1
|
Sanabria-Diaz G, Cagol A, Lu PJ, Barakovic M, Ocampo-Pineda M, Chen X, Weigel M, Ruberte E, Siebenborn NDOS, Galbusera R, Schädelin S, Benkert P, Kuhle J, Kappos L, Melie-Garcia L, Granziera C. Advanced MRI Measures of Myelin and Axon Volume Identify Repair in Multiple Sclerosis. Ann Neurol 2024. [PMID: 39390658 DOI: 10.1002/ana.27102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 08/10/2024] [Accepted: 09/04/2024] [Indexed: 10/12/2024]
Abstract
OBJECTIVE Pathological studies suggest that multiple sclerosis (MS) lesions endure multiple waves of damage and repair; however, the dynamics and characteristics of these processes are poorly understood in patients living with MS. METHODS We studied 128 MS patients (75 relapsing-remitting, 53 progressive) and 72 healthy controls who underwent advanced magnetic resonance imaging and clinical examination at baseline and 2 years later. Magnetization transfer saturation and multi-shell diffusion imaging were used to quantify longitudinal changes in myelin and axon volumes within MS lesions. Lesions were grouped into 4 classes (repair, damage, mixed repair damage, and stable). The frequency of each class was correlated to clinical measures, demographic characteristics, and levels of serum neurofilament light chain (sNfL). RESULTS Stable lesions were the most frequent (n = 2,276; 44%), followed by lesions with patterns of "repair" (n = 1,352; 26.2%) and damage (n = 1,214; 23.5%). The frequency of "repair" lesion was negatively associated with disability (β = -0.04; p < 0.001) and sNfL (β = -0.16; p < 0.001) at follow-up. The frequency of the "damage" class was higher in progressive than relapsing-remitting patients (p < 0.05) and was related to disability (baseline: β = -0.078; follow-up: β = -0.076; p < 0.001) and age (baseline: β = -0.078; p < 0.001). Stable lesions were more frequent in relapsing-remitting than in progressive patients (p < 0.05), and in younger patients versus older (β = -0.07; p < 0.001) at baseline. Further, "mixed" lesions were most frequent in older patients (β = 0.004; p < 0.001) at baseline. INTERPRETATION These findings show that repair and damage processes within MS lesions occur across the entire disease spectrum and that their frequency correlates with patients disability, age, disease duration, and extent of neuroaxonal damage. ANN NEUROL 2024.
Collapse
Affiliation(s)
- Gretel Sanabria-Diaz
- Neurology Clinic and Policlinic, Department of Medicine, Clinical Research and Biomedical Engineering, University Hospital Basel and University of Basel, Basel, Switzerland
- Translational Imaging in Neurology (ThINk) Basel, Department of Biomedical Engineering, University Hospital Basel and University of Basel, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel, Switzerland
| | - Alessandro Cagol
- Neurology Clinic and Policlinic, Department of Medicine, Clinical Research and Biomedical Engineering, University Hospital Basel and University of Basel, Basel, Switzerland
- Translational Imaging in Neurology (ThINk) Basel, Department of Biomedical Engineering, University Hospital Basel and University of Basel, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel, Switzerland
- Department of Health Sciences, University of Genova, Genoa, Italy
| | - Po-Jui Lu
- Neurology Clinic and Policlinic, Department of Medicine, Clinical Research and Biomedical Engineering, University Hospital Basel and University of Basel, Basel, Switzerland
- Translational Imaging in Neurology (ThINk) Basel, Department of Biomedical Engineering, University Hospital Basel and University of Basel, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel, Switzerland
| | - Muhamed Barakovic
- Neurology Clinic and Policlinic, Department of Medicine, Clinical Research and Biomedical Engineering, University Hospital Basel and University of Basel, Basel, Switzerland
- Translational Imaging in Neurology (ThINk) Basel, Department of Biomedical Engineering, University Hospital Basel and University of Basel, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel, Switzerland
| | - Mario Ocampo-Pineda
- Neurology Clinic and Policlinic, Department of Medicine, Clinical Research and Biomedical Engineering, University Hospital Basel and University of Basel, Basel, Switzerland
- Translational Imaging in Neurology (ThINk) Basel, Department of Biomedical Engineering, University Hospital Basel and University of Basel, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel, Switzerland
| | - Xinjie Chen
- Neurology Clinic and Policlinic, Department of Medicine, Clinical Research and Biomedical Engineering, University Hospital Basel and University of Basel, Basel, Switzerland
- Translational Imaging in Neurology (ThINk) Basel, Department of Biomedical Engineering, University Hospital Basel and University of Basel, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel, Switzerland
| | - Matthias Weigel
- Neurology Clinic and Policlinic, Department of Medicine, Clinical Research and Biomedical Engineering, University Hospital Basel and University of Basel, Basel, Switzerland
- Translational Imaging in Neurology (ThINk) Basel, Department of Biomedical Engineering, University Hospital Basel and University of Basel, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel, Switzerland
- Division of Radiological Physics, Department of Radiology, University Hospital Basel, Basel, Switzerland
| | - Esther Ruberte
- Neurology Clinic and Policlinic, Department of Medicine, Clinical Research and Biomedical Engineering, University Hospital Basel and University of Basel, Basel, Switzerland
- Translational Imaging in Neurology (ThINk) Basel, Department of Biomedical Engineering, University Hospital Basel and University of Basel, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel, Switzerland
- Medical Image Analysis Center (MIAC), Basel, Switzerland
| | - Nina de Oliveira S Siebenborn
- Neurology Clinic and Policlinic, Department of Medicine, Clinical Research and Biomedical Engineering, University Hospital Basel and University of Basel, Basel, Switzerland
- Translational Imaging in Neurology (ThINk) Basel, Department of Biomedical Engineering, University Hospital Basel and University of Basel, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel, Switzerland
- Medical Image Analysis Center (MIAC), Basel, Switzerland
| | - Riccardo Galbusera
- Neurology Clinic and Policlinic, Department of Medicine, Clinical Research and Biomedical Engineering, University Hospital Basel and University of Basel, Basel, Switzerland
- Translational Imaging in Neurology (ThINk) Basel, Department of Biomedical Engineering, University Hospital Basel and University of Basel, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel, Switzerland
| | - Sabine Schädelin
- Neurology Clinic and Policlinic, Department of Medicine, Clinical Research and Biomedical Engineering, University Hospital Basel and University of Basel, Basel, Switzerland
- Translational Imaging in Neurology (ThINk) Basel, Department of Biomedical Engineering, University Hospital Basel and University of Basel, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel, Switzerland
- Department of Clinical Research, University Hospital and University of Basel, Basel, Switzerland
| | - Pascal Benkert
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel, Switzerland
- Department of Clinical Research, University Hospital and University of Basel, Basel, Switzerland
| | - Jens Kuhle
- Neurology Clinic and Policlinic, Department of Medicine, Clinical Research and Biomedical Engineering, University Hospital Basel and University of Basel, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel, Switzerland
| | - Ludwig Kappos
- Multiple Sclerosis Centre, Department of Neurology, Biomedicine and Clinical Research, University Hospital and University of Basel, Basel, Switzerland
- Department of Neurology, University Hospital and University of Basel, Basel, Switzerland
| | - Lester Melie-Garcia
- Neurology Clinic and Policlinic, Department of Medicine, Clinical Research and Biomedical Engineering, University Hospital Basel and University of Basel, Basel, Switzerland
- Translational Imaging in Neurology (ThINk) Basel, Department of Biomedical Engineering, University Hospital Basel and University of Basel, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel, Switzerland
| | - Cristina Granziera
- Neurology Clinic and Policlinic, Department of Medicine, Clinical Research and Biomedical Engineering, University Hospital Basel and University of Basel, Basel, Switzerland
- Translational Imaging in Neurology (ThINk) Basel, Department of Biomedical Engineering, University Hospital Basel and University of Basel, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel, Switzerland
| |
Collapse
|
2
|
Bianco A, Di Sante G, Colò F, De Arcangelis V, Cicia A, Del Giacomo P, De Bonis M, Morganti TG, Carlomagno V, Lucchini M, Minucci A, Calabresi P, Mirabella M. Multiple Sclerosis Onset before and after COVID-19 Vaccination: Can HLA Haplotype Be Determinant? Int J Mol Sci 2024; 25:4556. [PMID: 38674141 PMCID: PMC11050425 DOI: 10.3390/ijms25084556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 04/13/2024] [Accepted: 04/18/2024] [Indexed: 04/28/2024] Open
Abstract
A few cases of multiple sclerosis (MS) onset after COVID-19 vaccination have been reported, although the evidence is insufficient to establish causality. The aim of this study is to compare cases of newly diagnosed relapsing-remitting MS before and after the outbreak of the COVID-19 pandemic and the impact of COVID-19 vaccination. Potential environmental and genetic predisposing factors were also investigated, as well as clinical patterns. This is a single-centre retrospective cohort study including all patients who presented with relapsing-remitting MS onset between January 2018 and July 2022. Data on COVID-19 vaccination administration, dose, and type were collected. HLA-DRB1 genotyping was performed in three subgroups. A total of 266 patients received a new diagnosis of relapsing-remitting MS in our centre, 143 before the COVID-19 pandemic (until and including March 2020), and 123 during the COVID-19 era (from April 2020). The mean number of new MS onset cases per year was not different before and during the COVID-19 era and neither were baseline patients' characteristics, type of onset, clinical recovery, or radiological patterns. Fourteen (11.4%) patients who subsequently received a new diagnosis of MS had a history of COVID-19 vaccination within one month before symptoms onset. Patients' characteristics, type of onset, clinical recovery, and radiological patterns did not differ from those of patients with non-vaccine-related new diagnoses of MS. The allele frequencies of HLA-DRB1*15 were 17.6% and 22.2% in patients with non-vaccine-related disease onset before and during the COVID-19 era, respectively, while no case of HLA-DRB1*15 was identified among patients with a new diagnosis of MS post-COVID-19 vaccine. In contrast, HLA-DRB1*08+ or HLA-DRB1*10+ MS patients were present only in this subgroup. Although a causal link between COVID-19 vaccination and relapsing-remitting MS cannot be detected, it is interesting to note and speculate about the peculiarities and heterogeneities underlying disease mechanisms of MS, where the interactions of genetics and the environment could be crucial also for the follow-up and the evaluation of therapeutic options.
Collapse
Affiliation(s)
- Assunta Bianco
- Division of Neurology, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Department of Neurosciences, Centro di Ricerca per la Sclerosi Multipla “Anna Paola Batocchi”, Catholic University of Sacred Heart, 00168 Rome, Italy
| | - Gabriele Di Sante
- Department of Medicine and Surgery, Section of Human, Clinical and Forensic Anatomy, University of Perugia, 06123 Perugia, Italy
| | - Francesca Colò
- Department of Neurosciences, Centro di Ricerca per la Sclerosi Multipla “Anna Paola Batocchi”, Catholic University of Sacred Heart, 00168 Rome, Italy
| | - Valeria De Arcangelis
- Division of Neurology, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Alessandra Cicia
- Department of Neurosciences, Centro di Ricerca per la Sclerosi Multipla “Anna Paola Batocchi”, Catholic University of Sacred Heart, 00168 Rome, Italy
| | - Paola Del Giacomo
- Department of Laboratory and Infectious Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Maria De Bonis
- Departmental Unit of Molecular and Genomic Diagnostics, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Genomics Core Facility, Gemelli Science and Technology Park (G-STeP), Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Tommaso Giuseppe Morganti
- Department of Neurosciences, Centro di Ricerca per la Sclerosi Multipla “Anna Paola Batocchi”, Catholic University of Sacred Heart, 00168 Rome, Italy
| | - Vincenzo Carlomagno
- Department of Neurosciences, Centro di Ricerca per la Sclerosi Multipla “Anna Paola Batocchi”, Catholic University of Sacred Heart, 00168 Rome, Italy
| | - Matteo Lucchini
- Division of Neurology, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Department of Neurosciences, Centro di Ricerca per la Sclerosi Multipla “Anna Paola Batocchi”, Catholic University of Sacred Heart, 00168 Rome, Italy
| | - Angelo Minucci
- Departmental Unit of Molecular and Genomic Diagnostics, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Genomics Core Facility, Gemelli Science and Technology Park (G-STeP), Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Paolo Calabresi
- Division of Neurology, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Department of Neurosciences, Centro di Ricerca per la Sclerosi Multipla “Anna Paola Batocchi”, Catholic University of Sacred Heart, 00168 Rome, Italy
| | - Massimiliano Mirabella
- Division of Neurology, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Department of Neurosciences, Centro di Ricerca per la Sclerosi Multipla “Anna Paola Batocchi”, Catholic University of Sacred Heart, 00168 Rome, Italy
| |
Collapse
|
3
|
Tredicine M, Ria F, Poerio N, Lucchini M, Bianco A, De Santis F, Valentini M, De Arcangelis V, Rende M, Stabile AM, Pistilli A, Camponeschi C, Nociti V, Mirabella M, Fraziano M, Di Sante G. Liposome-based nanoparticles impact on regulatory and effector phenotypes of macrophages and T cells in multiple Sclerosis patients. Biomaterials 2023; 292:121930. [PMID: 36493716 DOI: 10.1016/j.biomaterials.2022.121930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 11/19/2022] [Accepted: 11/22/2022] [Indexed: 11/30/2022]
Abstract
Current available treatments of Multiple Sclerosis (MS) reduce neuroinflammation acting on different targets on the immune system, but potentially lead to severe side effects and have a limited efficacy in slowing the progression of the disease. Here, we evaluated in vitro the immunomodulatory potential of a new class of nanoparticles - liposomes, constituted by a double-layer of phosphatidylserine (PSCho/PS), and double-faced, with an outer layer of phosphatidylserine and an inner layer of phosphatidic acid (PSCho/PA), either alone or in the presence of the myelin basic protein (MBP) peptide (residues 85-99) (PSCho/PS-MBP and PSCho/PA-MBP). Results showed that PSCho/PS are equally and efficiently internalized by pro- and anti-inflammatory macrophages (M1 and M2 respectively), while PSCho/PA were internalized better by M2 than M1. PSCho/PS liposomes were able to inhibit the secretion of innate pro-inflammatory cytokine IL-1β. PSCho/PS liposomes expanded Tregs, reducing Th1 and Th17 cells, while PSCho/PA liposomes were unable to dampen pro-inflammatory T cells and to promote immune-regulatory phenotype (Treg). The ability of PSCho/PS liposomes to up-regulate Treg cells was more pronounced in MS patients with high basal expression of M2 markers. PSCho/PS liposomes were more effective in decreasing Th1 (but not Th17) cells in MS patients with a disease duration >3 months. On the other hand, down-modulation of Th17 cells was evident in MS patients with active, Gadolinium enhancing lesions at MRI and in MS patients with a high basal expression of M1-associated markers in the monocytes. The same findings were observed for the modulation of MBP-driven Th1/Th17/Treg responses. These observations suggest that early MS associate to a hard-wired pro-Th1 phenotype of M1 that is lost later during disease course. On the other hand, acute inflammatory events reflect a temporary decrease of M2 phenotype that however is amenable to restauration upon treatment with PSCho/PS liposomes. Thus, together these data indicate that monocytes/macrophages may play an important regulatory function during MS course and suggest a role for PSCho/PS and PSCho/PS-MBP as new therapeutic tools to dampen the pro-inflammatory immune responses and to promote its regulatory branch.
Collapse
Affiliation(s)
- Maria Tredicine
- Section of General Pathology, Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy.
| | - Francesco Ria
- Section of General Pathology, Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCCS, Department Laboratory and Infectious diseases Sciences, Largo Agostino Gemelli 1-8, 00168, Rome, Italy.
| | - Noemi Poerio
- Department of Biology, University of Rome "TorVergata", Via della Ricerca Scientifica 1, 00173, Rome, Italy.
| | - Matteo Lucchini
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, UOC of Neurology, Largo Agostino Gemelli 8, 00168, Rome, Italy; Department of Neurosciences, Centro di Ricerca Sclerosi Multipla (CERSM), Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy.
| | - Assunta Bianco
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, UOC of Neurology, Largo Agostino Gemelli 8, 00168, Rome, Italy; Department of Neurosciences, Centro di Ricerca Sclerosi Multipla (CERSM), Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy.
| | - Federica De Santis
- Department of Biology, University of Rome "TorVergata", Via della Ricerca Scientifica 1, 00173, Rome, Italy.
| | - Mariagrazia Valentini
- Section of Pathology, Department of Woman, Child and Public Health Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli 1-8, 00168, Rome, Italy.
| | - Valeria De Arcangelis
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, UOC of Neurology, Largo Agostino Gemelli 8, 00168, Rome, Italy; Department of Neurosciences, Centro di Ricerca Sclerosi Multipla (CERSM), Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy.
| | - Mario Rende
- Department of Surgery and Medicine, Institute of Human, Clinical and Forensic Anatomy, Piazza L. Severi 1, 06125, Perugia, Italy.
| | - Anna Maria Stabile
- Department of Surgery and Medicine, Institute of Human, Clinical and Forensic Anatomy, Piazza L. Severi 1, 06125, Perugia, Italy.
| | - Alessandra Pistilli
- Department of Surgery and Medicine, Institute of Human, Clinical and Forensic Anatomy, Piazza L. Severi 1, 06125, Perugia, Italy.
| | - Chiara Camponeschi
- Section of General Pathology, Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy; Institute of Chemical Sciences and Technologies ''Giulio Natta'' (SCITEC)-CNR, Largo Francesco Vito 1, 00168, Rome, Italy.
| | - Viviana Nociti
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, UOC of Neurology, Largo Agostino Gemelli 8, 00168, Rome, Italy; Department of Neurosciences, Centro di Ricerca Sclerosi Multipla (CERSM), Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy.
| | - Massimiliano Mirabella
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, UOC of Neurology, Largo Agostino Gemelli 8, 00168, Rome, Italy; Department of Neurosciences, Centro di Ricerca Sclerosi Multipla (CERSM), Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy.
| | - Maurizio Fraziano
- Department of Biology, University of Rome "TorVergata", Via della Ricerca Scientifica 1, 00173, Rome, Italy.
| | - Gabriele Di Sante
- Section of General Pathology, Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy; Department of Surgery and Medicine, Institute of Human, Clinical and Forensic Anatomy, Piazza L. Severi 1, 06125, Perugia, Italy.
| |
Collapse
|
4
|
Butler R, Bradford D, Rodgers KE. Analysis of shared underlying mechanism in neurodegenerative disease. Front Aging Neurosci 2022; 14:1006089. [PMID: 36523957 PMCID: PMC9745190 DOI: 10.3389/fnagi.2022.1006089] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 11/08/2022] [Indexed: 08/27/2023] Open
Abstract
In this review, the relationship between bioenergetics, mitochondrial dysfunction, and inflammation will be and how they contribute to neurodegeneration, specifically in Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), and multiple sclerosis (MS) will be reviewed. Long-term changes in mitochondrial function, autophagy dysfunction, and immune activation are commonalities shared across these age-related disorders. Genetic risk factors for these diseases support an autophagy-immune connection in the underlying pathophysiology. Critical areas of deeper evaluation in these bioenergetic processes may lead to potential therapeutics with efficacy across multiple neurodegenerative diseases.
Collapse
Affiliation(s)
| | | | - Kathleen E. Rodgers
- Department of Medical Pharmacology, Center for Innovation in Brain Science, University of Arizona College of Medicine, Tucson, AZ, United States
| |
Collapse
|
5
|
Vitamin D Receptor Polymorphisms Among the Turkish Population are Associated with Multiple Sclerosis. Balkan J Med Genet 2022; 25:41-50. [PMID: 36880035 PMCID: PMC9985364 DOI: 10.2478/bjmg-2022-0003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2023] Open
Abstract
Multiple sclerosis (MS) is an inflammatory disease characterized by demyelination and axonal degeneration affecting the central nervous system. Among the genetic factors suggested to be associated with this disease are polymorphisms to the vitamin D receptor (VDR) gene. We tested the hypothesis that polymorphisms in the vitamin D receptor (VDR) gene are associated with MS. The aim of the study was to investigate the relationship of MS with the VDR gene Fok-I, Bsm-I and Taq-I polymorphisms among the Turkish population. This study contains 271 MS patients and 203 healthy controls. Genomic DNA was isolated from the samples and the VDR gene Fok-I, Bsm-I and Taq-I polymorphism regions were amplified by polymerase chain reaction (PCR). The PCR products were digested, and the genotypes were determined based on size of digested PCR products. Our results demonstrate associations between MS and the distribution of the VDR gene Fok-I T/T polymorphism genotype in a dominant model, VDR gene Fok-I T allele frequency, distribution of VDR gene Taq-I C/C polymorphism genotype in a dominant model and VDR gene Taq-I C allele frequency (Pearson test, p<0.05). However, there was no association between MS and the VDR gene Bsm-I polymorphisms for the genotype distribution (Pearson test, p>0.05) or allele frequency (Pearson test, p>0.05). Fok-I and Taq-I VDR gene polymorphisms are significantly associated with MS in dominant, homozygote and heterozygote inheritance models among the Turkish population.
Collapse
|
6
|
McCombe PA. The role of sex and pregnancy in multiple sclerosis: what do we know and what should we do? Expert Rev Neurother 2022; 22:377-392. [PMID: 35354378 DOI: 10.1080/14737175.2022.2060079] [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: 11/04/2022]
Abstract
INTRODUCTION Multiple sclerosis (MS) is more prevalent in women than in men. The sex of the patient, and pregnancy, are reported to be associated with the clinical features of MS. The mechanism of this is unclear. AREAS COVERED This review summarizes data about sex differences in MS and the role of pregnancy. Possible mechanisms for the effects of sex and pregnancy are summarized, and practical suggestions for addressing these issues are provided. EXPERT OPINION There is considerable interdependence of the variables that are associated with MS. Men have a worse outcome of MS, and this could be due to the same factors that lead to greater incidence of neurodegenerative disease in men. The possible role of parity on the long-term outcome of MS is of interest. Future studies that look at the mechanisms of the effects of the sex of the patient on the outcome of MS are required. However, there are some actions that can be taken without further research. We can concentrate on public health measures that address the modifiable risk factors for MS and ensure that disease is controlled in women who intend to become pregnant and use appropriate disease modifying agents during pregnancy.
Collapse
Affiliation(s)
- Pamela A McCombe
- The University of Queensland, Centre for Clinical Research, Royal Brisbane and Women's Hospital, Herston, Australia
| |
Collapse
|
7
|
Genetics and familial distribution of multiple sclerosis: A review. Rev Neurol (Paris) 2022; 178:512-520. [DOI: 10.1016/j.neurol.2021.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 09/30/2021] [Accepted: 11/04/2021] [Indexed: 11/18/2022]
|
8
|
Genetics and functional genomics of multiple sclerosis. Semin Immunopathol 2022; 44:63-79. [PMID: 35022889 DOI: 10.1007/s00281-021-00907-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 12/13/2021] [Indexed: 12/14/2022]
Abstract
Multiple sclerosis (MS) is an inflammatory neurodegenerative disease with genetic predisposition. Over the last decade, genome-wide association studies with increasing sample size led to the discovery of robustly associated genetic variants at an exponential rate. More than 200 genetic loci have been associated with MS susceptibility and almost half of its heritability can be accounted for. However, many challenges and unknowns remain. Definitive studies of disease progression and endophenotypes are yet to be performed, whereas the majority of the identified MS variants are not yet functionally characterized. Despite these shortcomings, the unraveling of MS genetics has opened up a new chapter on our understanding MS causal mechanisms.
Collapse
|
9
|
Aravilli RK, Vikram SL, Kohila V. The Functional Impact of Alternative Splicing and Single Nucleotide Polymorphisms in Rheumatoid Arthritis. Curr Pharm Biotechnol 2021; 22:1014-1029. [PMID: 33001009 DOI: 10.2174/1389201021666201001142416] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 09/05/2020] [Accepted: 09/08/2020] [Indexed: 11/22/2022]
Abstract
Advances in genomics and proteomics aid the identification of genes associated with various diseases. Genome-Wide Association Studies (GWAS) have identified multiple loci as risk alleles for susceptibility to Rheumatoid Arthritis (RA). A bisection of RA risk can be attributed to genetic factors. Over 100 associated genetic loci that encompass immune regulatory factors have been found to be linked with RA. Aberrant Single Nucleotide Polymorphisms (SNPs) and alternative splicing mechanisms in such loci induce RA. These aberrations are viewed as potential therapeutic targets due to their association with a multitude of diseases. This review presents a few imperious genes whose alterations can cause severe bone deformities culminating in RA.
Collapse
Affiliation(s)
- R Kowshik Aravilli
- Department of Biotechnology, National Institute of Technology Warangal, Warangal, India
| | - S Laveen Vikram
- Department of Computer Science and Engineering, Alagappa University, Karaikudi, India
| | - V Kohila
- Department of Biotechnology, National Institute of Technology Warangal, Warangal, India
| |
Collapse
|
10
|
Influence of immunomodulatory drugs on the gut microbiota. Transl Res 2021; 233:144-161. [PMID: 33515779 PMCID: PMC8184576 DOI: 10.1016/j.trsl.2021.01.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 12/10/2020] [Accepted: 01/26/2021] [Indexed: 12/17/2022]
Abstract
Immunomodulatory medications are a mainstay of treatment for autoimmune diseases and malignancies. In addition to their direct effects on immune cells, these medications also impact the gut microbiota. Drug-induced shifts in commensal microbes can lead to indirect but important changes in the immune response. We performed a comprehensive literature search focusing on immunotherapy/microbe interactions. Immunotherapies were categorized into 5 subtypes based on their mechanisms of action: cell trafficking inhibitors, immune checkpoint inhibitors, immunomodulators, antiproliferative drugs, and inflammatory cytokine inhibitors. Although no consistent relationships were observed between types of immunotherapy and microbiota, most immunotherapies were associated with shifts in specific colonizing bacterial taxa. The relationships between colonizing microbes and drug efficacy were not well-studied for autoimmune diseases. In contrast, the efficacy of immune checkpoint inhibitors for cancer was tied to the baseline composition of the gut microbiota. There was a paucity of high-quality data; existing data were generated using heterogeneous sampling and analytic techniques, and most studies involved small numbers of participants. Further work is needed to elucidate the extent and clinical significance of immunotherapy effects on the human microbiome.
Collapse
|
11
|
Exome sequencing reveals novel rare variants in Iranian familial multiple sclerosis: The importance of POLD2 in the disease pathogenesis. Genomics 2021; 113:2645-2655. [PMID: 34116171 DOI: 10.1016/j.ygeno.2021.06.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 04/20/2021] [Accepted: 06/06/2021] [Indexed: 02/07/2023]
Abstract
The prevalence of familial multiple sclerosis (FMS) is increasing worldwide which endorses the heritability of the disease. Given that many genome variations are ethnicity-specific and consanguineous marriage could affect genetic diseases, hereditary disease gene analysis among FMS patients from Iran, a country with high rates of parental consanguinity, could be highly effective in finding mutations underlying disease pathogenesis. To examine rare genetic mutations, we selected three Iranian FMS cases with ≥3 MS patients in more than one generation and performed whole exome sequencing. We identified a homozygous rare missense variant in POLD2 (p. Arg141Cys; rs372336011). Molecular dynamics analysis showed reduced polar dehydration energy and conformational changes in POLD2 mutant. Further, we found a heterozygote rare missense variant in NBFP1 (p. Gly487Asp; rs778806175). Our study revealed the possible role of novel rare variants in FMS. Molecular dynamic simulation provided the initial evidence of the structural changes behind POLD2 mutant.
Collapse
|
12
|
Manouchehri N, Zhang Y, Salter A, Hussain RZ, Hartung HP, Hemmer B, Linker R, Segal BM, Cutter G, Stüve O. Clinical trials in multiple sclerosis: potential future trial designs. Ther Adv Neurol Disord 2019; 12:1756286419847095. [PMID: 31205492 PMCID: PMC6535717 DOI: 10.1177/1756286419847095] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 03/29/2019] [Indexed: 12/13/2022] Open
Abstract
Clinical trials of new treatments in multiple sclerosis (MS) currently require large sample sizes and long durations in order to yield reliable results. The differential responses of an already heterogeneous population of MS patients to individual disease-modifying therapies (DMTs) will further complicate future trials. MS trials with smaller samples and faster outcomes are conceivable through the substitution of current clinical and MRI outcomes with objectively measureable genomic and proteomic biomarkers. Currently, biomarkers that could be utilized for diagnosis and monitoring of MS disease activity are in the early validation phase. The power of single biomarkers or multiple correlated biomarkers to predict prognosis and response to treatment could initially be compared with currently accepted methods. These prospectively validated disease biomarkers could then be used to subcategorize the spectrum of MS patients into a finite number of endophenotypes with demonstrable different molecular pathogeneses and DMT response profiles. Newly developed DMT could potentially be assessed within specific endophenotypes and compared with pharmacogenomically relevant active comparator DMT. This approach may increase the efficiency of MS trials through homogenization of patient population and minimization of nonresponders in study groups, providing the potential for the development of targeted therapies.
Collapse
Affiliation(s)
- Navid Manouchehri
- Department of Neurology and Neurotherapeutics, The University of Texas Southwestern Medical Center, Dallas, TX
| | - Yinan Zhang
- Department of Neurology and Neurotherapeutics, The University of Texas Southwestern Medical Center, Dallas, TX
| | - Amber Salter
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO
| | - Rehana Z Hussain
- Department of Neurology and Neurotherapeutics, The University of Texas Southwestern Medical Center, Dallas, TX
| | - Hans-Peter Hartung
- Department of Neurology, UKD and Center for Neurology and Neuropsychiatry, LVR Klinikum, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Bernhard Hemmer
- Department of Neurology, Klinikum rechts der Isar, Technische Universität München, Germany
| | - Ralf Linker
- Department of Neurology, University of Regensburg, Germany
| | | | - Gary Cutter
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, AL
| | - Olaf Stüve
- Neurology Section, VA North Texas Health Care System, Medical Service, 500 South Lancaster Rd., Dallas, TX 75216, USA Department of Neurology and Neurotherapeutics, The University of Texas Southwestern Medical Center, Dallas, TX Department of Neurology, Klinikum rechts der Isar, Technische Universität München, Germany
| |
Collapse
|
13
|
Abstract
The contribution of genetic inheritance in multiple sclerosis was established early on. Although multiple sclerosis is not a Mendelian disease, its incidence and prevalence is higher in family members of affected individuals compared with the general population. Throughout the last decade, several small studies failed to identify any robust genetic associations besides the classic associations in the major histocompatibility complex region. During the past few years, genome-wide association studies (GWAS) have revolutionized the genetics of multiple sclerosis, uncovering more than 200 implicated genetic loci. Here, we describe these main findings and discuss the new avenues that these discoveries lay open.
Collapse
Affiliation(s)
- Nikolaos A Patsopoulos
- Department of Neurology, Brigham & Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115
- Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts 02142
| |
Collapse
|
14
|
|
15
|
Baranzini SE, Oksenberg JR. The Genetics of Multiple Sclerosis: From 0 to 200 in 50 Years. Trends Genet 2017; 33:960-970. [PMID: 28987266 PMCID: PMC5701819 DOI: 10.1016/j.tig.2017.09.004] [Citation(s) in RCA: 142] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 08/26/2017] [Accepted: 09/11/2017] [Indexed: 12/12/2022]
Abstract
Multiple sclerosis (MS) is a common autoimmune disease that targets myelin in the central nervous system (CNS). Multiple genome-wide association studies (GWAS) over the past 10 years have uncovered more than 200 loci that independently contribute to disease pathogenesis. As with many other complex diseases, risk of developing MS is driven by multiple common variants whose biological effects are not immediately clear. Here, we present a historical perspective on the progress made in MS genetics and discuss current work geared towards creating a more complete model that accurately represents the genetic landscape of MS susceptibility. Such a model necessarily includes a better understanding of the individual contributions of each common variant to the cellular phenotypes, and interactions with other genes and with the environment. Future genetic studies in MS will likely focus on the role of rare variants and endophenotypes.
Collapse
Affiliation(s)
- Sergio E Baranzini
- Weill Institute for Neurosciences. Department of Neurology, University of California San Francisco, San Francisco, CA, USA; Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA; Graduate Program in Bioinformatics, University of California San Francisco, San Francisco, CA, USA.
| | - Jorge R Oksenberg
- Weill Institute for Neurosciences. Department of Neurology, University of California San Francisco, San Francisco, CA, USA; Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
16
|
Kelly MEM, Lehmann C, Zhou J. The Endocannabinoid System in Local and Systemic Inflammation. ACTA ACUST UNITED AC 2017. [DOI: 10.4199/c00151ed1v01y201702isp074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
17
|
Haghighi S, Andersen O, Nilsson S, Rydberg L, Wahlström J. A linkage study in two families with multiple sclerosis and healthy members with oligoclonal CSF immunopathy. Mult Scler 2016; 12:723-30. [PMID: 17262999 DOI: 10.1177/1352458506070972] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
We studied two extended families in which not only multiple sclerosis (MS) segregates, but also approximately 18% of the cerebrospinal fluid (CSF) investigated blood relatives have ‘MS immunopathic trait’, an oligoclonal CSF immunopathy similar to that seen in MS, but with no neurological symptoms. Both families fit a genetic model for autosomal dominant inheritance for MS immunopathic trait, although with reduced penetrance in family A. In order to identify genetic factors of importance for the development of MS immunopathic trait, we performed a genome scan using the CHLC/Weber Screening Set (ver 6A), with 285 successful markers, to test the hypothesis that a single gene is causing the MS immunopathic trait in these families. Using a parametric method, we identified regions with suggestive linkage at chromosome 6q12 with a LOD-score of 2.4, putative linkage with LOD-score 1.5 at chromosome 6p21 (HLA region), putative linkage at chromosome 12q24 with a LOD-score of 1.7 and suggestive linkage at chromosome 19q13.2 with a LOD-score of 1.8. The LOD-score at chromosome 19q13.2 increased to 2.2 when only family A was analysed. In family A, all MS patients and two of five individuals with MS immunopathic trait had HLA DRB1*(15) and in family B, all blood relatives had the rare HLA type DRB1*0103, which is associated with other autoimmune diseases. We suggest that DRB1*0103 is a necessary but not sufficient condition for the susceptibility for MS immunopathic trait in this family.
Collapse
Affiliation(s)
- S Haghighi
- Institute of Clinical Neuroscience, The Sahlgrenska Academy at Göteborg University, Göteborg, Sweden.
| | | | | | | | | |
Collapse
|
18
|
Nakane S, Matsuo H, Goto H, Yoshinaga-Matsumoto M, Ohtsuru I, Ichinose K, Onodera H, Yoshida M, Shibuya N. Cytapheresis with a filter for selective removal of CD4+ T cells in experimental autoimmune encephalomyelitis. Mult Scler 2016; 9:579-84. [PMID: 14664470 DOI: 10.1191/1352458503ms968oa] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Experimental autoimmune encephalomyelitis (EAE) is a major animal model of human multiple sclerosis (MS). CD4+ T cells are thought to play a pivotal role in the patho genesis of EAE and MS. In order to investigate the depletio n of CD4+ T cells from the systemic circulation as an effective strategy for the treatment of MS, we performed extracorporeal CD4+ T cell adsorption, using a filter to which anti-CD4+ antibody is immobilized as a ligand, in adoptively transferred EAE. Rats treated with CD4+ T cell removal filter (C D4RF) exhibited milder clinical signs of EAE and earlier recovery than those receiving sham treatment. Moreover, the thymic cells from EAE rats treated with C D4RF exhibited a suppressed proliferative response and IFN-g production to myelin basic protein. These results suggest that depletion of CD4+ T cells from the systemic circulation by extracorporeal treatment is a potentially useful strategy for treatment of acute phase and relapsing MS.
Collapse
MESH Headings
- Acute Disease
- Adoptive Transfer
- Animals
- CD4-Positive T-Lymphocytes/cytology
- CD4-Positive T-Lymphocytes/immunology
- Cell Line
- Cytapheresis/instrumentation
- Cytapheresis/methods
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/therapy
- Interferon-gamma/metabolism
- Multiple Sclerosis, Relapsing-Remitting/immunology
- Multiple Sclerosis, Relapsing-Remitting/pathology
- Multiple Sclerosis, Relapsing-Remitting/therapy
- Myelin Basic Protein/immunology
- Rats
- Rats, Inbred Lew
- Thymus Gland/cytology
- Thymus Gland/immunology
- Thymus Gland/metabolism
Collapse
Affiliation(s)
- Shunya Nakane
- Department of Neurology, Kawatana National Hospital, Nagasaki, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Kallaur AP, Reiche EMV, Oliveira SR, Simão ANC, Pereira WLDCJ, Alfieri DF, Flauzino T, Proença CDM, Lozovoy MAB, Kaimen-Maciel DR, Maes M. Genetic, Immune-Inflammatory, and Oxidative Stress Biomarkers as Predictors for Disability and Disease Progression in Multiple Sclerosis. Mol Neurobiol 2016; 54:31-44. [PMID: 26732588 DOI: 10.1007/s12035-015-9648-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 12/17/2015] [Indexed: 12/17/2022]
Abstract
The aim of this study was to evaluate the TNFβ NcoI polymorphism (rs909253) and immune-inflammatory, oxidative, and nitrosative stress (IO&NS) biomarkers as predictors of disease progression in multiple sclerosis (MS). We included 212 MS patients (150 female, 62 male, mean (±standard deviation (SD)) age = 42.7 ± 13.8 years) and 249 healthy controls (177 female, 72 male, 36.8 ± 11 years). The disability was measured the Expanded Disability Status Scale (EDSS) in 2006 and 2011. We determined the TNFβ NcoI polymorphism and serum levels of interleukin (IL)-6, tumor necrosis factor (TNF)-α, interferon (IFN)-γ, IL-4, IL-10, and IL-17, albumin, ferritin, and plasma levels of lipid hydroperoxides (CL-LOOH), carbonyl protein, advanced oxidation protein products (AOPPs), nitric oxide metabolites (NOx), and total radical-trapping antioxidant parameter (TRAP). The mean EDSS (±SD) in 2006 was 1.62 ± 2.01 and in 2011 3.16 ± 2.29, and disease duration was 7.34 ± 7.0 years. IL-10, TNF-α, IFN-γ, AOPP, and NOx levels were significantly higher and IL-4 lower in MS patients with a higher 2011 EDSS scores (≥3) as compared with those with EDSS < 3. The actual increases in EDSS from 2006 to 2011 were positively associated with TNF-α and IFN-γ. Increased IFN-γ values were associated with higher pyramidal symptoms and increased IL-6 with sensitive symptoms. Increased carbonyl protein and IL-10 but lowered albumin levels predicted cerebellar symptoms. The TNFB1/B2 genotype decreased risk towards progression of pyramidal symptoms. Treatments with IFN-β and glatiramer acetate significantly reduced TNF-α but did not affect the other IO&NS biomarkers or disease progression. Taken together, IO&NS biomarkers and NcoI TNFβ genotypes predict high disability in MS and are associated with different aspects of disease progression. New drugs to treat MS should also target oxidative stress pathways.
Collapse
Affiliation(s)
- Ana Paula Kallaur
- Health Sciences Postgraduate Program, Health Sciences Center, State University of Londrina, Londrina, Paraná, 86.038-440, Brazil
| | - Edna Maria Vissoci Reiche
- Department of Pathology, Clinical Analysis and Toxicology, Health Sciences Center, State University of Londrina, Av. Robert Koch, 60, Londrina, Paraná, CEP 86.038-440, Brazil.
| | - Sayonara Rangel Oliveira
- Health Sciences Postgraduate Program, Health Sciences Center, State University of Londrina, Londrina, Paraná, 86.038-440, Brazil
| | - Andrea Name Colado Simão
- Department of Pathology, Clinical Analysis and Toxicology, Health Sciences Center, State University of Londrina, Av. Robert Koch, 60, Londrina, Paraná, CEP 86.038-440, Brazil
| | - Wildea Lice de Carvalho Jennings Pereira
- Health Sciences Postgraduate Program, Health Sciences Center, State University of Londrina, Londrina, Paraná, 86.038-440, Brazil.,Outpatient Clinic for Demyelinating Diseases, University Hospital, State University of Londrina, Londrina, Paraná, 86061-335, Brazil
| | - Daniela Frizon Alfieri
- Health Sciences Postgraduate Program, Health Sciences Center, State University of Londrina, Londrina, Paraná, 86.038-440, Brazil
| | - Tamires Flauzino
- Health Sciences Postgraduate Program, Health Sciences Center, State University of Londrina, Londrina, Paraná, 86.038-440, Brazil
| | - Caio de Meleck Proença
- Outpatient Clinic for Demyelinating Diseases, University Hospital, State University of Londrina, Londrina, Paraná, 86061-335, Brazil
| | - Marcell Alysson Batisti Lozovoy
- Department of Pathology, Clinical Analysis and Toxicology, Health Sciences Center, State University of Londrina, Av. Robert Koch, 60, Londrina, Paraná, CEP 86.038-440, Brazil
| | - Damacio Ramón Kaimen-Maciel
- Outpatient Clinic for Demyelinating Diseases, University Hospital, State University of Londrina, Londrina, Paraná, 86061-335, Brazil.,Department of Clinical Medicine, Health Sciences Center, State University of Londrina, Londrina, Paraná, 86.038-440, Brazil
| | - Michael Maes
- Impact Strategic Research Centre, School of Medicine, Deakin University, Geelong, Victoria, Australia.,Department of Psychiatry, King Chulalongkorn Memorial Hospital, Chulalongkorn, Bangkok, Thailand
| |
Collapse
|
20
|
Croxford JL, Miyake S. Animal Models for the Study of Neuroimmunological Disease. NEUROIMMUNOLOGICAL DISEASES 2016. [PMCID: PMC7122656 DOI: 10.1007/978-4-431-55594-0_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2022]
|
21
|
A review of genome-wide association studies for multiple sclerosis: classical and hypothesis-driven approaches. Hum Genet 2015; 134:1143-62. [DOI: 10.1007/s00439-015-1601-2] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 09/10/2015] [Indexed: 12/17/2022]
|
22
|
Case LK, Wall EH, Osmanski EE, Dragon JA, Saligrama N, Zachary JF, Lemos B, Blankenhorn EP, Teuscher C. Copy number variation in Y chromosome multicopy genes is linked to a paternal parent-of-origin effect on CNS autoimmune disease in female offspring. Genome Biol 2015; 16:28. [PMID: 25886764 PMCID: PMC4396973 DOI: 10.1186/s13059-015-0591-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 01/20/2015] [Indexed: 12/11/2022] Open
Abstract
Background The prevalence of some autoimmune diseases is greater in females compared with males, although disease severity is often greater in males. The reason for this sexual dimorphism is unknown, but it may reflect negative selection of Y chromosome-bearing sperm during spermatogenesis or male fetuses early in the course of conception/pregnancy. Previously, we showed that the sexual dimorphism in experimental autoimmune encephalomyelitis (EAE) is associated with copy number variation (CNV) in Y chromosome multicopy genes. Here, we test the hypothesis that CNV in Y chromosome multicopy genes influences the paternal parent-of-origin effect on EAE susceptibility in female mice. Results We show that C57BL/6 J consomic strains of mice possessing an identical X chromosome and CNV in Y chromosome multicopy genes exhibit sperm head abnormalities and female-biased sex ratio. This is consistent with X-Y intragenomic conflict arising from an imbalance in CNV between homologous X:Y chromosome multicopy genes. These males also display paternal transmission of EAE to female offspring and differential loading of microRNAs within the sperm nucleus. Furthermore, in humans, families of probands with multiple sclerosis similarly exhibit a female-biased sex ratio, whereas families of probands affected with non-sexually dimorphic autoimmune diseases exhibit unbiased sex ratios. Conclusions These findings provide evidence for a mechanism at the level of the male gamete that contributes to the sexual dimorphism in EAE and paternal parent-of-origin effects in female mice, raising the possibility that a similar mechanism may contribute to the sexual dimorphism in multiple sclerosis. Electronic supplementary material The online version of this article (doi:10.1186/s13059-015-0591-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Laure K Case
- Department of Medicine, University of Vermont, Given Medical Building C317, Burlington, VT, 05405, USA.
| | - Emma H Wall
- Department of Medicine, University of Vermont, Given Medical Building C317, Burlington, VT, 05405, USA.
| | - Erin E Osmanski
- Department of Medicine, University of Vermont, Given Medical Building C317, Burlington, VT, 05405, USA.
| | - Julie A Dragon
- Department of Microbiology and Molecular Genetics, University of Vermont, Burlington, VT, 05405, USA.
| | - Naresha Saligrama
- Department of Medicine, University of Vermont, Given Medical Building C317, Burlington, VT, 05405, USA. .,Current address: Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| | - James F Zachary
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL, 61802, USA.
| | - Bernardo Lemos
- Department of Environmental Health, Harvard School of Public Health, Boston, MA, 02115, USA.
| | - Elizabeth P Blankenhorn
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, 19129, USA.
| | - Cory Teuscher
- Department of Medicine, University of Vermont, Given Medical Building C317, Burlington, VT, 05405, USA. .,Department of Pathology, University of Vermont, Burlington, VT, 05405, USA.
| |
Collapse
|
23
|
Paradowska-Gorycka A, Jurkowska M, Felis-Giemza A, Romanowska-Próchnicka K, Manczak M, Maslinski S, Olesinska M. Genetic polymorphisms of Foxp3 in patients with rheumatoid arthritis. J Rheumatol 2014; 42:170-80. [PMID: 25448791 DOI: 10.3899/jrheum.131381] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
OBJECTIVE The aim of the study was to identify 2 polymorphic variants in the promoter region of the Foxp3 gene and their possible association with susceptibility to and severity of rheumatoid arthritis (RA). The association between genetic factors and pathogenesis suggests that T cells take part in the induction of RA. The CD4+CD25highFoxp3+ subset of regulatory T cells plays an essential role in preventing autoimmunity and maintaining immune homeostasis. METHODS Patients with RA (n = 274) and healthy individuals (n = 295) were examined for -3279 C/A and -924 A/G Foxp3 gene polymorphisms by the polymerase chain reaction-restriction fragment-length polymorphism method. Serum Foxp3 levels in patients with RA and controls were measured with ELISA. RESULTS Foxp3 -3279 A and -924 G alleles were associated with significantly elevated risk of RA in the population tested (p = 0.003 and p = 0.004, respectively) compared to the wild-type alleles. Overall, -3279 C/A and -924 A/G Foxp3 gene polymorphisms were in indistinct linkage disequilibrium with D' = 0.481 and r(2) = 0.225. From 4 possible haplotypes, frequencies of 2 (AG and CA) showed significant differences between both examined groups (respectively, p < 0.001 and p = 0.007). After appropriate adjustment of Bonferroni correction for multiple testing, the genotype-phenotype analysis showed no significant correlation of the Foxp3 -3279 C/A and -924 A/G polymorphisms with the disease activity, joint damage, laboratory variables, and extraarticular manifestation in patients with RA. Serum Foxp3 level was significantly higher in patients than in controls (p < 0.0001). CONCLUSION Current findings indicated that the Foxp3 genetic polymorphism and the Foxp3 protein level may be associated with susceptibility to RA in the Polish population.
Collapse
Affiliation(s)
- Agnieszka Paradowska-Gorycka
- From the departments of Biochemistry and Molecular Biology, Connective Tissue Diseases, and Epidemiology and Health Promotion, Institute of Rheumatology; the Department of Pathophysiology, Medical University of Warsaw, Warsaw, Poland.A. Paradowska-Gorycka, PhD; M. Jurkowska, PhD, Department of Biochemistry and Molecular Biology, Institute of Rheumatology; A. Felis-Giemza, MD; K. Romanowska-Próchnicka, MD, Department of Connective Tissue Diseases, Institute of Rheumatology; M. Manczak, MS, Department of Epidemiology and Health Promotion, Institute of Rheumatology; S. Maslinski, Professor, Department of Pathophysiology, Medical University of Warsaw; M. Olesinska, Professor, Department of Connective Tissue Diseases, Institute of Rheumatology.
| | - Monika Jurkowska
- From the departments of Biochemistry and Molecular Biology, Connective Tissue Diseases, and Epidemiology and Health Promotion, Institute of Rheumatology; the Department of Pathophysiology, Medical University of Warsaw, Warsaw, Poland.A. Paradowska-Gorycka, PhD; M. Jurkowska, PhD, Department of Biochemistry and Molecular Biology, Institute of Rheumatology; A. Felis-Giemza, MD; K. Romanowska-Próchnicka, MD, Department of Connective Tissue Diseases, Institute of Rheumatology; M. Manczak, MS, Department of Epidemiology and Health Promotion, Institute of Rheumatology; S. Maslinski, Professor, Department of Pathophysiology, Medical University of Warsaw; M. Olesinska, Professor, Department of Connective Tissue Diseases, Institute of Rheumatology
| | - Anna Felis-Giemza
- From the departments of Biochemistry and Molecular Biology, Connective Tissue Diseases, and Epidemiology and Health Promotion, Institute of Rheumatology; the Department of Pathophysiology, Medical University of Warsaw, Warsaw, Poland.A. Paradowska-Gorycka, PhD; M. Jurkowska, PhD, Department of Biochemistry and Molecular Biology, Institute of Rheumatology; A. Felis-Giemza, MD; K. Romanowska-Próchnicka, MD, Department of Connective Tissue Diseases, Institute of Rheumatology; M. Manczak, MS, Department of Epidemiology and Health Promotion, Institute of Rheumatology; S. Maslinski, Professor, Department of Pathophysiology, Medical University of Warsaw; M. Olesinska, Professor, Department of Connective Tissue Diseases, Institute of Rheumatology
| | - Katarzyna Romanowska-Próchnicka
- From the departments of Biochemistry and Molecular Biology, Connective Tissue Diseases, and Epidemiology and Health Promotion, Institute of Rheumatology; the Department of Pathophysiology, Medical University of Warsaw, Warsaw, Poland.A. Paradowska-Gorycka, PhD; M. Jurkowska, PhD, Department of Biochemistry and Molecular Biology, Institute of Rheumatology; A. Felis-Giemza, MD; K. Romanowska-Próchnicka, MD, Department of Connective Tissue Diseases, Institute of Rheumatology; M. Manczak, MS, Department of Epidemiology and Health Promotion, Institute of Rheumatology; S. Maslinski, Professor, Department of Pathophysiology, Medical University of Warsaw; M. Olesinska, Professor, Department of Connective Tissue Diseases, Institute of Rheumatology
| | - Malgorzata Manczak
- From the departments of Biochemistry and Molecular Biology, Connective Tissue Diseases, and Epidemiology and Health Promotion, Institute of Rheumatology; the Department of Pathophysiology, Medical University of Warsaw, Warsaw, Poland.A. Paradowska-Gorycka, PhD; M. Jurkowska, PhD, Department of Biochemistry and Molecular Biology, Institute of Rheumatology; A. Felis-Giemza, MD; K. Romanowska-Próchnicka, MD, Department of Connective Tissue Diseases, Institute of Rheumatology; M. Manczak, MS, Department of Epidemiology and Health Promotion, Institute of Rheumatology; S. Maslinski, Professor, Department of Pathophysiology, Medical University of Warsaw; M. Olesinska, Professor, Department of Connective Tissue Diseases, Institute of Rheumatology
| | - Slawomir Maslinski
- From the departments of Biochemistry and Molecular Biology, Connective Tissue Diseases, and Epidemiology and Health Promotion, Institute of Rheumatology; the Department of Pathophysiology, Medical University of Warsaw, Warsaw, Poland.A. Paradowska-Gorycka, PhD; M. Jurkowska, PhD, Department of Biochemistry and Molecular Biology, Institute of Rheumatology; A. Felis-Giemza, MD; K. Romanowska-Próchnicka, MD, Department of Connective Tissue Diseases, Institute of Rheumatology; M. Manczak, MS, Department of Epidemiology and Health Promotion, Institute of Rheumatology; S. Maslinski, Professor, Department of Pathophysiology, Medical University of Warsaw; M. Olesinska, Professor, Department of Connective Tissue Diseases, Institute of Rheumatology
| | - Marzena Olesinska
- From the departments of Biochemistry and Molecular Biology, Connective Tissue Diseases, and Epidemiology and Health Promotion, Institute of Rheumatology; the Department of Pathophysiology, Medical University of Warsaw, Warsaw, Poland.A. Paradowska-Gorycka, PhD; M. Jurkowska, PhD, Department of Biochemistry and Molecular Biology, Institute of Rheumatology; A. Felis-Giemza, MD; K. Romanowska-Próchnicka, MD, Department of Connective Tissue Diseases, Institute of Rheumatology; M. Manczak, MS, Department of Epidemiology and Health Promotion, Institute of Rheumatology; S. Maslinski, Professor, Department of Pathophysiology, Medical University of Warsaw; M. Olesinska, Professor, Department of Connective Tissue Diseases, Institute of Rheumatology
| |
Collapse
|
24
|
Tumor necrosis factor beta NcoI polymorphism is associated with inflammatory and metabolic markers in multiple sclerosis patients. J Neurol Sci 2014; 346:156-63. [PMID: 25173940 DOI: 10.1016/j.jns.2014.08.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 08/10/2014] [Accepted: 08/13/2014] [Indexed: 12/19/2022]
Abstract
To evaluate the association between the tumor necrosis factor beta (TNF-β) NcoI polymorphism and inflammatory and metabolic markers in patients with multiple sclerosis (MS) patients and the association of these markers with disease disability, a 782 base-pair fragment of the TNF-β gene was amplified from genomic DNA and digested with the NcoI restriction enzyme. The serum levels of numerous cytokines (IL-1β, IL-12, IL-6, TNF-α, IFN-γ, IL-4, IL-10, and IL-17) serum lipid levels, plasma insulin levels, and the Homeostasis Model Assessment-Insulin Resistance (HOMA-IR) levels were evaluated in 123 female and 43 male patients with MS. Females carrying the TNFB2/B2 genotype presented with decreased IL-4 and IL-10 levels and increased TNF-α, glucose, insulin, and HOMA-IR levels; moreover, there were positive correlations between EDSS and glucose and between EDSS and HOMA-IR in these females. Males carrying the TNFB2/B2 genotype exhibited increased levels of TNF-α, IFN-γ, and IL-17 (p=0.0326) and decreased levels of IL-4, IL-10, insulin, and HOMA-IR; there was a positive correlation between EDSS and TNF-α levels. The TNFB2/B2 genotype of TNF-β NcoI polymorphism was associated with increased inflammatory and metabolic markers and this association was different according to sex of MS patients.
Collapse
|
25
|
The impact of the human genome project on complex disease. Genes (Basel) 2014; 5:518-35. [PMID: 25032678 PMCID: PMC4198915 DOI: 10.3390/genes5030518] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 06/03/2014] [Accepted: 06/24/2014] [Indexed: 02/06/2023] Open
Abstract
In the decade that has passed since the initial release of the Human Genome, numerous advancements in science and technology within and beyond genetics and genomics have been encouraged and enhanced by the availability of this vast and remarkable data resource. Progress in understanding three common, complex diseases: age-related macular degeneration (AMD), Alzheimer's disease (AD), and multiple sclerosis (MS), are three exemplars of the incredible impact on the elucidation of the genetic architecture of disease. The approaches used in these diseases have been successfully applied to numerous other complex diseases. For example, the heritability of AMD was confirmed upon the release of the first genome-wide association study (GWAS) along with confirmatory reports that supported the findings of that state-of-the art method, thus setting the foundation for future GWAS in other heritable diseases. Following this seminal discovery and applying it to other diseases including AD and MS, the genetic knowledge of AD expanded far beyond the well-known APOE locus and now includes more than 20 loci. MS genetics saw a similar increase beyond the HLA loci and now has more than 100 known risk loci. Ongoing and future efforts will seek to define the remaining heritability of these diseases; the next decade could very well hold the key to attaining this goal.
Collapse
|
26
|
Helal SI, Hegazi A, Al-Menabbawy K. Apitherapy Have a Role in Treatment of Multiple Sclerosis. Open Access Maced J Med Sci 2014. [DOI: 10.3889/oamjms.2014.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
AIM: Multiple sclerosis (MS) is an inflammatory disease in which the fatty myelin sheaths around the axons of the brain and spinal cord are damaged. We Study the effect of Apitherapy in treatment of MS.MATERIAL AND METHODS: Fifty patients with MS, their ages ranged between 26-71 years, were subjected to complete clinical and neurological history and examination to confirm the diagnosis. All cases were under their regular treatment they were divided into two main groups, Group I received honey, pollen, royal jelly and propolis and were treated with apiacupuncture 3 times weekly, for 12 months, in addition to their medical treatment, while group II remains on their ordinary medical treatment only. Apiacupuncture was done by bee stings for regulating the immune system.RESULTS: Results revealed that 4 patients showed some improvement regarding their defects in gait, bowel control, constipation and urination, while 12 cases, showed some mild improvement in their movement in bed, and better improvement in bed sores, sensation, and better motor power, only two cases of them were able to stand for few minutes with support.CONCLUSION: Although Apitherapy is not a curable therapy in MS, but it can be used to minimize the clinical symptoms of MS, and can be included among programs of MS therapy.
Collapse
|
27
|
Biomarkers of mercury exposure in the Amazon. BIOMED RESEARCH INTERNATIONAL 2014; 2014:867069. [PMID: 24895619 PMCID: PMC4020561 DOI: 10.1155/2014/867069] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2013] [Accepted: 04/08/2014] [Indexed: 11/22/2022]
Abstract
Mercury exposure in the Amazon has been studied since the 1980s decade and the assessment of human mercury exposure in the Amazon is difficult given that the natural occurrence of this metal is high and the concentration of mercury in biological samples of this population exceeds the standardized value of normality established by WHO. Few studies have focused on the discovery of mercury biomarkers in the region's population. In this way, some studies have used genetics as well as immunological and cytogenetic tools in order to find a molecular biomarker for assessing the toxicological effect of mercury in the Amazonian population. Most of those studies focused attention on the relation between mercury exposure and autoimmunity and, because of that, they will be discussed in more detail. Here we introduce the general aspects involved with each biomarker that was studied in the region in order to contextualize the reader and add information about the Amazonian life style and health that may be considered for future studies. We hope that, in the future, the toxicological studies in this field use high technological tools, such as the next generation sequencing and proteomics skills, in order to comprehend basic questions regarding the metabolic route of mercury in populations that are under constant exposure, such as in the Amazon.
Collapse
|
28
|
Tumor necrosis factor beta (TNF-β) NcoI polymorphism is associated with multiple sclerosis in Caucasian patients from Southern Brazil independently from HLA-DRB1. J Mol Neurosci 2014; 53:211-21. [DOI: 10.1007/s12031-014-0287-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 03/12/2014] [Indexed: 10/25/2022]
|
29
|
Levin MC, Douglas JN, Meyers L, Lee S, Shin Y, Gardner LA. Neurodegeneration in multiple sclerosis involves multiple pathogenic mechanisms. Degener Neurol Neuromuscul Dis 2014; 4:49-63. [PMID: 32669900 PMCID: PMC7337253 DOI: 10.2147/dnnd.s54391] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 03/06/2014] [Indexed: 12/18/2022] Open
Abstract
Multiple sclerosis (MS) is a complex autoimmune disease that impairs the central nervous system (CNS). The neurological disability and clinical course of the disease is highly variable and unpredictable from one patient to another. The cause of MS is still unknown, but it is thought to occur in genetically susceptible individuals who develop disease due to a nongenetic trigger, such as altered metabolism, a virus, or other environmental factors. MS patients develop progressive, irreversible, neurological disability associated with neuronal and axonal damage, collectively known as neurodegeneration. Neurodegeneration was traditionally considered as a secondary phenomenon to inflammation and demyelination. However, recent data indicate that neurodegeneration develops along with inflammation and demyelination. Thus, MS is increasingly recognized as a neurodegenerative disease triggered by an inflammatory attack of the CNS. While both inflammation and demyelination are well described and understood cellular processes, neurodegeneration might be defined by a diverse pool of any of the following: neuronal cell death, apoptosis, necrosis, and virtual hypoxia. In this review, we present multiple theories and supporting evidence that identify common biological processes that contribute to neurodegeneration in MS.
Collapse
Affiliation(s)
- Michael C Levin
- Veterans Administration Medical Center.,Department of Neuroscience, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Joshua N Douglas
- Veterans Administration Medical Center.,Department of Neuroscience, University of Tennessee Health Science Center, Memphis, TN, USA
| | | | - Sangmin Lee
- Veterans Administration Medical Center.,Department of Neurology
| | - Yoojin Shin
- Veterans Administration Medical Center.,Department of Neurology
| | - Lidia A Gardner
- Veterans Administration Medical Center.,Department of Neurology
| |
Collapse
|
30
|
Dagley LF, Emili A, Purcell AW. Application of quantitative proteomics technologies to the biomarker discovery pipeline for multiple sclerosis. Proteomics Clin Appl 2014; 7:91-108. [PMID: 23112123 DOI: 10.1002/prca.201200104] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Revised: 10/04/2012] [Accepted: 10/11/2012] [Indexed: 11/08/2022]
Abstract
Multiple sclerosis is an inflammatory-mediated demyelinating disorder most prevalent in young Caucasian adults. The various clinical manifestations of the disease present several challenges in the clinic in terms of diagnosis, monitoring disease progression and response to treatment. Advances in MS-based proteomic technologies have revolutionized the field of biomarker research and paved the way for the identification and validation of disease-specific markers. This review focuses on the novel candidates discovered by the application of quantitative proteomics to relevant disease-affected tissues in both the human context and within the animal model of the disease known as experimental autoimmune encephalomyelitis. The role of targeted MS approaches for biomarker validation studies, such as multiple reaction monitoring will also be discussed.
Collapse
Affiliation(s)
- Laura F Dagley
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia
| | | | | |
Collapse
|
31
|
Ooi JH, Waddell A, Lin YD, Albert I, Rust LT, Holden V, Cantorna MT. Dominant effects of the diet on the microbiome and the local and systemic immune response in mice. PLoS One 2014; 9:e86366. [PMID: 24489720 PMCID: PMC3906035 DOI: 10.1371/journal.pone.0086366] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 12/06/2013] [Indexed: 12/22/2022] Open
Abstract
Outside the nutrition community the effects of diet on immune-mediated diseases and experimental outcomes have not been appreciated. Investigators that study immune-mediated diseases and/or the microbiome have overlooked the potential of diet to impact disease phenotype. We aimed to determine the effects of diet on the bacterial microbiota and immune-mediated diseases. Three different laboratory diets were fed to wild-type mice for 2 weeks and resulted in three distinct susceptibilities to dextran sodium sulfate (DSS)-induced colitis. Examination of the fecal microbiota demonstrated a diet-mediated effect on the bacteria found there. Broad-spectrum antibiotics disturbed the gut microbiome and partially eliminated the diet-mediated changes in DSS susceptibility. Dietary changes 2 days after DSS treatment were protective and suggested that the diet-mediated effect occurred quickly. There were no diet-mediated effects on DSS susceptibility in germ-free mice. In addition, the diet-mediated effects were evident in a gastrointestinal infection model (Citrobacter rodentium) and in experimental autoimmune encephalomyelitis. Taken together, our study demonstrates a dominant effect of diet on immune-mediated diseases that act rapidly by changing the microbiota. These findings highlight the potential of using dietary manipulation to control the microbiome and prevent/treat immune-mediated disease.
Collapse
Affiliation(s)
- Jot Hui Ooi
- Center for Molecular Immunology and Infectious Disease, Department of Biochemistry and Molecular Biology, Department of Veterinary and Biomedical Science, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Amanda Waddell
- Center for Molecular Immunology and Infectious Disease, Department of Biochemistry and Molecular Biology, Department of Veterinary and Biomedical Science, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Yang-Ding Lin
- Center for Molecular Immunology and Infectious Disease, Department of Biochemistry and Molecular Biology, Department of Veterinary and Biomedical Science, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Istvan Albert
- Center for Molecular Immunology and Infectious Disease, Department of Biochemistry and Molecular Biology, Department of Veterinary and Biomedical Science, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Laura T. Rust
- Center for Molecular Immunology and Infectious Disease, Department of Biochemistry and Molecular Biology, Department of Veterinary and Biomedical Science, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Victoria Holden
- Center for Molecular Immunology and Infectious Disease, Department of Biochemistry and Molecular Biology, Department of Veterinary and Biomedical Science, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Margherita T. Cantorna
- Center for Molecular Immunology and Infectious Disease, Department of Biochemistry and Molecular Biology, Department of Veterinary and Biomedical Science, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
32
|
|
33
|
Stüve O, Cravens PD, Eagar TN. DNA-based vaccines: the future of multiple sclerosis therapy? Expert Rev Neurother 2014; 8:351-60. [DOI: 10.1586/14737175.8.3.351] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
34
|
Niino M, Kikuchi S, Fukazawa T, Tashiro K. Genetic susceptibility to multiple sclerosis: implications of genetic research on MS therapy. Expert Rev Neurother 2014; 2:329-8. [DOI: 10.1586/14737175.2.3.329] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
35
|
Naegele M, Martin R. The good and the bad of neuroinflammation in multiple sclerosis. HANDBOOK OF CLINICAL NEUROLOGY 2014; 122:59-87. [PMID: 24507513 DOI: 10.1016/b978-0-444-52001-2.00003-0] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Multiple sclerosis (MS) is the most common inflammatory, demyelinating, neurodegenerative disorder of the central nervous system (CNS). It is widely considered a T-cell mediated autoimmune disease that develops in genetically susceptible individuals, possibly under the influence of certain environmental trigger factors. The invasion of autoreactive CD4+ T-cells into the CNS is thought to be a central step that initiates the disease. Several other cell types, including CD8+ T-cells, B-cells and phagocytes appear to be involved in causing inflammation and eventually neurodegeneration. But inflammation is not entirely deleterious in MS. Evidence has accumulated in the recent years that show the importance of regulatory immune mechanisms which restrain tissue damage and initiate regeneration. More insight into the beneficial aspects of neuroinflammation might allow us to develop new treatment strategies for this enigmatic disease.
Collapse
Affiliation(s)
- Matthias Naegele
- Institute for Neuroimmunology and Clinical Multiple Sclerosis Research, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Roland Martin
- Neuroimmunology and MS Research, Neurology Clinic, University Hospital, Zurich, Switzerland.
| |
Collapse
|
36
|
Abstract
Familial aggregation and the studies of twins indicate that heredity contributes to multiple sclerosis (MS) risk. Immunologic studies of leukocyte antigens subsequently followed by gene-mapping techniques identified the primary MS susceptibility locus to be within the major histocompatibility complex (MHC). The primary risk allele is HLA-DRB1*15, although other alleles of this gene also influence MS susceptibility. Other genes within the MHC also contribute to MS susceptibility. Genome-wide association studies have identified over 50 additional common variants of genes across the genome. Estimates suggest that there may be as many as 200 genes involved in MS susceptibility. In addition to these common polymorphisms, studies have identified several rare risk alleles in some families. Interestingly, the majority of the genes identified have known immunologic functions and many contribute to the risk of inheriting other autoimmune diseases. Genetic variants in the vitamin D metabolic pathway have also been identified. That vitamin D contributes to MS susceptibility as both an environmental as well as genetic risk factor underscores the importance of this metabolic pathway in disease pathogenesis. Current efforts are focused on understanding how the myriad of genetic risk alleles interact within networks to influence MS risk at family level as well as within populations.
Collapse
Affiliation(s)
- Bruce A C Cree
- Department of Neurology, University of California, San Francisco, USA.
| |
Collapse
|
37
|
Hartung HP, Steinman L, Goodin DS, Comi G, Cook S, Filippi M, O'Connor P, Jeffery DR, Kappos L, Axtell R, Knappertz V, Bogumil T, Schwenke S, Croze E, Sandbrink R, Pohl C. Interleukin 17F level and interferon β response in patients with multiple sclerosis. JAMA Neurol 2013; 70:1017-21. [PMID: 23732754 DOI: 10.1001/jamaneurol.2013.192] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
IMPORTANCE High serum levels of interleukin 17F (IL-17F) at baseline have been associated with suboptimal response to interferon beta in patients with relapsing-remitting multiple sclerosis. OBJECTIVE To further investigate the role of IL-17F in predicting treatment response to interferon beta-1b in patients with relapsing-remitting multiple sclerosis using the Singulex Erenna IL-17F immunoassay. DESIGN, SETTING, AND PATIENTS Serum samples were analyzed from 239 randomly selected patients treated with interferon beta-1b, 250 μg, for at least 2 years in the Betaferon Efficacy Yielding Outcomes of a New Dose Study. EXPOSURE Treatment with interferon beta-1b, 250 μg, for at least 2 years. MAIN OUTCOME MEASURES Levels of IL-17F at baseline and month 6 as well as the difference between the IL-17F levels at month 6 and baseline were compared between the following: (1) patients with less disease activity vs more disease activity; (2) patients with no disease activity vs some disease activity; and (3) responders vs nonresponders. RESULTS Levels of IL-17F measured at baseline and month 6 did not correlate with lack of response to treatment after 2 years using clinical and magnetic resonance imaging criteria. Relapses and new lesions on magnetic resonance imaging were not associated with pretreatment serum IL-17F levels. When patients with neutralizing antibodies were excluded, the results did not change. All patients with levels of IL-17F greater than 200 pg/mL were associated with poor response with some clinical or radiological activity. CONCLUSIONS AND RELEVANCE An increase of IL-17F before and early after treatment with interferon beta-1b was not associated with poor response. These data do not support the value of IL-17F as a treatment response indicator for therapy of patients with multiple sclerosis with interferon beta, although high levels of IL-17F greater than 200 pg/mL may predict nonresponsiveness.
Collapse
Affiliation(s)
- Hans-Peter Hartung
- Department of Neurology, Heinrich-Heine-Universität, Düsseldorf, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Different mechanisms of inflammation induced in virus and autoimmune-mediated models of multiple sclerosis in C57BL6 mice. BIOMED RESEARCH INTERNATIONAL 2013; 2013:589048. [PMID: 24083230 PMCID: PMC3780522 DOI: 10.1155/2013/589048] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2013] [Accepted: 07/01/2013] [Indexed: 01/17/2023]
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the human central nervous system (CNS). Neurotropic demyelinating strain of MHV (MHV-A59 or its isogenic recombinant strain RSA59) induces MS-like disease in mice mediated by microglia, along with a small population of T cells. The mechanism of demyelination is at least in part due to microglia-mediated myelin stripping, with some direct axonal injury. Immunization with myelin oligodendrocyte glycoprotein (MOG) induces experimental autoimmune encephalomyelitis (EAE), a mainly CD4+ T-cell-mediated disease, although CD8+ T cells may play a significant role in demyelination. It is possible that both autoimmune and nonimmune mechanisms such as direct viral toxicity may induce MS. Our study directly compares CNS pathology in autoimmune and viral-induced MS models. Mice with viral-induced and EAE demyelinating diseases demonstrated similar patterns and distributions of demyelination that accumulated over the course of the disease. However, significant differences in acute inflammation were noted. Inflammation was restricted mainly to white matter at all times in EAE, whereas inflammation initially largely involved gray matter in acute MHV-induced disease and then is subsequently localized only in white matter in the chronic disease phase. The presence of dual mechanisms of demyelination may be responsible for the failure of immunosuppression to promote long-term remission in many MS patients.
Collapse
|
39
|
Microglia play a major role in direct viral-induced demyelination. Clin Dev Immunol 2013; 2013:510396. [PMID: 23864878 PMCID: PMC3705805 DOI: 10.1155/2013/510396] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Revised: 05/14/2013] [Accepted: 05/15/2013] [Indexed: 11/17/2022]
Abstract
Microglia are the resident macrophage-like populations in the central nervous system (CNS). Microglia remain quiescent, unable to perform effector and antigen presentation (APC) functions until activated by injury or infection, and have been suggested to represent the first line of defence for the CNS. Previous studies demonstrated that microglia can be persistently infected by neurotropic mouse hepatitis virus (MHV) which causes meningoencephalitis, myelitis with subsequent axonal loss, and demyelination and serve as a virus-induced model of human neurological disease multiple sclerosis (MS). Current studies revealed that MHV infection is associated with the pronounced activation of microglia during acute inflammation, as evidenced by characteristic changes in cellular morphology and increased expression of microglia-specific proteins, Iba1 (ionized calcium-binding adaptor molecule 1), which is a macrophage/microglia-specific novel calcium-binding protein and involved in membrane ruffling and phagocytosis. During chronic inflammation (day 30 postinfection), microglia were still present within areas of demyelination. Experiments performed in ex vivo spinal cord slice culture and in vitro neonatal microglial culture confirmed direct microglial infection. Our results suggest that MHV can directly infect and activate microglia during acute inflammation, which in turn during chronic inflammation stage causes phagocytosis of myelin sheath leading to chronic inflammatory demyelination.
Collapse
|
40
|
Goris A, Pauwels I, Dubois B. Progress in multiple sclerosis genetics. Curr Genomics 2013; 13:646-63. [PMID: 23730204 PMCID: PMC3492804 DOI: 10.2174/138920212803759695] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Revised: 09/20/2012] [Accepted: 09/24/2012] [Indexed: 01/06/2023] Open
Abstract
A genetic component in the susceptibility to multiple sclerosis (MS) has long been known, and the first and major genetic risk factor, the HLA region, was identified in the 1970’s. However, only with the advent of genome-wide association studies in the past five years did the list of risk factors for MS grow from 1 to over 50. In this review, we summarize the search for MS risk genes and the latest results. Comparison with data from other autoimmune and neurological diseases and from animal models indicates parallels and differences between diseases. We discuss how these translate into an improved understanding of disease mechanisms, and address current challenges such as genotype-phenotype correlations, functional mechanisms of risk variants and the missing heritability.
Collapse
Affiliation(s)
- An Goris
- Laboratory for Neuroimmunology, Section of Experimental Neurology, KU Leuven, Leuven, Belgium
| | | | | |
Collapse
|
41
|
Genetic variants of FOXP3 influence graft survival in kidney transplant patients. Hum Immunol 2013; 74:751-7. [DOI: 10.1016/j.humimm.2013.02.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Revised: 01/17/2013] [Accepted: 02/19/2013] [Indexed: 01/03/2023]
|
42
|
Swaminathan B, Cuapio A, Alloza I, Matesanz F, Alcina A, García-Barcina M, Fedetz M, Fernández Ó, Lucas M, Órpez T, Pinto-Medel MJ, Otaegui D, Olascoaga J, Urcelay E, Ortiz MA, Arroyo R, Oksenberg JR, Antigüedad A, Tolosa E, Vandenbroeck K. Fine mapping and functional analysis of the multiple sclerosis risk gene CD6. PLoS One 2013; 8:e62376. [PMID: 23638056 PMCID: PMC3634811 DOI: 10.1371/journal.pone.0062376] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Accepted: 03/22/2013] [Indexed: 12/18/2022] Open
Abstract
CD6 has recently been identified and validated as risk gene for multiple sclerosis (MS), based on the association of a single nucleotide polymorphism (SNP), rs17824933, located in intron 1. CD6 is a cell surface scavenger receptor involved in T-cell activation and proliferation, as well as in thymocyte differentiation. In this study, we performed a haptag SNP screen of the CD6 gene locus using a total of thirteen tagging SNPs, of which three were non-synonymous SNPs, and replicated the recently reported GWAS SNP rs650258 in a Spanish-Basque collection of 814 controls and 823 cases. Validation of the six most strongly associated SNPs was performed in an independent collection of 2265 MS patients and 2600 healthy controls. We identified association of haplotypes composed of two non-synonymous SNPs [rs11230563 (R225W) and rs2074225 (A257V)] in the 2nd SRCR domain with susceptibility to MS (Pmax(T) permutation = 1×10−4). The effect of these haplotypes on CD6 surface expression and cytokine secretion was also tested. The analysis showed significantly different CD6 expression patterns in the distinct cell subsets, i.e. – CD4+ naïve cells, P = 0.0001; CD8+ naïve cells, P<0.0001; CD4+ and CD8+ central memory cells, P = 0.01 and 0.05, respectively; and natural killer T (NKT) cells, P = 0.02; with the protective haplotype (RA) showing higher expression of CD6. However, no significant changes were observed in natural killer (NK) cells, effector memory and terminally differentiated effector memory T cells. Our findings reveal that this new MS-associated CD6 risk haplotype significantly modifies expression of CD6 on CD4+ and CD8+ T cells.
Collapse
MESH Headings
- Adult
- Antigens, CD/chemistry
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Antigens, Differentiation, T-Lymphocyte/chemistry
- Antigens, Differentiation, T-Lymphocyte/genetics
- Antigens, Differentiation, T-Lymphocyte/metabolism
- CD4-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/metabolism
- Chromosome Mapping
- Cluster Analysis
- Cytokines/metabolism
- Female
- Gene Order
- Genetic Loci
- Genetic Predisposition to Disease
- Haplotypes
- Humans
- Linkage Disequilibrium
- Lymphocyte Activation/immunology
- Male
- Multiple Sclerosis/genetics
- Multiple Sclerosis/metabolism
- Polymorphism, Single Nucleotide
- Protein Interaction Domains and Motifs
- Spain
- White People/genetics
- Young Adult
Collapse
Affiliation(s)
- Bhairavi Swaminathan
- Neurogenomiks Laboratory, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Angélica Cuapio
- Department of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Iraide Alloza
- Neurogenomiks Laboratory, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Fuencisla Matesanz
- Instituto de Parasitología y Biomedicina “López Neyra” Consejo Superior de Investigaciones Científicas (CSIC), Granada, Spain
| | - Antonio Alcina
- Instituto de Parasitología y Biomedicina “López Neyra” Consejo Superior de Investigaciones Científicas (CSIC), Granada, Spain
| | | | - Maria Fedetz
- Instituto de Parasitología y Biomedicina “López Neyra” Consejo Superior de Investigaciones Científicas (CSIC), Granada, Spain
| | - Óscar Fernández
- Department of Neurology, Institute of Clinical Neurosciences, Hospital Regional Universitario Carlos Haya, Málaga, Spain
| | - Miguel Lucas
- Unidad de Esclerosis Múltiple, Hospital Virgen Macarena, Sevilla, Spain
| | - Teresa Órpez
- Research Laboratory, Institute of Clinical Neurosciences, Hospital Regional Universitario Carlos Haya, Málaga, Spain
| | - Mª Jesus Pinto-Medel
- Research Laboratory, Institute of Clinical Neurosciences, Hospital Regional Universitario Carlos Haya, Málaga, Spain
| | - David Otaegui
- Área de Neurociencias, Instituto de Investigación Sanitaria Biodonostia, San Sebastián, Spain
| | - Javier Olascoaga
- Servicio de Neurología, Unidad de Esclerosis Múltiple, Hospital Donostia, San Sebastián, Spain
| | - Elena Urcelay
- Immunology Department H. Clínico S. Carlos, Instituto de Investigación Sanitaria S. Carlos (IdISSC), Madrid, Spain
| | - Miguel A. Ortiz
- Immunology Department H. Clínico S. Carlos, Instituto de Investigación Sanitaria S. Carlos (IdISSC), Madrid, Spain
| | - Rafael Arroyo
- Multiple Sclerosis Unit, Neurology Department H. Clínico S. Carlos, Instituto de Investigación Sanitaria S. Carlos (IdISSC), Madrid, Spain
| | - Jorge R. Oksenberg
- Department of Neurology, University of California San Francisco, San Francisco, California, United States of America
| | | | - Eva Tolosa
- Department of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Koen Vandenbroeck
- Neurogenomiks Laboratory, University of the Basque Country (UPV/EHU), Leioa, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
- * E-mail:
| |
Collapse
|
43
|
Gourraud PA, Harbo HF, Hauser SL, Baranzini SE. The genetics of multiple sclerosis: an up-to-date review. Immunol Rev 2012. [PMID: 22725956 DOI: 10.1111/j.1600-065x.2012.01134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Multiple sclerosis (MS) is a prevalent inflammatory disease of the central nervous system that often leads to disability in young adults. Treatment options are limited and often only partly effective. The disease is likely caused by a complex interaction between multiple genes and environmental factors, leading to inflammatory-mediated central nervous system deterioration. A series of genomic studies have confirmed a central role for the immune system in the development of MS, including genetic association studies that have now dramatically expanded the roster of MS susceptibility genes beyond the longstanding human leukocyte antigen (HLA) association in MS first identified nearly 40 years ago. Advances in technology together with novel models for collaboration across research groups have enabled the discovery of more than 50 non-HLA genetic risk factors associated with MS. However, with a large proportion of the disease heritability still unaccounted for, current studies are now geared towards identification of causal alleles, associated pathways, epigenetic mechanisms, and gene-environment interactions. This article reviews recent efforts in addressing the genetics of MS and the challenges posed by an ever increasing amount of analyzable data, which is spearheading development of novel statistical methods necessary to cope with such complexity.
Collapse
Affiliation(s)
- Pierre-Antoine Gourraud
- Department of Neurology, University of California San Francisco, San Francisco, CA 94143-0435, USA
| | | | | | | |
Collapse
|
44
|
Fontaine B. [Will multiple sclerosis (MS) susceptibility genes one day become biomarkers for MS diagnosis?]. Rev Neurol (Paris) 2012; 168:833-5. [PMID: 22959703 DOI: 10.1016/j.neurol.2012.07.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Over the past 5 years, studies of multiple sclerosis (MS) genes have made spectacular progress. Today, more than 50 genes have been found to have an established role in MS, and there is no doubt that this list will continue to grow. The genes identified so far point to the metabolic pathways involving the immune system, thereby suggesting than inflammation is an early process in the course of the disease and probably the major cause of the axonal degeneration that results in neurological deficiencies and handicap. They also raise the question of the usefulness of these biomarkers in the diagnosis of MS.
Collapse
Affiliation(s)
- B Fontaine
- INSERM, CNRS, université Pierre-et-Marie-Curie, Assistance publique-Hôpitaux de Paris, Paris, France.
| |
Collapse
|
45
|
Abstract
Multiple sclerosis (MS) is a prevalent inflammatory disease of the central nervous system that often leads to disability in young adults. Treatment options are limited and often only partly effective. The disease is likely caused by a complex interaction between multiple genes and environmental factors, leading to inflammatory-mediated central nervous system deterioration. A series of genomic studies have confirmed a central role for the immune system in the development of MS, including genetic association studies that have now dramatically expanded the roster of MS susceptibility genes beyond the longstanding human leukocyte antigen (HLA) association in MS first identified nearly 40 years ago. Advances in technology together with novel models for collaboration across research groups have enabled the discovery of more than 50 non-HLA genetic risk factors associated with MS. However, with a large proportion of the disease heritability still unaccounted for, current studies are now geared towards identification of causal alleles, associated pathways, epigenetic mechanisms, and gene-environment interactions. This article reviews recent efforts in addressing the genetics of MS and the challenges posed by an ever increasing amount of analyzable data, which is spearheading development of novel statistical methods necessary to cope with such complexity.
Collapse
Affiliation(s)
- Pierre-Antoine Gourraud
- Department of Neurology, University of California San Francisco. 513 Parnassus Ave. Room S-256. San Francisco, CA. 94143-0435’
| | - Hanne F. Harbo
- Department of Neurology, University of California San Francisco. 513 Parnassus Ave. Room S-256. San Francisco, CA. 94143-0435’
- Department of Neurology, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Stephen L. Hauser
- Department of Neurology, University of California San Francisco. 513 Parnassus Ave. Room S-256. San Francisco, CA. 94143-0435’
| | - Sergio E. Baranzini
- Department of Neurology, University of California San Francisco. 513 Parnassus Ave. Room S-256. San Francisco, CA. 94143-0435’
| |
Collapse
|
46
|
CRYAB-650 C>G (rs2234702) affects susceptibility to Type 1 diabetes and IAA-positivity in Swedish population. Hum Immunol 2012; 73:759-66. [DOI: 10.1016/j.humimm.2012.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Revised: 04/06/2012] [Accepted: 04/16/2012] [Indexed: 10/28/2022]
|
47
|
Recombinant T-Cell Receptor Ligand (RTL) for Treatment of Multiple Sclerosis: A Double-Blind, Placebo-Controlled, Phase 1, Dose-Escalation Study. Autoimmune Dis 2012; 2012:954739. [PMID: 22548151 PMCID: PMC3328144 DOI: 10.1155/2012/954739] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Revised: 01/15/2012] [Accepted: 01/16/2012] [Indexed: 11/23/2022] Open
Abstract
Background. Recombinant T-cell receptor ligand 1000 (RTL1000) is a single-chain protein construct containing the outer two domains of HLA-DR2 linked to myelin-oligodendrocyte-glycoprotein- (MOG-) 35–55 peptide. Analogues of RTL1000 induce T-cell tolerance, reverse clinical and histological disease, and promote repair in experimental autoimmune encephalomyelitis (EAE) in DR2 transgenic, C57BL/6, and SJL/J mice. Objective. Determining the maximum tolerated dose, safety, and tolerability of RTL1000 in multiple sclerosis (MS) subjects. Methods. This was a multicenter, Phase I dose-escalation study in HLA-DR2+ MS subjects. Consecutive cohorts received RTL1000 doses of 2, 6, 20, 60, 200, and 100 mg, respectively. Subjects within each cohort randomly received a single intravenous infusion of RTL1000 or placebo at a 4 : 2 ratio. Safety monitoring included clinical, laboratory, and brain magnetic resonance imaging (MRI) evaluations. Results. Thirty-four subjects completed the protocol. All subjects tolerated the 2–60 mg doses of RTL1000. Doses ≥100 mg caused hypotension and diarrhea in 3 of 4 subjects, leading to discontinuation of further enrollment. Conclusions. The maximum tolerated dose of RTL1000 in MS subjects is 60 mg, comparable to effective RTL doses in EAE. RTL1000 is a novel approach for MS treatment that may induce immunoregulation without immunosuppression and promote neural repair.
Collapse
|
48
|
Al Jumah M, Al Balwi M, Hussein M, Kojan S, Al Khathaami A, Al Fawaz M, Al Muzaini B, Jawhary A, Al Abdulkareem I. Association of SNPs rs6498169 and rs10984447 with multiple sclerosis in Saudi patients: a model of the usefulness of familial aggregates in identifying genetic linkage in a multifactorial disease. Mult Scler 2012; 18:1395-400. [PMID: 22492128 DOI: 10.1177/1352458512440832] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
OBJECTIVE Genome-Wide association studies (GWAS) showed an association between subset of single-nucleotide polymorphism (SNPs) and multiple sclerosis. Our study aims to study this association in Saudi familial multiple sclerosis patients. METHODS Four subject groups were used in this study: sporadic MS (MS patients without family history), FMS (MS patients who have at least one family member diagnosed with MS), related controls (relatives of FMS patients who appear to be free of the disease) and independent controls (healthy volunteers). Subjects were genotyped for 15 SNPs. The variation in the genotype distribution was analyzed across study groups by using logistic regression. RESULTS 342 subjects were included. 99 were in the sporadic MS group, 22 were FMS, 89 were related control, and 132 were independent control. SNPS rs3135388, rs7577363, rs1321172, rs6897932, rs6498169, rs12487066, and rs4763655were associated with MS when MS and independent control groups were compared. Same SNPS were identified but with stronger association when the FMS and independent control groups were compared. Finally, when the patients and the controls were selected from a much more homogenous genetic pool from which it would be expected that only SNPs highly linked to MS would persist, only two SNPs rs6498169[OR 4.26, CI (1.17 - 15.51)];, and rs10984447 [OR 13.63, CI(1.54, 120.83) ][were associated with MS. CONCLUSIONS Our results suggest that using a more homogenous genetic pool of cases and controls could help to identify the most significant MS-associated SNPs. Our finding is in agreement with other studies including larger sample size and more diverse populations.
Collapse
Affiliation(s)
- M Al Jumah
- King Abdullah International Medical Research Center, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia.
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Genetic factors of autoimmune thyroid diseases in Japanese. Autoimmune Dis 2012; 2012:236981. [PMID: 22242199 PMCID: PMC3254007 DOI: 10.1155/2012/236981] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Revised: 10/31/2011] [Accepted: 10/31/2011] [Indexed: 11/17/2022] Open
Abstract
Autoimmune thyroid diseases (AITDs), including Graves' disease (GD) and Hashimoto's thyroiditis (HT), are caused by immune response to self-thyroid antigens and affect approximately 2–5% of the general population. Genetic susceptibility in combination with external factors, such as smoking, viral/bacterial infection, and chemicals, is believed to initiate the autoimmune response against thyroid antigens. Abundant epidemiological data, including family and twin studies, point to a strong genetic influence on the development of AITDs. Various techniques have been employed to identify genes contributing to the etiology of AITDs, including candidate gene analysis and whole genome screening. These studies have enabled the identification of several loci (genetic regions) that are linked to AITDs, and, in some of these loci, putative AITD susceptibility genes have been identified. Some of these genes/loci are unique to GD and HT and some are common to both diseases, indicating that there is a shared genetic susceptibility to GD and HT. Known AITD-susceptibility genes are classified into three groups: HLA genes, non-HLA immune-regulatory genes (e.g., CTLA-4, PTPN22, and CD40), and thyroid-specific genes (e.g., TSHR and Tg). In this paper, we will summarize the latest findings on AITD susceptibility genes in Japanese.
Collapse
|
50
|
Broux B, Pannemans K, Zhang X, Markovic-Plese S, Broekmans T, Eijnde BO, Van Wijmeersch B, Somers V, Geusens P, van der Pol S, van Horssen J, Stinissen P, Hellings N. CX(3)CR1 drives cytotoxic CD4(+)CD28(-) T cells into the brain of multiple sclerosis patients. J Autoimmun 2011; 38:10-9. [PMID: 22123179 DOI: 10.1016/j.jaut.2011.11.006] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Revised: 11/10/2011] [Accepted: 11/12/2011] [Indexed: 10/25/2022]
Abstract
Immunosenescence, or ageing of the immune system, contributes to the increased morbidity and mortality seen in the elderly population. Premature immunosenescence is shown to occur in a subgroup of patients with autoimmune diseases. One of the main characteristics of immunosenescence is the expansion of CD4(+)CD28(-) T cells in the blood. In this study, we investigate the potential contribution of these cells to disease processes in a subgroup of multiple sclerosis (MS) and rheumatoid arthritis (RA) patients. Characterization of CD4(+)CD28(-) T cells in patients and healthy controls reveals that they have an inflammation-seeking effector-memory T cell phenotype with cytotoxic properties, as they expel cytotoxic granules in response to a polyclonal stimulus or MS-related autoantigens. We identify CX(3)CR1, the fractalkine receptor, as a selective marker to discriminate CD4(+)CD28(-) T cells from their CD4(+)CD28(+) counterparts. CX(3)CR1 expression enables CD4(+)CD28(-) T cells to migrate towards a fractalkine gradient in vitro. In addition, we find increased levels of fractalkine in the cerebrospinal fluid and inflammatory lesions of MS patients. We demonstrate for the first time that CD4(+)CD28(-) T cells accumulate in MS lesions of a subgroup of patients. Moreover, we have indications that these cells are cytotoxic in the target tissue. Overall, our findings suggest that CD4(+)CD28(-) T cells migrate in response to a chemotactic gradient of fractalkine to sites of inflammation, where they contribute to the inflammatory processes in a subgroup of patients with MS and RA.
Collapse
Affiliation(s)
- Bieke Broux
- Hasselt University, Biomedical Research Institute and transnationale Universiteit Limburg, School of Life Sciences, Agoralaan, Building C, 3590 Diepenbeek, Belgium.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|