101
|
Triantafyllou N, Thoda P, Armeni E, Rizos D, Kaparos G, Augoulea A, Alexandrou A, Creatsa M, Tsivgoulis G, Artemiades A, Panoulis C, Lambrinoudaki I. Association of sex hormones and glucose metabolism with the severity of multiple sclerosis. Int J Neurosci 2015; 126:797-804. [PMID: 26407165 DOI: 10.3109/00207454.2015.1069825] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE/AIM OF THE STUDY We evaluated possible associations between the severity of multiple sclerosis (MS) and levels of sex hormones as well as biochemical parameters in a sample of ambulatory patients. MATERIAL AND METHODS This cross-sectional study recruited 133 adults (52 men, 66 premenopausal and 15 postmenopausal women), with relapsing-remitting MS. Fasting venous blood samples were drawn for biochemical and hormonal evaluation. These parameters were tested for possible associations with MS severity, assessed using the Expanded Disability Status Scale (EDSS)-scores. RESULTS Follicle-stimulating hormone correlated with mean EDSS scores (r = -0.369, p = 0.038) in the premenopausal subgroup. However, this association became non-significant in the age-adjusted multivariate analysis (p = 0.141; power = 67%, type α error 0.10). Free androgen exhibited a borderline negative effect on EDSS-scores in the subgroup of men (r = -0.367, p = 0.093), which was lost after adjusting for age and duration of disease (p = 0.192; statistical power = 93%, type α error 0.05). Levels of estradiol tended to affect disability status of postmenopausal women (normal-mild vs. severe impairment: 23.33 ± 11.73pg/mL vs. 14.74 ± 6.30pg/mL, p = 0.095). Levels of sex hormones or indices of glycemic metabolism did not differ between patients presenting with EDSS scores higher or lower than the median value. CONCLUSION Sex hormones and indices of glucose metabolism exhibited only a middle effect on EDSS scoring, which was not independent from the presence of confounders like age and duration of MS. The present study highlights the need for additional research, in order to elucidate the role of sex hormones and insulin resistance in the course of MS.
Collapse
Affiliation(s)
- Nikolaos Triantafyllou
- a 1 1st Department of Neurology , University of Athens , Aiginiteio Hospital, Athens , Greece
| | - Pinelopi Thoda
- b 2 2nd Department of Obstetrics and Gynecology , University of Athens , Aretaieio Hospital, Athens , Greece
| | - Eleni Armeni
- b 2 2nd Department of Obstetrics and Gynecology , University of Athens , Aretaieio Hospital, Athens , Greece
| | - Demetrios Rizos
- c 3 Hormonal and Biochemical Laboratory , University of Athens , Aretaieio Hospital, Athens , Greece
| | - George Kaparos
- c 3 Hormonal and Biochemical Laboratory , University of Athens , Aretaieio Hospital, Athens , Greece
| | - Areti Augoulea
- b 2 2nd Department of Obstetrics and Gynecology , University of Athens , Aretaieio Hospital, Athens , Greece
| | - Andreas Alexandrou
- b 2 2nd Department of Obstetrics and Gynecology , University of Athens , Aretaieio Hospital, Athens , Greece
| | - Maria Creatsa
- b 2 2nd Department of Obstetrics and Gynecology , University of Athens , Aretaieio Hospital, Athens , Greece
| | - Georgios Tsivgoulis
- d 4 2nd Department of Neurology , University of Athens , Attiko Hospital, Athens , Greece
| | - Artemios Artemiades
- a 1 1st Department of Neurology , University of Athens , Aiginiteio Hospital, Athens , Greece
| | - Constantinos Panoulis
- b 2 2nd Department of Obstetrics and Gynecology , University of Athens , Aretaieio Hospital, Athens , Greece
| | - Irene Lambrinoudaki
- b 2 2nd Department of Obstetrics and Gynecology , University of Athens , Aretaieio Hospital, Athens , Greece
| |
Collapse
|
102
|
Kempe P, Hammar M, Brynhildsen J. Symptoms of multiple sclerosis during use of combined hormonal contraception. Eur J Obstet Gynecol Reprod Biol 2015. [PMID: 26196655 DOI: 10.1016/j.ejogrb.2015.06.030] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
OBJECTIVE The incidence and disease course of multiple sclerosis (MS) is influenced by sex steroids, and several studies have shown less disease activity during high estrogen states. We have previously shown variation in symptom experience related to the estrogen/progestogen phase in women using combined hormonal contraceptives (CHC) in a small sample. The aim of this study was to confirm these results in a larger sample. STUDY DESIGN Self-assessment of symptoms of MS in relation to CHC cycle by 22 female MS patients. A symptom diary based on a validated instrument for cyclical symptoms was used. Mean symptom scores for high and low estrogen/progestogen phases were compared. RESULTS The women scored four out of ten symptoms significantly higher during the pill-free week than during the CHC phase (p<.05). CONCLUSION Women with MS report more pronounced symptoms during the pill-free, low-estrogen/progestogen phase of CHC use. Future studies should investigate, with a prospective, controlled design, the effects that continuous-use regimens of CHC have in women with MS.
Collapse
Affiliation(s)
- Per Kempe
- Division of Obstetrics and Gynecology, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, S-581 85 Linköping, Sweden; Department of Obstetrics & Gynecology, County Hospital Sundsvall, Sweden.
| | - Mats Hammar
- Division of Obstetrics and Gynecology, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, S-581 85 Linköping, Sweden
| | - Jan Brynhildsen
- Division of Obstetrics and Gynecology, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, S-581 85 Linköping, Sweden
| |
Collapse
|
103
|
Abstract
Multiple sclerosis (MS) is a major neurologic disorder which preferentially affects young women of childbearing age. In the last two decades, a number of disease-modifying therapies have become available to treat relapsing forms of MS. None of these agents is approved for use in pregnancy. The timing of treatment versus conception, and risk of drug pregnancy exposures, are frequent discussion topics when caring for MS patients. This editorial will try to put into context available data, approaches, controversies and future needs.
Collapse
Affiliation(s)
- Patricia K Coyle
- Stony Brook University, Stony Brook MS Comprehensive Care Center, Department of Neurology , Stony Brook, NY , USA +1 631 444 8188 ; +1 631 444 1474 ;
| |
Collapse
|
104
|
Pozzilli C, De Giglio L, Barletta VT, Marinelli F, Angelis FD, Gallo V, Pagano VA, Marini S, Piattella MC, Tomassini V, Pantano P. Oral contraceptives combined with interferon β in multiple sclerosis. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2015; 2:e120. [PMID: 26140279 PMCID: PMC4476053 DOI: 10.1212/nxi.0000000000000120] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 05/04/2015] [Indexed: 01/28/2023]
Abstract
Objective: To test the effect of oral contraceptives (OCs) in combination with interferon β (IFN-β) on disease activity in patients with relapsing-remitting multiple sclerosis (RRMS). Methods: One hundred fifty women with RRMS were randomized in a 1:1:1 ratio to receive IFN-β-1a subcutaneously (SC) only (group 1), IFN-β-1a SC plus ethinylstradiol 20 μg and desogestrel 150 μg (group 2), or IFN-β-1a SC plus ethinylestradiol 40 μg and desogestrel 125 μg (group 3). The primary endpoint was the cumulative number of combined unique active (CUA) lesions on brain MRI at week 96. Secondary endpoints included MRI and clinical and safety measures. Results: The estimated number of cumulative CUA lesions at week 96 was 0.98 (95% confidence interval [CI] 0.81–1.14) in group 1, 0.84 (95% CI 0.66–1.02) in group 2, and 0.72 (95% CI 0.53–0.91) in group 3, with a decrease of 14.1% (p = 0.24) and 26.5% (p = 0.04) when comparing group 1 with groups 2 and 3, respectively. The number of patients with no gadolinium-enhancing lesions was greater in group 3 than in group 1 (p = 0.03). No significant differences were detected in other secondary endpoints. IFN-β or OC discontinuations were equally distributed across groups. Conclusions: Our results translate the observations derived from experimental models to patients, supporting the anti-inflammatory effects of OCs with high-dose estrogens, and suggest possible directions for future research. Classification of evidence: This study provides Class II evidence that in women with RRMS, IFN-β plus ethinylstradiol and desogestrel decreases the cumulative number of active brain MRI lesions compared with IFN-β alone.
Collapse
Affiliation(s)
- Carlo Pozzilli
- Department of Neurology and Psychiatry (C.P., L.D.G., F.D.A., M.C.P., V.T., P.P.), Sapienza University of Rome, Italy; MS Center (C.P., L.D.G., V.T.B., F.M.), S. Andrea Hospital, Sapienza University of Rome, Italy; Centre for Primary Care and Public Health (V.G.), Queen Mary University of London, UK; TFS Trial Form Support S.L. (V.A.P., S.M.), Rome, Italy; Institute of Psychological Medicine and Clinical Neurosciences (V.T.), Cardiff University School of Medicine University Hospital of Wales (V.T.), Cardiff, UK; Santa Lucia Foundation (V.T.), Rome, Italy; and IRCCS Neuromed (P.P.), Pozzilli (IS), Italy
| | - Laura De Giglio
- Department of Neurology and Psychiatry (C.P., L.D.G., F.D.A., M.C.P., V.T., P.P.), Sapienza University of Rome, Italy; MS Center (C.P., L.D.G., V.T.B., F.M.), S. Andrea Hospital, Sapienza University of Rome, Italy; Centre for Primary Care and Public Health (V.G.), Queen Mary University of London, UK; TFS Trial Form Support S.L. (V.A.P., S.M.), Rome, Italy; Institute of Psychological Medicine and Clinical Neurosciences (V.T.), Cardiff University School of Medicine University Hospital of Wales (V.T.), Cardiff, UK; Santa Lucia Foundation (V.T.), Rome, Italy; and IRCCS Neuromed (P.P.), Pozzilli (IS), Italy
| | - Valeria T Barletta
- Department of Neurology and Psychiatry (C.P., L.D.G., F.D.A., M.C.P., V.T., P.P.), Sapienza University of Rome, Italy; MS Center (C.P., L.D.G., V.T.B., F.M.), S. Andrea Hospital, Sapienza University of Rome, Italy; Centre for Primary Care and Public Health (V.G.), Queen Mary University of London, UK; TFS Trial Form Support S.L. (V.A.P., S.M.), Rome, Italy; Institute of Psychological Medicine and Clinical Neurosciences (V.T.), Cardiff University School of Medicine University Hospital of Wales (V.T.), Cardiff, UK; Santa Lucia Foundation (V.T.), Rome, Italy; and IRCCS Neuromed (P.P.), Pozzilli (IS), Italy
| | - Fabiana Marinelli
- Department of Neurology and Psychiatry (C.P., L.D.G., F.D.A., M.C.P., V.T., P.P.), Sapienza University of Rome, Italy; MS Center (C.P., L.D.G., V.T.B., F.M.), S. Andrea Hospital, Sapienza University of Rome, Italy; Centre for Primary Care and Public Health (V.G.), Queen Mary University of London, UK; TFS Trial Form Support S.L. (V.A.P., S.M.), Rome, Italy; Institute of Psychological Medicine and Clinical Neurosciences (V.T.), Cardiff University School of Medicine University Hospital of Wales (V.T.), Cardiff, UK; Santa Lucia Foundation (V.T.), Rome, Italy; and IRCCS Neuromed (P.P.), Pozzilli (IS), Italy
| | - Floriana De Angelis
- Department of Neurology and Psychiatry (C.P., L.D.G., F.D.A., M.C.P., V.T., P.P.), Sapienza University of Rome, Italy; MS Center (C.P., L.D.G., V.T.B., F.M.), S. Andrea Hospital, Sapienza University of Rome, Italy; Centre for Primary Care and Public Health (V.G.), Queen Mary University of London, UK; TFS Trial Form Support S.L. (V.A.P., S.M.), Rome, Italy; Institute of Psychological Medicine and Clinical Neurosciences (V.T.), Cardiff University School of Medicine University Hospital of Wales (V.T.), Cardiff, UK; Santa Lucia Foundation (V.T.), Rome, Italy; and IRCCS Neuromed (P.P.), Pozzilli (IS), Italy
| | - Valentina Gallo
- Department of Neurology and Psychiatry (C.P., L.D.G., F.D.A., M.C.P., V.T., P.P.), Sapienza University of Rome, Italy; MS Center (C.P., L.D.G., V.T.B., F.M.), S. Andrea Hospital, Sapienza University of Rome, Italy; Centre for Primary Care and Public Health (V.G.), Queen Mary University of London, UK; TFS Trial Form Support S.L. (V.A.P., S.M.), Rome, Italy; Institute of Psychological Medicine and Clinical Neurosciences (V.T.), Cardiff University School of Medicine University Hospital of Wales (V.T.), Cardiff, UK; Santa Lucia Foundation (V.T.), Rome, Italy; and IRCCS Neuromed (P.P.), Pozzilli (IS), Italy
| | - Veronica A Pagano
- Department of Neurology and Psychiatry (C.P., L.D.G., F.D.A., M.C.P., V.T., P.P.), Sapienza University of Rome, Italy; MS Center (C.P., L.D.G., V.T.B., F.M.), S. Andrea Hospital, Sapienza University of Rome, Italy; Centre for Primary Care and Public Health (V.G.), Queen Mary University of London, UK; TFS Trial Form Support S.L. (V.A.P., S.M.), Rome, Italy; Institute of Psychological Medicine and Clinical Neurosciences (V.T.), Cardiff University School of Medicine University Hospital of Wales (V.T.), Cardiff, UK; Santa Lucia Foundation (V.T.), Rome, Italy; and IRCCS Neuromed (P.P.), Pozzilli (IS), Italy
| | - Stefano Marini
- Department of Neurology and Psychiatry (C.P., L.D.G., F.D.A., M.C.P., V.T., P.P.), Sapienza University of Rome, Italy; MS Center (C.P., L.D.G., V.T.B., F.M.), S. Andrea Hospital, Sapienza University of Rome, Italy; Centre for Primary Care and Public Health (V.G.), Queen Mary University of London, UK; TFS Trial Form Support S.L. (V.A.P., S.M.), Rome, Italy; Institute of Psychological Medicine and Clinical Neurosciences (V.T.), Cardiff University School of Medicine University Hospital of Wales (V.T.), Cardiff, UK; Santa Lucia Foundation (V.T.), Rome, Italy; and IRCCS Neuromed (P.P.), Pozzilli (IS), Italy
| | - Maria C Piattella
- Department of Neurology and Psychiatry (C.P., L.D.G., F.D.A., M.C.P., V.T., P.P.), Sapienza University of Rome, Italy; MS Center (C.P., L.D.G., V.T.B., F.M.), S. Andrea Hospital, Sapienza University of Rome, Italy; Centre for Primary Care and Public Health (V.G.), Queen Mary University of London, UK; TFS Trial Form Support S.L. (V.A.P., S.M.), Rome, Italy; Institute of Psychological Medicine and Clinical Neurosciences (V.T.), Cardiff University School of Medicine University Hospital of Wales (V.T.), Cardiff, UK; Santa Lucia Foundation (V.T.), Rome, Italy; and IRCCS Neuromed (P.P.), Pozzilli (IS), Italy
| | - Valentina Tomassini
- Department of Neurology and Psychiatry (C.P., L.D.G., F.D.A., M.C.P., V.T., P.P.), Sapienza University of Rome, Italy; MS Center (C.P., L.D.G., V.T.B., F.M.), S. Andrea Hospital, Sapienza University of Rome, Italy; Centre for Primary Care and Public Health (V.G.), Queen Mary University of London, UK; TFS Trial Form Support S.L. (V.A.P., S.M.), Rome, Italy; Institute of Psychological Medicine and Clinical Neurosciences (V.T.), Cardiff University School of Medicine University Hospital of Wales (V.T.), Cardiff, UK; Santa Lucia Foundation (V.T.), Rome, Italy; and IRCCS Neuromed (P.P.), Pozzilli (IS), Italy
| | - Patrizia Pantano
- Department of Neurology and Psychiatry (C.P., L.D.G., F.D.A., M.C.P., V.T., P.P.), Sapienza University of Rome, Italy; MS Center (C.P., L.D.G., V.T.B., F.M.), S. Andrea Hospital, Sapienza University of Rome, Italy; Centre for Primary Care and Public Health (V.G.), Queen Mary University of London, UK; TFS Trial Form Support S.L. (V.A.P., S.M.), Rome, Italy; Institute of Psychological Medicine and Clinical Neurosciences (V.T.), Cardiff University School of Medicine University Hospital of Wales (V.T.), Cardiff, UK; Santa Lucia Foundation (V.T.), Rome, Italy; and IRCCS Neuromed (P.P.), Pozzilli (IS), Italy
| |
Collapse
|
105
|
Airas L. Hormonal and gender-related immune changes in multiple sclerosis. Acta Neurol Scand 2015; 132:62-70. [PMID: 26046561 DOI: 10.1111/ane.12433] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2015] [Indexed: 01/10/2023]
Abstract
Similarly to many other autoimmune diseases, multiple sclerosis (MS) is more common among women than men, and its incidence among women is rising. There are also qualitative differences in the disease course between men and women, with male patients experiencing increased disease progression, brain atrophy, and cognitive impairment. During pregnancy, women with MS typically have a greatly reduced relapse rate, whereas very soon after the delivery, the disease activity returns, often even at a higher level than seen in the prepregnancy year. The reasons for the increased postpartum activity are not entirely clear, but factors such as the abrupt decrease in estrogen levels immediately after the delivery and the loss of the immunosuppressive state of pregnancy are likely of importance. There is compelling evidence that estrogen, progesterone, and testosterone control MS pathology by influencing immune responses and by contributing to repair mechanisms in the nervous system. Hormones may thus offer important insights into MS disease prevention and treatment. In this review, the possible reasons for the sex bias in autoimmune diseases will be discussed. The pregnancy-related alterations in MS, including the effect of pregnancy on disease activity, long-term disability accumulation, and prevalence will be reviewed, as well as the hormonal and immunological mechanisms potentially underlying these changes. Finally, the present thinking on the effect of hormones on the changing incidence of MS will be elucidated.
Collapse
Affiliation(s)
- L. Airas
- Division of Clinical Neurosciences; Turku University Hospital; Turku Finland
| |
Collapse
|
106
|
|
107
|
Singh V, Stingl C, Stoop MP, Zeneyedpour L, Neuteboom RF, Smitt PS, Hintzen RQ, Luider TM. Proteomics urine analysis of pregnant women suffering from multiple sclerosis. J Proteome Res 2015; 14:2065-73. [PMID: 25793971 DOI: 10.1021/pr501162w] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Multiple sclerosis (MScl) frequently is remitted during the third trimester of pregnancy but exacerbated in the first postpartum period. In this context, we investigated protein identification, its abundance, and its change in urine related to these two periods. Using mass spectrometry (LTQ Orbitrap), we identified 1699 tryptic peptides (related to 402 proteins) in urine from 31 MScl and 8 control at these two periods. Pregnancy-related peptides were significantly elevated (p < 0.01) in MScl patients compared with controls (Analysis 1: 531 peptides in MScl and 36 peptides in controls higher abundant in the third trimester compared to postpartum). When comparing the longitudinal differences (Analysis 2), we identified 43 (related to 35 proteins) MScl disease-associated peptides (p < 0.01) with increased or decreased difference ratio in MScl compared with controls. The most discriminating peptides identified were trefoil factor 3 and lysosomal-associated membrane protein 2. Both proteins have a role in the innate immune system. Three proteins with a significant decreased ratio were plasma glutamate carboxypeptidase, Ig mu chain C region, and osteoclast associated immune like receptor. Our results indicate that the protein expression pattern in urine of MScl patients contains information about remote CNS and brain disease processes.
Collapse
Affiliation(s)
- Vaibhav Singh
- Department of Neurology, Erasmus University Medical Center, Dr. Molewaterplein 50, Rotterdam 3015 GE, The Netherlands
| | - Christoph Stingl
- Department of Neurology, Erasmus University Medical Center, Dr. Molewaterplein 50, Rotterdam 3015 GE, The Netherlands
| | - Marcel P Stoop
- Department of Neurology, Erasmus University Medical Center, Dr. Molewaterplein 50, Rotterdam 3015 GE, The Netherlands
| | - Lona Zeneyedpour
- Department of Neurology, Erasmus University Medical Center, Dr. Molewaterplein 50, Rotterdam 3015 GE, The Netherlands
| | - Rinze F Neuteboom
- Department of Neurology, Erasmus University Medical Center, Dr. Molewaterplein 50, Rotterdam 3015 GE, The Netherlands
| | - Peter Sillevis Smitt
- Department of Neurology, Erasmus University Medical Center, Dr. Molewaterplein 50, Rotterdam 3015 GE, The Netherlands
| | - Rogier Q Hintzen
- Department of Neurology, Erasmus University Medical Center, Dr. Molewaterplein 50, Rotterdam 3015 GE, The Netherlands
| | - Theo M Luider
- Department of Neurology, Erasmus University Medical Center, Dr. Molewaterplein 50, Rotterdam 3015 GE, The Netherlands
| |
Collapse
|
108
|
Luessi F, Kraus S, Trinschek B, Lerch S, Ploen R, Paterka M, Roberg T, Poisa-Beiro L, Klotz L, Wiendl H, Bopp T, Jonuleit H, Jolivel V, Zipp F, Witsch E. FTY720 (fingolimod) treatment tips the balance towards less immunogenic antigen-presenting cells in patients with multiple sclerosis. Mult Scler 2015; 21:1811-22. [PMID: 25732840 DOI: 10.1177/1352458515574895] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 01/26/2015] [Indexed: 11/16/2022]
Abstract
OBJECTIVE We aimed to clarify whether fingolimod has direct effects on antigen-presenting cells in multiple sclerosis patients. METHODS Frequency and phenotype of directly ex vivo dendritic cells and monocytes were analyzed in 43 individuals, including fingolimod-treated and untreated multiple sclerosis patients as well as healthy subjects. These cells were further stimulated with lipopolysaccharide to determine functional effects of fingolimod treatment. RESULTS Absolute numbers of CD1c+ dendritic cells and monocytes were not significantly reduced in fingolimod-treated patients indicating that fingolimod did not block the migration of antigen-presenting cells to peripheral blood. CD86 was upregulated on CD1c+ dendritic cells and thus their activation was not impaired under fingolimod treatment. Quantitative analyses of gene transcription in cells and protein content in supernatants from ex vivo CD1c+ dendritic cells and monocytes, however, showed lower secretion of TNFα, IL1-β and IL-6 upon lipopolysaccharide-stimulation. These results could be matched with CD4+MOG-specific transgenic T cells exhibiting reduced levels of TNFα and IFN-γ but not IL-4 upon stimulation with murine dendritic cells loaded with MOG, when treated with fingolimod. CONCLUSIONS Our data indicate that fingolimod - apart from trapping lymphocytes in lymph nodes - exerts its disease-modulating activity by rebalancing the immune tolerance networks by modulation of antigen-presenting cells.
Collapse
Affiliation(s)
- Felix Luessi
- Department of Neurology, University Medical Center of the Johannes Gutenberg-University of Mainz, Germany
| | - Stefan Kraus
- Department of Neurology, University Medical Center of the Johannes Gutenberg-University of Mainz, Germany
| | - Bettina Trinschek
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University of Mainz, Germany
| | - Steffen Lerch
- Department of Neurology, University Medical Center of the Johannes Gutenberg-University of Mainz, Germany
| | - Robert Ploen
- Department of Neurology, University Medical Center of the Johannes Gutenberg-University of Mainz, Germany
| | - Magdalena Paterka
- Department of Neurology, University Medical Center of the Johannes Gutenberg-University of Mainz, Germany
| | - Torsten Roberg
- Department of Neurology, University Medical Center of the Johannes Gutenberg-University of Mainz, Germany
| | - Laura Poisa-Beiro
- Department of Neurology, University Medical Center of the Johannes Gutenberg-University of Mainz, Germany
| | - Luisa Klotz
- Department of Neurology, University of Münster, Germany
| | - Heinz Wiendl
- Department of Neurology, University of Münster, Germany
| | - Tobias Bopp
- Institute for Immunology, University Medical Center of the Johannes Gutenberg-University of Mainz, Germany
| | - Helmut Jonuleit
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University of Mainz, Germany
| | - Valérie Jolivel
- Department of Neurology, University Medical Center of the Johannes Gutenberg-University of Mainz, Germany
| | - Frauke Zipp
- Department of Neurology, University Medical Center of the Johannes Gutenberg-University of Mainz, Germany
| | - Esther Witsch
- Department of Neurology, University Medical Center of the Johannes Gutenberg-University of Mainz, Germany
| |
Collapse
|
109
|
Koong LY, Watson CS. Rapid, nongenomic signaling effects of several xenoestrogens involved in early- vs. late-stage prostate cancer cell proliferation. ACTA ACUST UNITED AC 2015. [DOI: 10.4161/23273747.2014.995003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Luke Y Koong
- Biochemistry & Molecular Biology Department; University of Texas Medical Branch; Galveston, TX USA
| | | |
Collapse
|
110
|
Management of multiple sclerosis during pregnancy and the reproductive years: a systematic review. Obstet Gynecol 2015; 124:1157-1168. [PMID: 25415167 DOI: 10.1097/aog.0000000000000541] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
OBJECTIVE To examine the evidence guiding management of multiple sclerosis (MS) in reproductive-aged women. DATA SOURCES We conducted an electronic literature search using PubMed, ClinicalTrials.gov, and other available resources. The following keywords were used: "multiple sclerosis" and "pregnancy." We manually searched the reference lists of identified studies. METHODS OF STUDY SELECTION Two reviewers categorized all studies identified in the search by management topic, including effect of pregnancy on MS course, fetal risks associated with disease-modifying treatments during pregnancy, and management of patients off disease-modifying treatment. We categorized studies by strength of evidence and included prior meta-analyses and systematic studies. These studies were then summarized and discussed by an expert multidisciplinary team. TABULATION, INTEGRATION, AND RESULTS The risk of MS relapses is decreased during pregnancy and increased postpartum. Data are lacking regarding the risks of disease-modifying treatments during pregnancy. There may be an increased risk of MS relapses after use of assisted reproductive techniques. There does not appear to be a major increase in adverse outcomes in newborns of mothers with MS. CONCLUSION Although there are many unmet research needs, the reviewed data support the conclusion that in the majority of cases, women with MS can safely choose to become pregnant, give birth, and breastfeed children. Clinical management should be individualized to optimize both the mother's reproductive outcomes and MS course.
Collapse
|
111
|
Dunn SE, Gunde E, Lee H. Sex-Based Differences in Multiple Sclerosis (MS): Part II: Rising Incidence of Multiple Sclerosis in Women and the Vulnerability of Men to Progression of this Disease. Curr Top Behav Neurosci 2015; 26:57-86. [PMID: 25690592 DOI: 10.1007/7854_2015_370] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
It is well known that a number of autoimmune diseases including multiple sclerosis (MS) predominantly affect women and there has been much attention directed toward understanding why this is the case. Past research has revealed a number of sex differences in autoimmune responses that can account for the female bias in MS. However, much less is known about why the incidence of MS has increased exclusively in women over the past half century. The recency of this increase suggests that changing environmental or lifestyle factors are interacting with biological sex to increase MS risk predominantly in females. Indeed, a number of recent studies have identified sex-specific differences in the effect of environmental factors on MS incidence. The first part of this chapter will overview this evidence and will discuss the possible scenarios of how the environment may be interacting with autoimmune mechanisms to contribute to the preferential rise in MS incidence in women. Despite the strong female bias in MS incidence, culminating evidence from natural history studies, and imaging and pathology studies suggests that males who develop MS may exhibit a more rapid decline in disability and cognitive functioning than women. Very little is known about the biological basis of this more rapid deterioration, but some insights have been provided by studies in rodent models of demyelination/remyelination. The second part of this chapter will overview the evidence that males with relapsing-onset MS undergo a more rapid progression of disease than females and will discuss potential biological mechanisms that account for this sex difference.
Collapse
Affiliation(s)
- Shannon E Dunn
- Department of Immunology, University of Toronto, Toronto, ON, Canada. .,General Research Institute, University Health Network, Women's College Research Institute, Toronto, ON, Canada.
| | - Eva Gunde
- Department of Medical Neuroscience, Dalhousie University, Halifax, NS, Canada.
| | - Hyunwoo Lee
- Montreal Neurological Institute, McGill University, Montreal, Canada.
| |
Collapse
|
112
|
Multiple functional therapeutic effects of the estrogen receptor β agonist indazole-Cl in a mouse model of multiple sclerosis. Proc Natl Acad Sci U S A 2014; 111:18061-6. [PMID: 25453074 DOI: 10.1073/pnas.1411294111] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Currently available immunomodulatory therapies do not stop the pathogenesis underlying multiple sclerosis (MS) and are only partially effective in preventing the onset of permanent disability in patients with MS. Identifying a drug that stimulates endogenous remyelination and/or minimizes axonal degeneration would reduce the rate and degree of disease progression. Here, the effects of the highly selective estrogen receptor (ER) β agonist indazole chloride (Ind-Cl) on functional remyelination in chronic experimental autoimmune encephalomyelitis (EAE) mice were investigated by assessing pathologic, functional, and behavioral consequences of both prophylactic and therapeutic (peak EAE) treatment with Ind-Cl. Peripheral cytokines from autoantigen-stimulated splenocytes were measured, and central nervous system infiltration by immune cells, axon health, and myelination were assessed by immunohistochemistry and electron microscopy. Therapeutic Ind-Cl improved clinical disease and rotorod performance and also decreased peripheral Th1 cytokines and reactive astrocytes, activated microglia, and T cells in brains of EAE mice. Increased callosal myelination and mature oligodendrocytes correlated with improved callosal conduction and refractoriness. Therapeutic Ind-Cl-induced remyelination was independent of its effects on the immune system, as Ind-Cl increased remyelination within the cuprizone diet-induced demyelinating model. We conclude that Ind-Cl is a refined pharmacologic agent capable of stimulating functionally relevant endogenous myelination, with important implications for progressive MS treatment.
Collapse
|
113
|
Schmitz K, Barthelmes J, Stolz L, Beyer S, Diehl O, Tegeder I. "Disease modifying nutricals" for multiple sclerosis. Pharmacol Ther 2014; 148:85-113. [PMID: 25435020 DOI: 10.1016/j.pharmthera.2014.11.015] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 11/20/2014] [Indexed: 12/26/2022]
Abstract
The association between vitamin D and multiple sclerosis has (re)-opened new interest in nutrition and natural compounds in the prevention and treatment of this neuroinflammatory disease. The dietary amount and type of fat, probiotics and biologicals, salmon proteoglycans, phytoestrogens and protease inhibitor of soy, sodium chloride and trace elements, and fat soluble vitamins including D, A and E were all considered as disease-modifying nutraceuticals. Studies in experimental autoimmune encephalomyelitis mice suggest that poly-unsaturated fatty acids and their 'inflammation-resolving' metabolites and the gut microflora may reduce auto-aggressive immune cells and reduce progression or risk of relapse, and infection with whipworm eggs may positively change the gut-brain communication. Encouraged by the recent interest in multiple sclerosis-nutrition nature's pharmacy has been searched for novel compounds with anti-inflammatory, immune-modifying and antioxidative properties, the most interesting being the scorpion toxins that inhibit specific potassium channels of T cells and antioxidative compounds including the green tea flavonoid epigallocatechin-3-gallate, curcumin and the mustard oil glycoside from e.g. broccoli and sulforaphane. They mostly also inhibit pro-inflammatory signaling through NF-κB or toll-like receptors and stabilize the blood brain barrier. Disease modifying functions may also complement analgesic and anti-spastic effects of cannabis, its constituents, and of 'endocannabinoid enhancing' drugs or nutricals like inhibitors of fatty acid amide hydrolase. Nutricals will not solve multiple sclerosis therapeutic challenges but possibly support pharmacological interventions or unearth novel structures.
Collapse
Affiliation(s)
- Katja Schmitz
- The MS Study Group of the TRIP-Graduate School, Goethe-University Frankfurt, Germany
| | - Julia Barthelmes
- The MS Study Group of the TRIP-Graduate School, Goethe-University Frankfurt, Germany
| | - Leonie Stolz
- The MS Study Group of the TRIP-Graduate School, Goethe-University Frankfurt, Germany
| | - Susanne Beyer
- The MS Study Group of the TRIP-Graduate School, Goethe-University Frankfurt, Germany
| | - Olaf Diehl
- The MS Study Group of the TRIP-Graduate School, Goethe-University Frankfurt, Germany
| | - Irmgard Tegeder
- The MS Study Group of the TRIP-Graduate School, Goethe-University Frankfurt, Germany.
| |
Collapse
|
114
|
Multiple sclerosis at menopause: Potential neuroprotective effects of estrogen. Maturitas 2014; 80:133-9. [PMID: 25544310 DOI: 10.1016/j.maturitas.2014.11.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 11/19/2014] [Accepted: 11/20/2014] [Indexed: 12/17/2022]
Abstract
Multiple sclerosis (MS) is an autoimmune demyelinating and neurodegenerative condition of the central nervous system that preferentially afflicts women more than men. Low estrogen states such as menopause and the postpartum period favor exacerbations of multiple sclerosis in women with the disease. Existing and emerging evidence suggests a role for estrogen in the alleviation of symptoms and reversal of pathology associated with MS. While clinical evidence is sparse regarding the benefit of estrogen therapy for women at risk for MS exacerbations, scientific data demonstrates that estrogen potentiates numerous neuroprotective effects on the central nervous system (CNS). Estrogens play a wide range of roles involved in MS disease pathophysiology, including increasing antiinflammatory cytokines, decreasing demyelination, and enhancing oxidative and energy producing processes in CNS cells.
Collapse
|
115
|
Ochoa-Repáraz J, Kasper LH. Gut microbiome and the risk factors in central nervous system autoimmunity. FEBS Lett 2014; 588:4214-22. [PMID: 25286403 PMCID: PMC4254300 DOI: 10.1016/j.febslet.2014.09.024] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 09/16/2014] [Accepted: 09/16/2014] [Indexed: 02/07/2023]
Abstract
Humans are colonized after birth by microbial organisms that form a heterogeneous community, collectively termed microbiota. The genomic pool of this macro-community is named microbiome. The gut microbiota is essential for the complete development of the immune system, representing a binary network in which the microbiota interact with the host providing important immune and physiologic function and conversely the bacteria protect themselves from host immune defense. Alterations in the balance of the gut microbiome due to a combination of environmental and genetic factors can now be associated with detrimental or protective effects in experimental autoimmune diseases. These gut microbiome alterations can unbalance the gastrointestinal immune responses and influence distal effector sites leading to CNS disease including both demyelination and affective disorders. The current range of risk factors for MS includes genetic makeup and environmental elements. Of interest to this review is the consistency between this range of MS risk factors and the gut microbiome. We postulate that the gut microbiome serves as the niche where different MS risk factors merge, thereby influencing the disease process.
Collapse
Affiliation(s)
- Javier Ochoa-Repáraz
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth College, Hanover, NH 03755, USA.
| | - Lloyd H Kasper
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth College, Hanover, NH 03755, USA
| |
Collapse
|
116
|
Coyle PK. Multiple sclerosis and pregnancy prescriptions. Expert Opin Drug Saf 2014. [DOI: 10.1517/14740338.2015.973848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
117
|
Hsu P, Nanan R. Foetal immune programming: hormones, cytokines, microbes and regulatory T cells. J Reprod Immunol 2014; 104-105:2-7. [DOI: 10.1016/j.jri.2014.02.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 02/05/2014] [Accepted: 02/10/2014] [Indexed: 01/19/2023]
|
118
|
Chakrabarti M, Haque A, Banik NL, Nagarkatti P, Nagarkatti M, Ray SK. Estrogen receptor agonists for attenuation of neuroinflammation and neurodegeneration. Brain Res Bull 2014; 109:22-31. [PMID: 25245209 DOI: 10.1016/j.brainresbull.2014.09.004] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 09/09/2014] [Accepted: 09/10/2014] [Indexed: 01/05/2023]
Abstract
Recent results from laboratory investigations and clinical trials indicate important roles for estrogen receptor (ER) agonists in protecting the central nervous system (CNS) from noxious consequences of neuroinflammation and neurodegeneration. Neurodegenerative processes in several CNS disorders including spinal cord injury (SCI), multiple sclerosis (MS), Parkinson's disease (PD), and Alzheimer's disease (AD) are associated with activation of microglia and astrocytes, which drive the resident neuroinflammatory response. During neurodegenerative processes, activated microglia and astrocytes cause deleterious effects on surrounding neurons. The inhibitory activity of ER agonists on microglia activation might be a beneficial therapeutic option for delaying the onset or progression of neurodegenerative injuries and diseases. Recent studies suggest that ER agonists can provide neuroprotection by modulation of cell survival mechanisms, synaptic reorganization, regenerative responses to axonal injury, and neurogenesis process. The anti-inflammatory and neuroprotective actions of ER agonists are mediated mainly via two ERs known as ERα and ERβ. Although some studies have suggested that ER agonists may be deleterious to some neuronal populations, the potential clinical benefits of ER agonists for augmenting cognitive function may triumph over the associated side effects. Also, understanding the modulatory activities of ER agonists on inflammatory pathways will possibly lead to the development of selective anti-inflammatory molecules with neuroprotective roles in different CNS disorders such as SCI, MS, PD, and AD in humans. Future studies should be concentrated on finding the most plausible molecular pathways for enhancing protective functions of ER agonists in treating neuroinflammatory and neurodegenerative injuries and diseases in the CNS.
Collapse
Affiliation(s)
- Mrinmay Chakrabarti
- University of South Carolina School of Medicine, Department of Pathology, Microbiology, and Immunology, Columbia, SC 29209, USA
| | - Azizul Haque
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Naren L Banik
- Department of Neurosurgery and Neurology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Prakash Nagarkatti
- University of South Carolina School of Medicine, Department of Pathology, Microbiology, and Immunology, Columbia, SC 29209, USA
| | - Mitzi Nagarkatti
- University of South Carolina School of Medicine, Department of Pathology, Microbiology, and Immunology, Columbia, SC 29209, USA
| | - Swapan K Ray
- University of South Carolina School of Medicine, Department of Pathology, Microbiology, and Immunology, Columbia, SC 29209, USA.
| |
Collapse
|
119
|
Abstract
The modern treatment era for multiple sclerosis (MS) began in 1993 with the approval of the first disease-modifying agent. Since then the field has greatly expanded, with 10 therapies currently approved to treat MS. These treatments are effective to reduce relapses and changes on MRI, and slow disability. However, despite these medications some patients continue to have exacerbations, accumulate disability, and develop progressive disease due to partial effectiveness. New molecules with novel mechanisms of action and targets are being explored. Hopefully these agents will yield even greater efficacy without significant safety concerns. As more aggressive therapies are available to treat MS, the goals and expectations of treatment are also likely to change. Some of the emerging therapies, including alemtuzumab, daclizumab, rituximab, ocrelizumab, laquinimod, estriol, 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitors (statins), vitamin D, and stem cell transplantation, will be discussed in this chapter. In the future, therapies with different mechanisms may be combined, but this will need to be evaluated in clinical trials. Neuroprotection and repair definitely warrant further study. The future of MS treatment is very exciting, especially as our armamentarium expands.
Collapse
|
120
|
Namjooyan F, Ghanavati R, Majdinasab N, Jokari S, Janbozorgi M. Uses of complementary and alternative medicine in multiple sclerosis. J Tradit Complement Med 2014; 4:145-52. [PMID: 25161918 PMCID: PMC4142451 DOI: 10.4103/2225-4110.136543] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic, disabling, recurrent demyelination of the central nervous system (CNS). It could affect different regions in the brain and spinal cord, and according to the domain which is affected, it could cause different symptoms such as motor, sensory, or visual impairment; fatigue; bowel, bladder, and sexual dysfunction; cognitive impairment; and depression. MS patients also face reduced quality of life. Drugs that are used in MS are not fully efficient and patients suffer from many symptoms and adverse effects. Today there is an increasing trend of using complementary and alternative medicine (CAM). People are more likely to use this type of treatment. Using appropriate lifestyle and CAM therapy can subside some of the symptoms and could improve the quality of life in these patients. Many people with MS explore CAM therapies for their symptoms. This review is aimed to introduce CAM therapies that could be used in MS patients.
Collapse
Affiliation(s)
- Foroogh Namjooyan
- Department of Pharmacognosy, Marine Natural Pharmaceutical Research Center, School of Pharmacy, Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Rahil Ghanavati
- Department of Traditional Pharmacy, School of Pharmacy, Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Nastaran Majdinasab
- Department of Neurology, School of Medicine, Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Shiva Jokari
- Jundishapur University of Medical Sciences, Arvand International Branch, Abadan, Iran
| | | |
Collapse
|
121
|
He YX, Du M, Shi HL, Huang F, Liu HS, Wu H, Zhang BB, Dou W, Wu XJ, Wang ZT. Astragalosides from Radix Astragali benefits experimental autoimmune encephalomyelitis in C57BL /6 mice at multiple levels. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2014; 14:313. [PMID: 25150364 PMCID: PMC4155103 DOI: 10.1186/1472-6882-14-313] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Accepted: 08/20/2014] [Indexed: 12/03/2022]
Abstract
Background Radix Astragali is famous for its beneficial effect on inflammation associated diseases. This study was to assess the efficacy of astragalosides (AST) extracted from Radix Astragali, on the progression of experimental autoimmune encephalomyelitis (EAE), and explore its possible underlying molecular mechanisms. Methods EAE was induced by subcutaneous immunization of MOG35–55. Infiltration of inflammatory cells was examined by HE staining. ROS level was detected by measuring infiltrated hydroethidine. Leakage of blood brain barrier (BBB) was assessed using Evan’s blue dye extravasation method. Levels of inflammatory cytokines were measured using ELISA kits. Activities of total-SOD, GSH-Px, and iNOS and MDA concentration were measured using biochemical analytic kits. Gene expression was detected using real-time PCR method. Protein expression was assayed using western blotting approach. Results AST administration attenuated the progression of EAE in mice remarkably. Further studies manifested that AST treatment inhibited infiltration of inflammatory cells, lessened ROS production and decreased BBB leakage. In peripheral immune-systems, AST up-regulated mRNA expression of transcriptional factors T-bet and Foxp3 but decreased that of RORγt to modulate T cell differentiation. In CNS, AST stopped BBB leakage, reduced ROS production by up-regulation of T-SOD, and reduced neuroinflammation by inhibition of iNOS and other inflammatory cytokines. Moreover, AST inhibited production of p53 and phosphorylation of tau by modulation of the Bcl-2/Bax ratio. Conclusions AST orchestrated multiple pathways, including immuno-regulation, anti-oxidative stress, anti-neuroinflammation and anti-neuroapoptosis involved in the MS pathogenesis, to prevent the deterioration of EAE, which paves the way for the application of it in clinical prevention/therapy of MS.
Collapse
|
122
|
Ngo ST, Steyn FJ, McCombe PA. Gender differences in autoimmune disease. Front Neuroendocrinol 2014; 35:347-69. [PMID: 24793874 DOI: 10.1016/j.yfrne.2014.04.004] [Citation(s) in RCA: 666] [Impact Index Per Article: 60.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Revised: 04/20/2014] [Accepted: 04/22/2014] [Indexed: 12/21/2022]
Abstract
Autoimmune diseases are a range of diseases in which the immune response to self-antigens results in damage or dysfunction of tissues. Autoimmune diseases can be systemic or can affect specific organs or body systems. For most autoimmune diseases there is a clear sex difference in prevalence, whereby females are generally more frequently affected than males. In this review, we consider gender differences in systemic and organ-specific autoimmune diseases, and we summarize human data that outlines the prevalence of common autoimmune diseases specific to adult males and females in countries commonly surveyed. We discuss possible mechanisms for sex specific differences including gender differences in immune response and organ vulnerability, reproductive capacity including pregnancy, sex hormones, genetic predisposition, parental inheritance, and epigenetics. Evidence demonstrates that gender has a significant influence on the development of autoimmune disease. Thus, considerations of gender should be at the forefront of all studies that attempt to define mechanisms that underpin autoimmune disease.
Collapse
Affiliation(s)
- S T Ngo
- School of Biomedical Sciences, University of Queensland, St Lucia, Queensland, Australia; University of Queensland Centre for Clinical Research, University of Queensland, Herston, Queensland, Australia
| | - F J Steyn
- School of Biomedical Sciences, University of Queensland, St Lucia, Queensland, Australia
| | - P A McCombe
- University of Queensland Centre for Clinical Research, University of Queensland, Herston, Queensland, Australia; Department of Neurology, Royal Brisbane & Women's Hospital, Herston, Queensland, Australia.
| |
Collapse
|
123
|
Avivi I, Farbstein D, Brenner B, Horowitz NA. Non-Hodgkin lymphomas in pregnancy: tackling therapeutic quandaries. Blood Rev 2014; 28:213-20. [PMID: 25108745 DOI: 10.1016/j.blre.2014.06.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 06/23/2014] [Accepted: 06/27/2014] [Indexed: 12/15/2022]
Abstract
Hodgkin lymphoma (HL) and non-Hodgkin lymphoma (NHL) often present with systemic symptoms such as fatigue, shortness of breath and night sweats, mimicking pregnancy-related features which may result in delayed disease diagnosis. Furthermore, the wish to avoid investigational imaging, aiming to protect the fetus from radiation exposure, may lead to a further delay, which does not often result in significant changes in HL clinical nature and patient outcome. In contrast, a more aggressive behavior (i.e., advanced disease stage and reproductive organ involvement) of most NHL types diagnosed in pregnancy may require urgent therapeutic intervention to prevent disease progression. Current management of pregnancy-associated NHL depends on histological subtype of the disease, gestational stage at diagnosis and the urgency of treatment for a specific patient. Patients diagnosed with indolent lymphoma may often be just followed, whereas those presenting with aggressive or highly aggressive disease need to be urgently treated with chemoimmunotherapy, either after undergoing an elective pregnancy termination if diagnosed at an early gestational stage, or with pregnancy preservation, if diagnosed later. Supportive care of NHL is also important; however, granulocyte colony stimulating factor (G-CSF) which is commonly used outside of pregnancy, should be cautiously employed, considering its established teratogenicity in animals, though this is less proven in humans. In conclusion, given the paucity of studies prospectively evaluating the outcome of pregnant women with NHL, international efforts are warranted to elucidate critical issues and develop guidelines for the management of such patients.
Collapse
Affiliation(s)
- Irit Avivi
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel; Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel.
| | - Dan Farbstein
- Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Benjamin Brenner
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel; Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Netanel A Horowitz
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel; Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| |
Collapse
|
124
|
Broadley SA, Barnett MH, Boggild M, Brew BJ, Butzkueven H, Heard R, Hodgkinson S, Kermode AG, Lechner-Scott J, Macdonell RAL, Marriott M, Mason DF, Parratt J, Reddel SW, Shaw CP, Slee M, Spies J, Taylor BV, Carroll WM, Kilpatrick TJ, King J, McCombe PA, Pollard JD, Willoughby E. Therapeutic approaches to disease modifying therapy for multiple sclerosis in adults: an Australian and New Zealand perspective: part 1 historical and established therapies. MS Neurology Group of the Australian and New Zealand Association of Neurologists. J Clin Neurosci 2014; 21:1835-46. [PMID: 24993135 DOI: 10.1016/j.jocn.2014.01.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 01/28/2014] [Indexed: 01/05/2023]
Abstract
Multiple sclerosis (MS) is a potentially life-changing immune mediated disease of the central nervous system. Until recently, treatment has been largely confined to acute treatment of relapses, symptomatic therapies and rehabilitation. Through persistent efforts of dedicated physicians and scientists around the globe for 160 years, a number of therapies that have an impact on the long term outcome of the disease have emerged over the past 20 years. In this three part series we review the practicalities, benefits and potential hazards of each of the currently available and emerging treatment options for MS. We pay particular attention to ways of abrogating the risks of these therapies and provide advice on the most appropriate indications for using individual therapies. In Part 1 we review the history of the development of MS therapies and its connection with the underlying immunobiology of the disease. The established therapies for MS are reviewed in detail and their current availability and indications in Australia and New Zealand are summarised. We examine the evidence to support their use in the treatment of MS.
Collapse
Affiliation(s)
- Simon A Broadley
- School of Medicine, Griffith University, Gold Coast Campus, QLD 4222, Australia; Department of Neurology, Gold Coast University Hospital, Southport, QLD, Australia.
| | - Michael H Barnett
- Brain and Mind Research Institute, University of Sydney, Camperdown, NSW, Australia
| | - Mike Boggild
- Department of Neurology, The Townsville Hospital, Douglas, QLD, Australia
| | - Bruce J Brew
- Department of Neurology and St Vincent's Centre for Applied Medical Research, St Vincent's Hospital, University of New South Wales, Darlinghurst, NSW, Australia
| | - Helmut Butzkueven
- Melbourne Brain Centre, Royal Melbourne Hospital, University of Melbourne, Parkville, VIC, Australia
| | - Robert Heard
- Westmead Clinical School, University of Sydney, NSW, Australia
| | - Suzanne Hodgkinson
- South Western Sydney Clinical School, University of New South Wales, NSW, Australia
| | - Allan G Kermode
- Centre for Neuromuscular and Neurological Disorders, University of Western Australia, WA, Australia; Institute of Immunology and Infectious Diseases, Murdoch University, WA, Australia
| | | | | | - Mark Marriott
- Melbourne Brain Centre, Royal Melbourne Hospital, University of Melbourne, Parkville, VIC, Australia
| | - Deborah F Mason
- Department of Neurology, Christchurch Hospital, Christchurch, New Zealand
| | - John Parratt
- Central Clinical School, University of Sydney, NSW, Australia
| | - Stephen W Reddel
- Brain and Mind Research Institute, University of Sydney, Camperdown, NSW, Australia
| | | | - Mark Slee
- Centre for Neuroscience and Flinders Medical Centre, Flinders University, SA, Australia
| | - Judith Spies
- Brain and Mind Research Institute, University of Sydney, Camperdown, NSW, Australia
| | - Bruce V Taylor
- Menzies Research Institute, University of Tasmania, TAS, Australia
| | - William M Carroll
- Centre for Neuromuscular and Neurological Disorders, University of Western Australia, WA, Australia
| | | | - John King
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Pamela A McCombe
- University of Queensland Centre for Clinical Research, QLD, Australia
| | - John D Pollard
- Brain and Mind Research Institute, University of Sydney, Camperdown, NSW, Australia
| | - Ernest Willoughby
- Department of Neurology, Auckland City Hospital, Auckland, New Zealand
| |
Collapse
|
125
|
|
126
|
Abstract
Microglia, the resident innate immune cells in the brain, have long been understood to be crucial to maintenance in the nervous system, by clearing debris, monitoring for infiltration of infectious agents, and mediating the brain's inflammatory and repair response to traumatic injury, stroke, or neurodegeneration. A wave of new research has shown that microglia are also active players in many basic processes in the healthy brain, including cell proliferation, synaptic connectivity, and physiology. Microglia, both in their capacity as phagocytic cells and via secretion of many neuroactive molecules, including cytokines and growth factors, play a central role in early brain development, including sexual differentiation of the brain. In this review, we present the vast roles microglia play in normal brain development and how perturbations in the normal neuroimmune environment during development may contribute to the etiology of brain-based disorders. There are notable differences between microglia and neuroimmune signaling in the male and female brain throughout the life span, and these differences may contribute to the vast differences in the incidence of neuropsychiatric and neurological disorders between males and females.
Collapse
Affiliation(s)
- Kathryn M Lenz
- Department of Psychology and Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| | - Margaret M McCarthy
- Department of Pharmacology and Program in Neuroscience, The University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
127
|
Faria DDP, Copray S, Buchpiguel C, Dierckx R, de Vries E. PET imaging in multiple sclerosis. J Neuroimmune Pharmacol 2014; 9:468-82. [PMID: 24809810 DOI: 10.1007/s11481-014-9544-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 04/21/2014] [Indexed: 01/03/2023]
Abstract
Positron emission tomography (PET) is a non-invasive technique for quantitative imaging of biochemical and physiological processes in animals and humans. PET uses probes labeled with a radioactive isotope, called PET tracers, which can bind to or be converted by a specific biological target and thus can be applied to detect and monitor different aspects of diseases. The number of applications of PET imaging in multiple sclerosis is still limited. Clinical studies using PET are basically focused on monitoring changes in glucose metabolism and the presence of activated microglia/macrophages in sclerotic lesions. In preclinical studies, PET imaging of targets for other processes, like demyelination and remyelination, has been investigated and may soon be translated to clinical applications. Moreover, more PET tracers that could be relevant for MS are available now, but have not been studied in this context yet. In this review, we summarize the PET imaging studies performed in multiple sclerosis up to now. In addition, we will identify potential applications of PET imaging of processes or targets that are of interest to MS research, but have yet remained largely unexplored.
Collapse
Affiliation(s)
- Daniele de Paula Faria
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | | | | | | | | |
Collapse
|
128
|
Gava G, Bartolomei I, Costantino A, Berra M, Venturoli S, Salvi F, Meriggiola MC. Long-term influence of combined oral contraceptive use on the clinical course of relapsing-remitting multiple sclerosis. Fertil Steril 2014; 102:116-22. [PMID: 24794311 DOI: 10.1016/j.fertnstert.2014.03.054] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2013] [Revised: 03/28/2014] [Accepted: 03/28/2014] [Indexed: 11/16/2022]
Abstract
OBJECTIVE To assess the long-term effects of combined oral contraceptives (COCs) on the clinical course of relapsing-remitting multiple sclerosis (RRMS), focusing on disability progression and evolution to secondary-progressive multiple sclerosis (SPMS). DESIGN Retrospective and exploratory study. SETTING Academic medical center. PATIENT(S) A total of 174 women with clinically confirmed MS; of these, 33 had evolved to SPMS at the time of enrollment in the study, whereas 141 still had a relapsing-remitting form of disease. INTERVENTION(S) Women were interviewed to obtain gynecologic and obstetric history. MAIN OUTCOME MEASURE(S) Expanded Disability Status Scale (EDSS); Multiple Sclerosis Severity Score (MSSS); annualized relapse rate; evolution to SPMS. RESULT(S) Mean±SD duration of disease was 14.3±9.8 years. Compared with non-users of COCs, COC users had lower EDSS scores and MSSS only in the subset of the population with prior or current immunomodulatory treatment. Nonuse of COCs was a predictor of disease evolution in SPMS, whether treated or not with immunomodulatory drugs. The annualized relapse rate was not influenced by COC use. No differences in EDSS scores and evolution to SPMS depending on COC formulation were detected. CONCLUSION(S) Our results suggest that COC use is associated with a less severe disease and less severe evolution. Whether different doses or types of progestin may have different effects remains to be defined.
Collapse
Affiliation(s)
- Giulia Gava
- Gynecology and Physiopathology of Human Reproduction, University of Bologna and S. Orsola-Malpighi Hospital, Bologna, Italy
| | - Ilaria Bartolomei
- Center for Rare and Neuroimmunological Diseases, Scientific Institute for Research, Hospitalization and Health Care (IRCCS), Istituto delle Scienze Neurologiche di Bologna, Bellaria Hospital, Bologna, Italy
| | - Antonietta Costantino
- Gynecology and Physiopathology of Human Reproduction, University of Bologna and S. Orsola-Malpighi Hospital, Bologna, Italy
| | - Marta Berra
- Gynecology and Physiopathology of Human Reproduction, University of Bologna and S. Orsola-Malpighi Hospital, Bologna, Italy
| | - Stefano Venturoli
- Gynecology and Physiopathology of Human Reproduction, University of Bologna and S. Orsola-Malpighi Hospital, Bologna, Italy
| | - Fabrizio Salvi
- Center for Rare and Neuroimmunological Diseases, Scientific Institute for Research, Hospitalization and Health Care (IRCCS), Istituto delle Scienze Neurologiche di Bologna, Bellaria Hospital, Bologna, Italy
| | - Maria Cristina Meriggiola
- Gynecology and Physiopathology of Human Reproduction, University of Bologna and S. Orsola-Malpighi Hospital, Bologna, Italy.
| |
Collapse
|
129
|
Li R, Xu W, Chen Y, Qiu W, Shu Y, Wu A, Dai Y, Bao J, Lu Z, Hu X. Raloxifene suppresses experimental autoimmune encephalomyelitis and NF-κB-dependent CCL20 expression in reactive astrocytes. PLoS One 2014; 9:e94320. [PMID: 24722370 PMCID: PMC3983123 DOI: 10.1371/journal.pone.0094320] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 03/14/2014] [Indexed: 12/19/2022] Open
Abstract
Recent clinical data have led to the consideration of sexual steroids as new potential therapeutic tools for multiple sclerosis. Selective estrogen receptor modulators can exhibit neuroprotective effects like estrogen, with fewer systemic estrogen side effects than estrogen, offering a more promising therapeutic modality for multiple sclerosis. The important role of astrocytes in a proinflammatory effect mediated by CCL20 signaling on inflammatory cells has been documented. Their potential contribution to selective estrogen receptor modulator-mediated protection is still unknown. Using a mouse model of chronic neuroinflammation, we report that raloxifene, a selective estrogen receptor modulator, alleviated experimental autoimmune encephalomyelitis–an animal model of multiple sclerosis–and decreased astrocytic production of CCL20. Enzyme-linked immunosorbent assay, immunohistochemistry imaging and transwell migration assays revealed that reactive astrocytes express CCL20, which promotes Th17 cell migration. In cultured rodent astrocytes, raloxifene inhibited IL-1β-induced CCL20 expression and chemotaxis ability for Th17 migration, whereas the estrogen receptor antagonist ICI 182,780 blocked this effect. Western blotting further indicated that raloxifene suppresses IL-1β-induced NF-κB activation (phosphorylation of p65) and translocation but does not affect phosphorylation of IκB. In conclusion, these data demonstrate that raloxifene provides robust neuroprotection against experimental autoimmune encephalomyelitis, partially via an inhibitory action on CCL20 expression and NF-κB pathways in reactive astrocytes. Our results contribute to a better understanding of the critical roles of raloxifene in treating experimental autoimmune encephalomyelitis and uncover reactive astrocytes as a new target for the inhibitory action of estrogen receptors on chemokine CCL20 expression.
Collapse
MESH Headings
- Animals
- Astrocytes/drug effects
- Astrocytes/pathology
- Cell Movement/drug effects
- Cells, Cultured
- Chemokine CCL20/antagonists & inhibitors
- Chemokine CCL20/genetics
- Chemokine CCL20/immunology
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Estradiol/analogs & derivatives
- Estradiol/pharmacology
- Female
- Fulvestrant
- Gene Expression Regulation
- Mice
- Mice, Inbred C57BL
- Multiple Sclerosis/drug therapy
- Multiple Sclerosis/genetics
- Multiple Sclerosis/pathology
- NF-kappa B/genetics
- NF-kappa B/metabolism
- Raloxifene Hydrochloride/pharmacology
- Receptors, Estrogen/antagonists & inhibitors
- Receptors, Estrogen/genetics
- Receptors, Estrogen/immunology
- Selective Estrogen Receptor Modulators/pharmacology
- Signal Transduction
- Th17 Cells/drug effects
- Th17 Cells/pathology
Collapse
Affiliation(s)
- Rui Li
- Multiple Sclerosis Center, Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Wen Xu
- Multiple Sclerosis Center, Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Ying Chen
- Multiple Sclerosis Center, Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Wei Qiu
- Multiple Sclerosis Center, Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Yaqing Shu
- Multiple Sclerosis Center, Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Aimin Wu
- Multiple Sclerosis Center, Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Yongqiang Dai
- Multiple Sclerosis Center, Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Jian Bao
- Multiple Sclerosis Center, Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Zhengqi Lu
- Multiple Sclerosis Center, Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Xueqiang Hu
- Multiple Sclerosis Center, Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
- * E-mail:
| |
Collapse
|
130
|
Miller DH, Fazekas F, Montalban X, Reingold SC, Trojano M. Pregnancy, sex and hormonal factors in multiple sclerosis. Mult Scler 2014; 20:527-36. [PMID: 24446387 PMCID: PMC5458871 DOI: 10.1177/1352458513519840] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 12/04/2013] [Indexed: 01/17/2023]
Abstract
BACKGROUND Multiple sclerosis (MS) is influenced by pregnancy, sex and hormonal factors. OBJECTIVES A comprehensive understanding of the role of pregnancy, sex and hormonal factors can provide insights into disease mechanisms, and new therapeutic developments and can provide improved patient care and treatment. METHODS Based on an international conference of experts and a comprehensive PubMed search for publications on these areas in MS, we provide a review of what is known about the impact of these factors on disease demographics, etiology, pathophysiology and clinical course and outcomes. RESULTS AND CONCLUSIONS Recommendations are provided for counseling and management of people with MS before conception, during pregnancy and after delivery. The use of disease-modifying and symptomatic therapies in pregnancy is problematic and such treatments are normally discontinued. Available knowledge about the impact of treatment on the mother, fetus and newborn is discussed. Recommendations for future research to fill knowledge gaps and clarify inconsistencies in available data are made.
Collapse
Affiliation(s)
- David H Miller
- Queen Square Multiple Sclerosis Centre, Department of Neuroinflammation, Institute of Neurology, University College London, UK
| | - Franz Fazekas
- Department of Neurology, Medical University of Graz, Austria
| | - Xavier Montalban
- Department of Neurology/Neuroimmunology, MS Center of Catalonia (Cemcat), Vall d’Hebron University Hospital and Research Institute, Spain
| | | | - Maria Trojano
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari, Italy
| |
Collapse
|
131
|
Abstract
Multiple sclerosis (MS) is a chronic progressive demyelinating disease of the central nervous system. Common manifestations include paresthesias, diplopia, loss of vision, numbness or weakness of the limbs, bowel or bladder dysfunction, spasticity, ataxia, fatigue, and mental changes. Four main patterns of MS are recognized: relapsing remitting, primary progressive, secondary progressive, and progressive relapsing. The cause of MS is unknown, although it appears to be an autoimmune disease. Much of what is known about MS has been learned from an animal model of the disease, experimental allergic encephalomyelitis.
Collapse
Affiliation(s)
- Alan Gaby
- Alan Gaby, MD, is internationally recognized as an expert in the field of nutritional therapy. He has recently completed a 30-year project, a textbook titled Nutritional Medicine . This article is adapted from chapter 137 of the textbook with permission from www.doctorgaby.com , Concord, New Hampshire, United States; 2011
| |
Collapse
|
132
|
Rahn EJ, Iannitti T, Donahue RR, Taylor BK. Sex differences in a mouse model of multiple sclerosis: neuropathic pain behavior in females but not males and protection from neurological deficits during proestrus. Biol Sex Differ 2014; 5:4. [PMID: 24581045 PMCID: PMC3974112 DOI: 10.1186/2042-6410-5-4] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Accepted: 01/31/2014] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Multiple sclerosis (MS), a demyelinating disease of the central nervous system, is one of the most prevalent neurological disorders in the industrialized world. This disease afflicts more than two million people worldwide, over two thirds of which are women. MS is typically diagnosed between the ages of 20-40 and can produce debilitating neurological impairments including muscle spasticity, muscle paralysis, and chronic pain. Despite the large sex disparity in MS prevalence, clinical and basic research investigations of how sex and estrous cycle impact development, duration, and severity of neurological impairments and pain symptoms are limited. To help address these questions, we evaluated behavioral signs of sensory and motor functions in one of the most widely characterized animal models of MS, the experimental autoimmune encephalomyelitis (EAE) model. METHODS C57BL/6 male and female mice received flank injection of complete Freund's adjuvant (CFA) or CFA plus myelin oligodendrocyte glycoprotein 35-55 (MOG35-55) to induce EAE. Experiment 1 evaluated sex differences of EAE-induced neurological motor deficits and neuropathic pain-like behavior over 3 weeks, while experiment 2 evaluated the effect of estrous phase in female mice on the same behavioral measures for 3 months. EAE-induced neurological motor deficits including gait analysis and forelimb grip strength were assessed. Neuropathic pain-like behaviors evaluated included sensitivity to mechanical, cold, and heat stimulations. Estrous cycle was determined daily via vaginal lavage. RESULTS MOG35-55-induced EAE produced neurological impairments (i.e., motor dysfunction) including mild paralysis and decreases in grip strength in both females and males. MOG35-55 produced behavioral signs of neuropathic pain-mechanical and cold hypersensitivity-in females, but not males. MOG35-55 did not change cutaneous heat sensitivity in either sex. Administration of CFA or CFA + MOG35-55 prolonged the time spent in diestrus for 2 weeks, after which normal cycling returned. MOG35-55 produced fewer neurological motor deficits when mice were in proestrus relative to non-proestrus phases. CONCLUSIONS We conclude that female mice are superior to males for the study of neuropathic pain-like behaviors associated with MOG35-55-induced EAE. Further, proestrus may be protective against EAE-induced neurological deficits, thus necessitating further investigation into the impact that estrous cycle exerts on MS symptoms.
Collapse
Affiliation(s)
| | | | | | - Bradley K Taylor
- Department of Physiology, University of Kentucky, 800 Rose Street, Lexington, KY 40536, USA.
| |
Collapse
|
133
|
Bove R, Chitnis T. The role of gender and sex hormones in determining the onset and outcome of multiple sclerosis. Mult Scler 2014; 20:520-6. [DOI: 10.1177/1352458513519181] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Intriguing sex differences both in multiple sclerosis (MS) susceptibility and its disease course may offer important insights into MS disease pathophysiology, prevention and treatment. In this review, we first summarize the key sex-related differences in MS risk, heritability and disease progression. One promising hypothesis we explore is whether sexually-dimorphic responsiveness to cultural and environmental changes may explain the observation of an increasing female:male sex ratio in MS. We then review the evidence for hormonal modulation of MS, during such transitions as puberty and pregnancy. Finally, we review sex differences in the non-inflammatory facets of MS. We highlight those research gaps that may point to important sex or sex hormone-mediated mechanistic and therapeutic insights.
Collapse
Affiliation(s)
- Riley Bove
- Department of Neurology, Brigham and Women’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Tanuja Chitnis
- Department of Neurology, Brigham and Women’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|
134
|
Abstract
PURPOSE OF REVIEW This article reviews the current understanding of the interactions between multiple sclerosis (MS) and pregnancy, and implications for reproductive counseling. This is a key topic in MS because the typical patient is a young woman of childbearing age. RECENT FINDINGS It has been known for some time that MS disease activity markedly reduces during the last trimester of pregnancy, then markedly increases in the 3 months postpartum before returning to the prepregnancy baseline. High relapse rate or disability before pregnancy, as well as relapse during pregnancy, have been associated with increased risk for postpartum attacks. Recent data continue to support the conclusion that long-term disease progression is not worsened (and may actually be lessened) with pregnancy in patients with relapsing MS; the data are not so clear for those with progressive MS. Among the MS disease-modifying therapies, the only one that requires contraception use by men is the new oral agent teriflunomide, because the drug is present in semen. It is reassuring that, to date, no human teratogenic effects have been documented for any of the MS disease-modifying therapies. SUMMARY Pregnancy has a profound effect on MS disease activity. Identification of the responsible mechanisms for this effect should lead to new disease insights and therapeutic strategies.
Collapse
|
135
|
|
136
|
Galimberti D, Bresolin N, Scarpini E. Chemokine network in multiple sclerosis: role in pathogenesis and targeting for future treatments. Expert Rev Neurother 2014; 4:439-53. [PMID: 15853541 DOI: 10.1586/14737175.4.3.439] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Multiple sclerosis is the most common inflammatory disorder of the CNS. Evidence suggests that an immunomediated mechanism plays a crucial role during the development of the disease. Currently, two classes of immunomodulatory agents -- interferon-beta and glatiramer acetate (Copaxone, Teva Pharmaceutical Industries), have been approved for the long-term treatment of multiple sclerosis. New drugs which effectively target the immunological processes occurring in multiple sclerosis have been proposed. This review summarizes the immunological background that occurs during the pathogenesis of multiple sclerosis focusing on chemokines and related receptors. The effects of standard treatments on the immune system are analyzed along with the current knowledge of potential new immunomodulatory molecules, such as antiadhesion molecules, statins, estriol, cannabinoids, neurotrophic factors and chemokine antagonists.
Collapse
Affiliation(s)
- Daniela Galimberti
- Department of Neurological Sciences, Dino Ferrari Center and Center of Excellence for Neurodegenerative Diseases, University of Milan, IRCCS Ospedale Maggiore Policlinico, Via F Sforza 35, 20122, Milan, Italy.
| | | | | |
Collapse
|
137
|
Lo A. Advancement of therapies for neuroprotection in multiple sclerosis. Expert Rev Neurother 2014; 8:1355-66. [DOI: 10.1586/14737175.8.9.1355] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
138
|
Angelo Ghezzi A, Zaffaroni M. Female-specific issues in multiple sclerosis. Expert Rev Neurother 2014; 8:969-77. [DOI: 10.1586/14737175.8.6.969] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
139
|
Neuroendocrine immunoregulation in multiple sclerosis. Clin Dev Immunol 2013; 2013:705232. [PMID: 24382974 PMCID: PMC3870621 DOI: 10.1155/2013/705232] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Revised: 09/29/2013] [Accepted: 09/30/2013] [Indexed: 12/03/2022]
Abstract
Currently, it is generally accepted that multiple sclerosis (MS) is a complex multifactorial disease involving genetic and environmental factors affecting the autoreactive immune responses that lead to damage of myelin. In this respect, intrinsic or extrinsic factors such as emotional, psychological, traumatic, or inflammatory stress as well as a variety of other lifestyle interventions can influence the neuroendocrine system. On its turn, it has been demonstrated that the neuroendocrine system has immunomodulatory potential. Moreover, the neuroendocrine and immune systems communicate bidirectionally via shared receptors and shared messenger molecules, variously called hormones, neurotransmitters, or cytokines. Discrepancies at any level can therefore lead to changes in susceptibility and to severity of several autoimmune and inflammatory diseases. Here we provide an overview of the complex system of crosstalk between the neuroendocrine and immune system as well as reported dysfunctions involved in the pathogenesis of autoimmunity, including MS. Finally, possible strategies to intervene with the neuroendocrine-immune system for MS patient management will be discussed. Ultimately, a better understanding of the interactions between the neuroendocrine system and the immune system can open up new therapeutic approaches for the treatment of MS as well as other autoimmune diseases.
Collapse
|
140
|
Dolezal O, Gabelic T, Horakova D, Bergsland N, Dwyer MG, Seidl Z, Krasensky J, Ramasamy DP, Vaneckova M, Havrdova E, Zivadinov R. Development of gray matter atrophy in relapsing–remitting multiple sclerosis is not gender dependent: Results of a 5-year follow-up study. Clin Neurol Neurosurg 2013; 115 Suppl 1:S42-8. [DOI: 10.1016/j.clineuro.2013.09.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
141
|
Bove R, Chitnis T, Houtchens M. Menopause in multiple sclerosis: therapeutic considerations. J Neurol 2013; 261:1257-68. [PMID: 24101131 DOI: 10.1007/s00415-013-7131-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Accepted: 09/24/2013] [Indexed: 11/25/2022]
Abstract
While the onset of multiple sclerosis (MS) typically occurs during the childbearing years, many women living with MS are of perimenopausal age. There is frequent overlap between menopausal and MS-related symptoms and co-morbidities (e.g. sexual dysfunction, mood disorders and bladder function). Furthermore, some MS symptoms may be exacerbated by perimenopausal changes such as hot flashes or sleep disturbance. The MS neurologist may frequently be the first to become aware of these symptoms and to play a role in monitoring and managing them. In this review, we describe immunological and neurologic changes at menopause as they may impact MS. We then review common symptoms, including fatigue, depression, sexual function, pain and insomnia, and provide both behavioral and pharmacological suggestions for their management. Next, we discuss the need for osteoporosis and cancer screening in perimenopausal women with MS. Finally, we highlight important research gaps, including what effect, if any, the menopausal transition may play on MS disease course as well as the potential modulatory role of hormone replacement therapies.
Collapse
Affiliation(s)
- Riley Bove
- Department of Neurology, Partners Multiple Sclerosis Center, Brigham and Women's Hospital, 1 Brookline Place West, Suite 225, Brookline, MA, 02445, USA,
| | | | | |
Collapse
|
142
|
Patas K, Engler JB, Friese MA, Gold SM. Pregnancy and multiple sclerosis: feto-maternal immune cross talk and its implications for disease activity. J Reprod Immunol 2013; 97:140-6. [PMID: 23432880 DOI: 10.1016/j.jri.2012.10.005] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Revised: 09/30/2012] [Accepted: 10/01/2012] [Indexed: 11/16/2022]
Abstract
Multiple sclerosis (MS) is an inflammatory, demyelinating disease of the central nervous system of presumed autoimmune origin. Intriguingly, pregnancy in female MS patients is associated with a substantial decrease in relapse rate. However, post-partum the relapse rate increases in a rebounding fashion above the rate seen before pregnancy. Wide gaps remain in our understanding of the biological mechanisms underlying these pregnancy-related effects in MS patients. To date, most attempts to explain MS disease amelioration during pregnancy have focused on levels of circulating hormones with immunomodulatory properties such as estrogens and global shifts in systemic maternal immune cell composition. However, recent advances in our understanding of feto-maternal tolerance have provided evidence that fetal antigens directly interact with the maternal immune system. This results in specific immunomodulation such as fetal-antigen-dependent induction of regulatory T cells. Thus, the "shaping" of maternal immune responses by fetal antigens may represent an endogenous pathway by which antigen-specific immunomodulation might also contribute to reinstalling tolerance to autoantigens in MS. Reproductive immunology therefore has great potential to provide insights into MS immunopathogenesis and highlight novel avenues for treatment of MS and other autoimmune diseases.
Collapse
Affiliation(s)
- Konstantinos Patas
- Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | | | | |
Collapse
|
143
|
Koch MW, Cutter G, Stys PK, Yong VW, Metz LM. Treatment trials in progressive MS—current challenges and future directions. Nat Rev Neurol 2013; 9:496-503. [DOI: 10.1038/nrneurol.2013.148] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
144
|
Koenig KA, Lowe MJ, Lin J, Sakaie KE, Stone L, Bermel RA, Beall EB, Rao SM, Trapp BD, Phillips MD. Sex differences in resting-state functional connectivity in multiple sclerosis. AJNR Am J Neuroradiol 2013; 34:2304-11. [PMID: 23811974 DOI: 10.3174/ajnr.a3630] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
BACKGROUND AND PURPOSE Multiple studies have demonstrated evidence of sex differences in patients with MS, including differences in disease progression, cognitive decline, and biologic markers. This study used functional connectivity MRI to investigate sex differences in the strength of functional connectivity of the default mode network in patients with MS and healthy control subjects. MATERIALS AND METHODS A total of 16 men and 16 women with MS and 32 age- and sex-matched healthy control subjects underwent a whole-brain resting-state functional connectivity MRI scan. A group-based seed in the posterior cingulate was used to create whole-brain correlation maps. A 2 × 2 ANOVA was used to assess whether disease status and sex affected the strength of connectivity to the posterior cingulate. RESULTS Patients with MS showed significantly stronger connectivity from the posterior cingulate to the bilateral medial frontal gyri, the left ventral anterior cingulate, the right putamen, and the left middle temporal gyrus (P < .0005). In the left dorsal lateral prefrontal cortex, female patients showed significantly stronger connectivity to the posterior cingulate cortex compared with female control subjects (P = 3 × 10(4)), and male control subjects showed stronger posterior cingulate cortex-left dorsal lateral prefrontal cortex connectivity in comparison to female control subjects (P = .002). Male patients showed significantly weaker connectivity to the caudate compared with female patients (P = .004). CONCLUSIONS Disease status and sex interact to produce differences in the strength of functional connectivity from the posterior cingulate to the caudate and the left dorsal lateral prefrontal cortex.
Collapse
|
145
|
Santner-Nanan B, Straubinger K, Hsu P, Parnell G, Tang B, Xu B, Makris A, Hennessy A, Peek MJ, Busch DH, da Costa CP, Nanan R. Fetal-maternal alignment of regulatory T cells correlates with IL-10 and Bcl-2 upregulation in pregnancy. THE JOURNAL OF IMMUNOLOGY 2013; 191:145-53. [PMID: 23733877 DOI: 10.4049/jimmunol.1203165] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Transplacental immune regulation refers to the concept that during pregnancy, significant cross-talk occurs between the maternal and fetal immune system with potential long-term effects for both the mother and child. In this study, we made the surprising observation that there is a strong correlation of peripheral blood regulatory T (Treg) cells between the mother and the fetus. In contrast, there is no significant Treg cell correlation between paternal fetal dyads (pairs), suggesting that the specific context of pregnancy, rather than the genetic parental similarity to the fetus, is responsible for this correlation. Gene microarray analysis of Treg cells identified a typical IL-10-dependent signature in maternal and fetal Treg cells. In addition, a direct correlation of serum IL-10 protein levels between maternal fetal dyads was observed. Furthermore, we show that maternal serum IL-10 levels correlate with serum estradiol and estriol, implicating hormonal involvement in this alignment. Interestingly, we show that Treg cells possess higher expression of IL-10 receptor α and that Treg cell IL-10 receptor α expression directly correlates with their Bcl-2 expression. Indeed, in vitro data in both humans and mice demonstrate that IL-10 upregulates Bcl-2 specifically in Treg cells but not non-Treg cells. Our results provide evidence for transplacental regulation of cellular immunity and suggest that IL-10 may influence Treg cell homeostasis through its effect on Treg cell Bcl-2 expression. These novel findings have important implications on immune tolerance in pregnancy and beyond in areas of autoimmunity, allergy, and transplantation.
Collapse
Affiliation(s)
- Brigitte Santner-Nanan
- Sydney Medical School Nepean, University of Sydney, Kingswood 2751, New South Wales, Australia
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
146
|
Reproductive organ involvement in non-Hodgkin lymphoma during pregnancy: a systematic review. Lancet Oncol 2013; 14:e275-82. [DOI: 10.1016/s1470-2045(12)70589-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
147
|
Wisdom AJ, Cao Y, Itoh N, Spence RD, Voskuhl RR. Estrogen receptor-β ligand treatment after disease onset is neuroprotective in the multiple sclerosis model. J Neurosci Res 2013; 91:901-8. [PMID: 23633287 DOI: 10.1002/jnr.23219] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Revised: 01/19/2013] [Accepted: 02/14/2013] [Indexed: 12/11/2022]
Abstract
Multiple sclerosis (MS) is an autoimmune disease characterized by inflammation and neurodegeneration. Current MS treatments were designed to reduce inflammation in MS rather than directly to prevent neurodegeneration. Estrogen has well-documented neuroprotective effects in a variety of disorders of the CNS, including experimental autoimmune encephalomyelitis (EAE), the most widely used mouse model of MS. Treatment with an estrogen receptor-β (ERβ) ligand is known to ameliorate clinical disease effectively and provide neuroprotection in EAE. However, the protective effects of this ERβ ligand have been demonstrated only when administered prior to disease (prophylactically). Here we tested whether ERβ ligand treatment could provide clinical protection when treatment was initiated after onset of disease (therapeutically). We found that therapeutic treatment effectively ameliorated clinical disease in EAE. Specifically, ERβ ligand-treated animals exhibited preserved axons and myelin compared with vehicle-treated animals. We observed no difference in the number of T lymphocytes, macrophages, or microglia in the CNS of vehicle- vs. ERβ ligand-treated animals. Our findings show that therapeutically administered ERβ ligand successfully treats clinical EAE, bearing translational relevance to MS as a candidate neuroprotective agent.
Collapse
Affiliation(s)
- Amy J Wisdom
- UCLA Multiple Sclerosis Program, Department of Neurology, University of California, Los Angeles, California 90095, USA
| | | | | | | | | |
Collapse
|
148
|
Bove R, Chitnis T. Sexual disparities in the incidence and course of MS. Clin Immunol 2013; 149:201-10. [PMID: 23608496 DOI: 10.1016/j.clim.2013.03.005] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Revised: 02/06/2013] [Accepted: 03/11/2013] [Indexed: 11/18/2022]
Abstract
Multiple sclerosis (MS) affects three times more women than men and this ratio appears to be increasing. However male patients experience increased disease progression, brain atrophy, and cognitive impairment. Gonadal hormones may modulate these sex differences. For example, female puberty heralds an increased risk of MS, and during pregnancy disease activity is milder, with an increased risk of postpartum relapses. Gonadal hormones likely have complex and inflammatory and neuroprotective effects, and may interact with other disease modulators, such as vitamin D. Sex differences in the heritability of disease susceptibility genes implicate a role for epigenetic modification. Many questions remain, including the impact of sex on treatment response and epigenetic changes, and the modulatory potential of hormonal treatments. This article summarizes what is known about sexual dimorphism in MS onset and course, as well as potential interactions between sex and other factors influencing MS pathogenesis, incidence and severity.
Collapse
Affiliation(s)
- Riley Bove
- Partners Multiple Sclerosis Center, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
149
|
Jolivel V, Luessi F, Masri J, Kraus SH, Hubo M, Poisa-Beiro L, Klebow S, Paterka M, Yogev N, Tumani H, Furlan R, Siffrin V, Jonuleit H, Zipp F, Waisman A. Modulation of dendritic cell properties by laquinimod as a mechanism for modulating multiple sclerosis. Brain 2013; 136:1048-66. [DOI: 10.1093/brain/awt023] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
|
150
|
Hu X, Qin X. Lentivirus-mediated estrogen receptor α overexpression in the central nervous system ameliorates experimental autoimmune encephalomyelitis in mice. Int J Mol Med 2013; 31:1209-21. [PMID: 23525227 DOI: 10.3892/ijmm.2013.1306] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Accepted: 03/05/2013] [Indexed: 11/05/2022] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disease characterized by inflammatory cell infiltration of the central nervous system (CNS) and multifocal demyelination. Clinical data and clinical indicators demonstrate that estrogen improves the relapse-remittance of MS patients. This study aimed to investigate the anti-inflammatory effects and the underlying mechanism(s) of action of estrogen and estrogen receptor α (ERα) in an experimental autoimmune encephalomyelitis (EAE) mouse model of MS. An ERα recombinant lentivirus was constructed. Mouse neurons were cultured in serum-free culture medium, and ERα recombinant lentivirus with a multiplicity of infection (MOI) of 5 was used to infect the neurons. Furthermore, neuronal ERα mRNA and protein expression were detected using real-time quantitative PCR and western blot analysis. We sterotaxically injected ERα recombinant lentivirus into the lateral ventricle of mouse brains, and successfully identified infected neurons using Flag immunofluorescence staining to determine the optimal dose. A total of 75 C57BL/6 mice were ovariectomized. After 2 weeks, EAE was induced with myelin oligodendrocyte glycoprotein (MOG) 35-55 peptide. The EAE mice were divided into 5 groups: the estrogen group (treatment with estradiol), the ERα agonist group (treatment with raloxifene), the ERα recombinant lentivirus group (ERα group, treatment with ERα recombinant lentivirus), the empty virus group and the normal saline (NS) group; clinical symptoms and body weight were compared among the groups. We assessed EAE-related parameters, detected pathological changes with immunohistochemistry and quantified the expression of myelin basic protein (MBP), matrix metalloproteinase-9 (MMP-9), and a subset of EAE-related cytokines using enzyme-linked immunosorbent assay (ELISA). We successfully constructed an ERα recombinant lentivirus. C57BL/6 mouse neurons can survive in culture for at least 8 weeks. During that period, the recombinant lentivirus was able to infect the neurons, while sustaining green fluorescence protein (GFP) expression. ERα recombinant lentivirus also infected the neurons at a MOI of 5. The ERα mRNA and protein expression levels were higher in the infected neurons compared to the uninfected ones. We successfully infected the CNS of C57BL/6 mice by stereotaxically injecting ERα recombinant lentivirus into the lateral ventricle of the mouse brains and induced EAE. The lentivirus-mediated overexpression of ERα reduced the incidence of EAE, ameliorated the clinical symptoms, inhibited inflammatory cell CNS infiltration, and reduced nerve fiber demyelination. MMP-9, tumor necrosis factor-α (TNF-α), interferon-γ (IFN-γ), interleukin (IL)-17 and IL-23 expression levels were decreased, while those of MBP and IL-4 were increased. These data demonstrate that it is possible to induce the overexpression of ERα using a recombinant lentivirus, and that this novel intervention ameliorates EAE in a mouse model. Mechanistically, estrogen and ERα inhibit inflammatory responses, and ERα alleviates damage to the myelin sheath. Collectively, our findings support the potential use of ERα as a therapeutic target for the treatment of MS.
Collapse
Affiliation(s)
- Xiao Hu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | | |
Collapse
|