet Training and Research Hospital, University of Health Sciences, Istanbul 34752, Turkey
| | - Nihan Parasız Yükselen
- Department of Neurology, Fatih Sultan Mehmet Training and Research Hospital, University of Health Sciences, Istanbul 34752, Turkey
| | - Banu Açıkalın
- Department of Ophthalmology, Fatih Sultan Mehmet Training and Research Hospital, University of Health Sciences, Istanbul 34752, Turkey
| |
Collapse
|