51
|
Atkinson KC, Lee JB, Hasselmann JPC, Kim SH, Drew A, Soto J, Katzenellenbogen JA, Harris NG, Obenaus A, Tiwari-Woodruff SK. Diffusion tensor imaging identifies aspects of therapeutic estrogen receptor β ligand-induced remyelination in a mouse model of multiple sclerosis. Neurobiol Dis 2019; 130:104501. [PMID: 31226301 DOI: 10.1016/j.nbd.2019.104501] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Revised: 04/25/2019] [Accepted: 06/10/2019] [Indexed: 12/31/2022] Open
Abstract
Diffusion tensor imaging (DTI) has been shown to detect white matter degeneration in multiple sclerosis (MS), a neurodegenerative autoimmune disease that presents with diffuse demyelination of the central nervous system. However, the utility of DTI in evaluating therapeutic remyelination has not yet been well-established. Here, we assessed the ability of DTI to distinguish between remyelination and neuroprotection following estrogen receptor β ligand (Indazole chloride, IndCl) treatment, which has been previously shown to stimulate functional remyelination, in the cuprizone (CPZ) diet mouse model of MS. Adult C57BL/6 J male and female mice received a normal diet (control), demyelination-inducing CPZ diet (9wkDM), or CPZ diet followed by two weeks of a normal diet (i.e., remyelination period) with either IndCl (RM + IndCl) or vehicle (RM + Veh) injections. We evaluated tissue microstructure of the corpus callosum utilizing in vivo and ex vivo DTI and immunohistochemistry (IHC) for validation. Compared to control mice, the 9wkDM group showed decreased fractional anisotropy (FA), increased radial diffusivity (RD), and no changes in axial diffusivity (AD) both in vivo and ex vivo. Meanwhile, RM + IndCl groups showed increased FA and decreased RD ex vivo compared to the RM + Veh group, in accordance with the evidence of remyelination by IHC. In conclusion, the DTI technology used in the present study can identify some changes in myelination and is a valuable translational tool for evaluating MS pathophysiology and therapeutic efficacy.
Collapse
Affiliation(s)
- Kelley C Atkinson
- Division of Biomedical Sciences, School of Medicine at UCR, Riverside, CA, USA
| | - Jeong Bin Lee
- Division of Physiology, School of Medicine at Loma Linda University, Loma Linda, CA, USA
| | | | - Sung Hoon Kim
- Department of Chemistry, University of Illinois at Urbana-Champaign, IL, USA
| | - Alyson Drew
- Division of Physiology, School of Medicine at Loma Linda University, Loma Linda, CA, USA
| | - Joselyn Soto
- Division of Biomedical Sciences, School of Medicine at UCR, Riverside, CA, USA
| | | | - Neil G Harris
- Department of Neurosurgery, School of Medicine at UCLA, Los Angeles, CA, USA
| | - Andre Obenaus
- Division of Physiology, School of Medicine at Loma Linda University, Loma Linda, CA, USA; Department of Pediatrics, School of Medicine at UCI, Irvine, CA, USA
| | | |
Collapse
|
52
|
Mendibe Bilbao M, Boyero Durán S, Bárcena Llona J, Rodriguez-Antigüedad A. Multiple sclerosis: pregnancy and women's health issues. NEUROLOGÍA (ENGLISH EDITION) 2019. [DOI: 10.1016/j.nrleng.2016.06.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
53
|
Coyle PK, Oh J, Magyari M, Oreja-Guevara C, Houtchens M. Management strategies for female patients of reproductive potential with multiple sclerosis: An evidence-based review. Mult Scler Relat Disord 2019; 32:54-63. [PMID: 31030020 DOI: 10.1016/j.msard.2019.04.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 03/08/2019] [Accepted: 04/02/2019] [Indexed: 12/22/2022]
Abstract
Multiple sclerosis (MS) is an inflammatory, demyelinating, neurodegenerative, immune-mediated disease primarily diagnosed in early adulthood. Multiple sclerosis mostly impacts women of reproductive potential, with pregnancy and birth outcomes being major concerns for many patients. While there is ample evidence that the disease itself has no impact on pregnancy, many women living with MS still question their ability to have children, and the impact of childbearing on their disease in the short and long term. Such questions emphasize the importance of proper guidance from healthcare professionals, particularly neurologists. Management considerations are also complicated by the growing list of available treatment options. This review will summarize current evidence and expert opinion around the management of female MS patients of reproductive potential, from family planning to the postpartum period. Current guidelines on the use of disease-modifying therapies throughout pregnancy will be discussed, as well as other general medical recommendations, to minimize MS disease activity in the peripartum period.
Collapse
Affiliation(s)
- Patricia K Coyle
- Department of Neurology, Stony Brook University, Stony Brook, NY, USA.
| | - Jiwon Oh
- St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Melinda Magyari
- Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Celia Oreja-Guevara
- Hospital Clínico San Carlos, Universidad Complutense de Madrid, IdISSC, Madrid, Spain
| | - Maria Houtchens
- Brigham and Women's Hospital, Harvard Medical School, Brookline, MA, USA
| |
Collapse
|
54
|
Kim RY, Mangu D, Hoffman AS, Kavosh R, Jung E, Itoh N, Voskuhl R. Oestrogen receptor β ligand acts on CD11c+ cells to mediate protection in experimental autoimmune encephalomyelitis. Brain 2019; 141:132-147. [PMID: 29228214 PMCID: PMC5837360 DOI: 10.1093/brain/awx315] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 10/14/2017] [Indexed: 11/12/2022] Open
Abstract
Oestrogen treatments are neuroprotective in a variety of neurodegenerative disease models. Selective oestrogen receptor modifiers are needed to optimize beneficial effects while minimizing adverse effects to achieve neuroprotection in chronic diseases. Oestrogen receptor beta (ERβ) ligands are potential candidates. In the multiple sclerosis model chronic experimental autoimmune encephalomyelitis, ERβ-ligand treatment is neuroprotective, but mechanisms underlying this neuroprotection remain unclear. Specifically, whether there are direct effects of ERβ-ligand on CD11c+ microglia, myeloid dendritic cells or macrophages in vivo during disease is unknown. Here, we generated mice with ERβ deleted from CD11c+ cells to show direct effects of ERβ-ligand treatment in vivo on these cells to mediate neuroprotection during experimental autoimmune encephalomyelitis. Further, we use bone marrow chimeras to show that ERβ in peripherally derived myeloid cells, not resident microglia, are the CD11c+ cells mediating this protection. CD11c+ dendritic cell and macrophages isolated from the central nervous system of wild-type experimental autoimmune encephalomyelitis mice treated with ERβ-ligand expressed less iNOS and T-bet, but more IL-10, and this treatment effect was lost in mice with specific deletion of ERβ in CD11c+ cells. Also, we extend previous reports of ERβ-ligand’s ability to enhance remyelination through a direct effect on oligodendrocytes by showing that the immunomodulatory effect of ERβ-ligand acting on CD11c+ cells is necessary to permit the maturation of oligodendrocytes. Together these results demonstrate that targeting ERβ signalling pathways in CD11c+ myeloid cells is a novel strategy for regulation of the innate immune system in neurodegenerative diseases. To our knowledge, this is the first report showing how direct effects of a candidate neuroprotective treatment on two distinct cell lineages (bone marrow derived myeloid cells and oligodendrocytes) can have complementary neuroprotective effects in vivo.awx315media15688130498001.
Collapse
Affiliation(s)
- Roy Y Kim
- Multiple Sclerosis Program, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA.,Molecular, Cellular and Integrative Physiology Ph.D. Program, University of California, Los Angeles, CA 90095, USA
| | - Darian Mangu
- Multiple Sclerosis Program, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Alexandria S Hoffman
- Multiple Sclerosis Program, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Rojan Kavosh
- Multiple Sclerosis Program, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Eunice Jung
- Multiple Sclerosis Program, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Noriko Itoh
- Multiple Sclerosis Program, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Rhonda Voskuhl
- Multiple Sclerosis Program, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
55
|
Nuzzi R, Scalabrin S, Becco A, Panzica G. Sex Hormones and Optic Nerve Disorders: A Review. Front Neurosci 2019; 13:57. [PMID: 30804741 PMCID: PMC6378504 DOI: 10.3389/fnins.2019.00057] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 01/21/2019] [Indexed: 01/31/2023] Open
Abstract
Aim: This review article presents a comprehensive overview of the literature on sex hormones (estrogens, androgens, progesterone) and optic nerve disorders, with a discussion of the implications for therapy and prevention. Methods: Epidemiological, pre-clinical and clinical studies were reviewed. Results: Analysis of the biological basis for a relationship between eye diseases and sex hormones showed that some types of hormones can exert a protective effect either directly on the retina and optic nerve or indirectly by modulating ocular blood flow. For example, it seems that estrogen exposure has a protective effect against glaucoma, whereas its deficit may lead to early onset of the disease. If further studies confirm the data in the literature, estrogen therapy, because of its antioxidant action, may be effective in the treatment of Leber's hereditary optic neuropathy, whereas, in the light of current studies, there does not seem to be an influence of estrogen on non-arteritic anterior ischemic optic neuritis (NAION). Conclusions: Although there is some evidence that in some optic nerve pathologies the sex hormones seem to play an important role there are still too few studies providing evidence for its wider use in clinical practice.
Collapse
Affiliation(s)
- Raffaele Nuzzi
- Eye Clinic, Department of Surgical Sciences, AOU Città della Salute e della Scienza, Ophtalmic Clinic, University of Turin, Turin, Italy
| | - Simona Scalabrin
- Eye Clinic, Department of Surgical Sciences, AOU Città della Salute e della Scienza, Ophtalmic Clinic, University of Turin, Turin, Italy
| | - Alice Becco
- Eye Clinic, Department of Surgical Sciences, AOU Città della Salute e della Scienza, Ophtalmic Clinic, University of Turin, Turin, Italy
| | - Giancarlo Panzica
- Laboratory of Neuroendocrinology, Department of Neuroscience Rita Levi-Montalcini, University of Turin, Turin, Italy.,Neuroscience Institute Cavalieri-Ottolenghi, Orbassano, Italy
| |
Collapse
|
56
|
Iannello A, Rolla S, Maglione A, Ferrero G, Bardina V, Inaudi I, De Mercanti S, Novelli F, D'Antuono L, Cardaropoli S, Todros T, Turrini MV, Cordioli C, Puorro G, Marsili A, Lanzillo R, Brescia Morra V, Cordero F, De Bortoli M, Durelli L, Visconti A, Cutrupi S, Clerico M. Pregnancy Epigenetic Signature in T Helper 17 and T Regulatory Cells in Multiple Sclerosis. Front Immunol 2019; 9:3075. [PMID: 30671056 PMCID: PMC6331474 DOI: 10.3389/fimmu.2018.03075] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 12/12/2018] [Indexed: 12/22/2022] Open
Abstract
Increasing evidence supports the anti-inflammatory role of estrogens in Multiple Sclerosis (MS), originating from the observation of reduction in relapse rates among women with MS during pregnancy, but the molecular mechanisms are still not completely understood. Using an integrative data analysis, we identified T helper (Th) 17 and T regulatory (Treg) cell-type-specific regulatory regions (CSR) regulated by estrogen receptor alpha (ERα). These CSRs were validated in polarized Th17 from healthy donors (HD) and in peripheral blood mononuclear cells, Th17 and Treg cells from relapsing remitting (RR) MS patients and HD during pregnancy. 17β-estradiol induces active histone marks enrichment at Forkhead Box P3 (FOXP3)-CSRs and repressive histone marks enrichment at RAR related orphan receptor C (RORC)-CSRs in polarized Th17 cells. A disease-associated epigenetic profile was found in RRMS patients during pregnancy, suggesting a FOXP3 positive regulation and a RORC negative regulation in the third trimester of pregnancy. Altogether, these data indicate that estrogens act as immunomodulatory factors on the epigenomes of CD4+ T cells in RRMS; the identified CSRs may represent potential biomarkers for monitoring disease progression or new potential therapeutic targets.
Collapse
Affiliation(s)
- Andrea Iannello
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Simona Rolla
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Alessandro Maglione
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Giulio Ferrero
- Department of Computer Science, University of Turin, Turin, Italy
| | - Valentina Bardina
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Ilenia Inaudi
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Stefania De Mercanti
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Francesco Novelli
- Department of Molecular Biotechnology and Healthy Sciences, University of Turin, Turin, Italy
| | - Lucrezia D'Antuono
- Obstetric and Gynecologic Department, OIRM-Sant'Anna Hospital, Città della Salute e della Scienza, Turin, Italy
| | | | - Tullia Todros
- Department of Surgical Sciences, University of Turin, Turin, Italy
| | - Maria Vittoria Turrini
- Multiple Sclerosis Center, Ospedali Civili di Brescia, Montichiari Hospital, Montichiari, Italy
| | - Cinzia Cordioli
- Multiple Sclerosis Center, Ospedali Civili di Brescia, Montichiari Hospital, Montichiari, Italy
| | - Giorgia Puorro
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples Federico II, Naples, Italy
| | - Angela Marsili
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples Federico II, Naples, Italy
| | - Roberta Lanzillo
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples Federico II, Naples, Italy
| | - Vincenzo Brescia Morra
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples Federico II, Naples, Italy
| | | | - Michele De Bortoli
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Luca Durelli
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | | | - Santina Cutrupi
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Marinella Clerico
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| |
Collapse
|
57
|
Ohgomori T, Jinno S. Cuprizone-induced demyelination in the mouse hippocampus is alleviated by phytoestrogen genistein. Toxicol Appl Pharmacol 2019; 363:98-110. [DOI: 10.1016/j.taap.2018.11.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 11/15/2018] [Accepted: 11/19/2018] [Indexed: 12/28/2022]
|
58
|
Ysrraelit MC, Correale J. Impact of sex hormones on immune function and multiple sclerosis development. Immunology 2019; 156:9-22. [PMID: 30222193 PMCID: PMC6283654 DOI: 10.1111/imm.13004] [Citation(s) in RCA: 166] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 08/25/2018] [Accepted: 09/05/2018] [Indexed: 12/20/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) affecting young people and leading to demyelination and neurodegeneration. The disease is clearly more common in women, in whom incidence has been rising. Gender differences include: earlier disease onset and more frequent relapses in women; and faster progression and worse outcomes in men. Hormone-related physiological conditions in women such as puberty, pregnancy, puerperium, and menopause also exert significant influence both on disease prevalence as well as on outcomes. Hormonal and/or genetic factors are therefore believed to be involved in regulating the course of disease. In this review, we discuss clinical evidence for the impact of sex hormones (estrogens, progesterone, prolactin, and testosterone) on MS and attempt to elucidate the hormonal and immunological mechanisms potentially underlying these changes. We also review current knowledge on the relationship between sex hormones and resident CNS cells and provide new insights in the context of MS. Understanding these molecular mechanisms may contribute to the development of new and safer treatments for both men and women.
Collapse
Affiliation(s)
- María C. Ysrraelit
- Department of NeurologyRaúl Carrea Institute for Neurological Research (FLENI)Buenos AiresArgentina
| | - Jorge Correale
- Department of NeurologyRaúl Carrea Institute for Neurological Research (FLENI)Buenos AiresArgentina
| |
Collapse
|
59
|
Oliveira Filho RVD, Antunes NDJ, Ilha JDO, Moreno RA, Wedemeyer RS, Warnke A, De Nucci G. Pharmacokinetics and pharmacodynamics of three dosages of oestriol after continuous vaginal ring administration for 21 days in healthy, postmenopausal women. Br J Clin Pharmacol 2018; 85:551-562. [PMID: 30480330 DOI: 10.1111/bcp.13822] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 10/28/2018] [Accepted: 11/18/2018] [Indexed: 01/18/2023] Open
Affiliation(s)
| | - Natalícia de Jesus Antunes
- Department of Pharmacology, Faculty of Medical Sciences, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | | | - Ronilson Agnaldo Moreno
- Department of Pharmacology, Faculty of Medical Sciences, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | | | | | - Gilberto De Nucci
- Department of Pharmacology, Faculty of Medical Sciences, State University of Campinas (UNICAMP), Campinas, SP, Brazil.,Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
60
|
Ghanaatian N, Lashgari N, Abdolghaffari AH, Rajaee SM, Panahi Y, Barreto GE, Butler AE, Sahebkar A. Curcumin as a therapeutic candidate for multiple sclerosis: Molecular mechanisms and targets. J Cell Physiol 2018; 234:12237-12248. [DOI: 10.1002/jcp.27965] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 11/19/2018] [Indexed: 12/20/2022]
Affiliation(s)
- Negar Ghanaatian
- Faculty of Pharmacy, Pharmaceutical Sciences Branch, Islamic Azad University (IAUPS) Tehran Iran
| | - Naser‐Aldin Lashgari
- Faculty of Pharmacy, Pharmaceutical Sciences Branch, Islamic Azad University (IAUPS) Tehran Iran
| | - Amir Hossein Abdolghaffari
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR Karaj Iran
- Department of Pharmacology and Toxicology Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences Tehran Iran
- Department of Toxicology & Pharmacology Faculty of Pharmacy, Pharmaceutical Sciences Branch, Islamic Azad University (IAUPS) Tehran Iran
- Gastrointestinal Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN) Tehran Iran
| | - Seyed M. Rajaee
- Gastrointestinal Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN) Tehran Iran
| | - Yunes Panahi
- Clinical Pharmacy Department Faculty of Pharmacy, Baqiyatallah University of Medical Sciences Tehran Iran
| | - George E. Barreto
- Departamento de Nutrición y Bioquímica Facultad de Ciencias Pontificia Universidad Javeriana Bogotá Colombia
- Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile Santiago Chile
| | | | - Amirhossein Sahebkar
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences Mashhad Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences Mashhad Iran
- School of Pharmacy, Mashhad University of Medical Sciences Mashhad Iran
| |
Collapse
|
61
|
Landi D, Ragonese P, Prosperini L, Nociti V, Haggiag S, Cortese A, Fantozzi R, Pontecorvo S, Ferraro E, Buscarinu MC, Mataluni G, Monteleone F, Salvetti M, Di Battista G, Francia A, Millefiorini E, Gasperini C, Mirabella M, Salemi G, Boffa L, Pozzilli C, Centonze D, Marfia GA. Abortion induces reactivation of inflammation in relapsing-remitting multiple sclerosis. J Neurol Neurosurg Psychiatry 2018; 89:1272-1278. [PMID: 29970387 DOI: 10.1136/jnnp-2018-318468] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 05/25/2018] [Accepted: 06/11/2018] [Indexed: 02/04/2023]
Abstract
OBJECTIVE To investigate clinical and radiological outcomes of women with relapsing-remitting multiple sclerosis (RRMS) undergoing abortion. METHODS An independent, multicentre retrospective study was conducted collecting data from eight Italian MS centres. We compared the preconception and postabortion annualised relapse rate (ARR) and number of Gadolinium enhancing (Gd+) lesions, by analyses of covariance. Variables associated with postabortion clinical and MRI activity were investigated using Poisson regression models; each abortion was considered as a statistical unit. RESULTS From 1995 to 2017, we observed 188 abortions (17 elective) in 153 women with RRMS. Abortions occurred after a mean time of 9.5 (4.4) weeks from estimated conception date. In 86 events out of 188, conception happened during treatment with disease modifying drugs. The mean postabortion ARR (0.63±0.74) was significantly increased (p=0.037) compared with the preconception year (0.50±0.71) as well as the postabortion mean number of new Gd+ lesions (0.77±1.40 vs 0.39±1.04; p=0.004). Higher likelihood of relapses was predicted by higher preconception ARR, discontinuation of preconception treatment and elective abortion; the occurrence of new Gd+ lesions was associated with higher preconception number of active lesions, discontinuation of preconception treatment, shorter length of pregnancy maintenance and elective abortion. CONCLUSIONS Abortion was associated with clinical and radiological inflammatory rebound remarkably in the first 12 months postevent. Deregulated proinflammatory processes arising at the early stages of pregnancy might play a role both in MS reactivation and abortion. Women with MS should be counselled about these risks of abortion and followed up accordingly.
Collapse
Affiliation(s)
- Doriana Landi
- Multiple Sclerosis Clinical and Research Unit, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Paolo Ragonese
- Department of Biomedicine and Clinical Neuroscience, P. Giaccone University Hospital, Palermo, Italy
| | - Luca Prosperini
- Multiple Sclerosis Center, S. Camillo-Forlanini Hospital, Rome, Italy
| | - Viviana Nociti
- Multiple Sclerosis Center, Cattolica University, A. Gemelli University Hospital Foundation, Rome, Italy.,Neurorehabilitation Department, Don C. Gnocchi Foundation Onlus, Milan, Italy
| | - Shalom Haggiag
- Multiple Sclerosis Center, S. Camillo-Forlanini Hospital, Rome, Italy
| | - Antonio Cortese
- Multiple Sclerosis Center, Policlinico Umberto I, Department of Neurology and Psychiatry, Sapienza University, Rome, Italy
| | - Roberta Fantozzi
- Neurology and Neurorehabilitation Units, IRCCS Istituto Neurologico Mediterraneo NEUROMED, Pozzilli, Italy
| | - Simona Pontecorvo
- Multiple Sclerosis Center, Policlinico Umberto I, Department of Neurology and Psychiatry, Sapienza University, Rome, Italy
| | - Elisabetta Ferraro
- Multiple Sclerosis Center, San Filippo Neri Hospital - ASL Roma 1, Rome, Italy
| | - Maria Chiara Buscarinu
- Multiple Sclerosis Center, Department of Neurology and Psychiatry, Sapienza University, Rome, Italy
| | - Giorgia Mataluni
- Multiple Sclerosis Clinical and Research Unit, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Fabrizia Monteleone
- Multiple Sclerosis Clinical and Research Unit, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Marco Salvetti
- Neurology and Neurorehabilitation Units, IRCCS Istituto Neurologico Mediterraneo NEUROMED, Pozzilli, Italy.,Multiple Sclerosis Center, Department of Neurology and Psychiatry, Sapienza University, Rome, Italy
| | | | - Ada Francia
- Multiple Sclerosis Center, Policlinico Umberto I, Department of Neurology and Psychiatry, Sapienza University, Rome, Italy
| | - Enrico Millefiorini
- Multiple Sclerosis Center, Policlinico Umberto I, Department of Neurology and Psychiatry, Sapienza University, Rome, Italy
| | - Claudio Gasperini
- Multiple Sclerosis Center, S. Camillo-Forlanini Hospital, Rome, Italy
| | - Massimiliano Mirabella
- Multiple Sclerosis Center, Cattolica University, A. Gemelli University Hospital Foundation, Rome, Italy
| | - Giuseppe Salemi
- Department of Biomedicine and Clinical Neuroscience, P. Giaccone University Hospital, Palermo, Italy
| | - Laura Boffa
- Multiple Sclerosis Clinical and Research Unit, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Carlo Pozzilli
- Multiple Sclerosis Center, Department of Neurology and Psychiatry, Sapienza University, Rome, Italy
| | - Diego Centonze
- Multiple Sclerosis Clinical and Research Unit, Department of Systems Medicine, Tor Vergata University, Rome, Italy.,Neurology and Neurorehabilitation Units, IRCCS Istituto Neurologico Mediterraneo NEUROMED, Pozzilli, Italy
| | - Girolama Alessandra Marfia
- Multiple Sclerosis Clinical and Research Unit, Department of Systems Medicine, Tor Vergata University, Rome, Italy.,Neurology and Neurorehabilitation Units, IRCCS Istituto Neurologico Mediterraneo NEUROMED, Pozzilli, Italy
| |
Collapse
|
62
|
Wang CK, Lin CK, Wang TJ, Wang CY, Hsu PC, Su HY. Pregnancy and childbirth in a patient with multiple sclerosis: A case report of successful management. Taiwan J Obstet Gynecol 2018; 57:447-448. [PMID: 29880182 DOI: 10.1016/j.tjog.2018.04.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/30/2017] [Indexed: 11/25/2022] Open
Abstract
OBJECTIVE Multiple sclerosis (MS) preferentially affects females of reproductive age, making reproduction an important issue for women with MS. An increased incidence of poor labor progress often results in assisted vaginal delivery or a cesarean section. However, with good pre-pregnancy counseling and management, women with MS can conceive and give birth safely. Here, we present a case of pregnancy with MS, which was carried to term uneventfully and ended with unassisted vaginal delivery. CASE REPORT A 36-year-old woman was treated for MS for three years before she conceived. Because of her mild clinical presentation, medication was discontinued when her pregnancy was confirmed. Counseling was completed, and she had a smooth pregnancy course and gave birth vaginally at 38 weeks and two days. CONCLUSION Based on this case report, women with mild clinical presentation of MS before pregnancy can conceive and carry successfully to term with no or improved disease presentation.
Collapse
Affiliation(s)
- Chun-Kai Wang
- Department of Obstetrics and Gynecology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chi-Kang Lin
- Department of Obstetrics and Gynecology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Tun-Jun Wang
- Department of Obstetrics and Gynecology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chen-Yu Wang
- Department of Obstetrics and Gynecology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Po-Chao Hsu
- Department of Obstetrics and Gynecology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Her-Young Su
- Department of Obstetrics and Gynecology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.
| |
Collapse
|
63
|
Garnier L, Laffont S, Lélu K, Yogev N, Waisman A, Guéry JC. Estrogen Signaling in Bystander Foxp3neg CD4+ T Cells Suppresses Cognate Th17 Differentiation in Trans and Protects from Central Nervous System Autoimmunity. THE JOURNAL OF IMMUNOLOGY 2018; 201:3218-3228. [DOI: 10.4049/jimmunol.1800417] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 09/25/2018] [Indexed: 12/31/2022]
|
64
|
Moulton VR. Sex Hormones in Acquired Immunity and Autoimmune Disease. Front Immunol 2018; 9:2279. [PMID: 30337927 PMCID: PMC6180207 DOI: 10.3389/fimmu.2018.02279] [Citation(s) in RCA: 379] [Impact Index Per Article: 54.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 09/13/2018] [Indexed: 12/15/2022] Open
Abstract
Women have stronger immune responses to infections and vaccination than men. Paradoxically, the stronger immune response comes at a steep price, which is the high incidence of autoimmune diseases in women. The reasons why women have stronger immunity and higher incidence of autoimmunity are not clear. Besides gender, sex hormones contribute to the development and activity of the immune system, accounting for differences in gender-related immune responses. Both innate and adaptive immune systems bear receptors for sex hormones and respond to hormonal cues. This review focuses on the role of sex hormones particularly estrogen, in the adaptive immune response, in health, and autoimmune disease with an emphasis on systemic lupus erythematosus.
Collapse
Affiliation(s)
- Vaishali R Moulton
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
65
|
Tong Y, Liu J, Yang T, Kang Y, Wang J, Zhao T, Cheng C, Fan Y. Influences of pregnancy on neuromyelitis optica spectrum disorders and multiple sclerosis. Mult Scler Relat Disord 2018; 25:61-65. [DOI: 10.1016/j.msard.2018.07.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
66
|
Houtchens MK, Desai N. It is time to conduct phase 3 clinical trials of sex hormones in MS – No. Mult Scler 2018; 24:1415-1417. [DOI: 10.1177/1352458518771840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Maria K Houtchens
- Partners Multiple Sclerosis Center, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Neelam Desai
- Breast Oncology, Department of Medical Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
67
|
MacKenzie‐Graham A, Brook J, Kurth F, Itoh Y, Meyer C, Montag MJ, Wang H, Elashoff R, Voskuhl RR. Estriol-mediated neuroprotection in multiple sclerosis localized by voxel-based morphometry. Brain Behav 2018; 8:e01086. [PMID: 30144306 PMCID: PMC6160650 DOI: 10.1002/brb3.1086] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 07/05/2018] [Accepted: 07/08/2018] [Indexed: 11/11/2022] Open
Abstract
INTRODUCTION Progressive gray matter (GM) atrophy is a hallmark of multiple sclerosis (MS). Cognitive impairment has been observed in 40%-70% of MS patients and has been linked to GM atrophy. In a phase 2 trial of estriol treatment in women with relapsing-remitting MS (RRMS), higher estriol levels correlated with greater improvement on the paced auditory serial addition test (PASAT) and imaging revealed sparing of localized GM in estriol-treated compared to placebo-treated patients. To better understand the significance of this GM sparing, the current study explored the relationships between the GM sparing and traditional MRI measures and clinical outcomes. METHODS Sixty-two estriol- and forty-nine placebo-treated RRMS patients underwent clinical evaluations and brain MRI. Voxel-based morphometry (VBM) was used to evaluate voxelwise GM sparing from high-resolution T1-weighted scans. RESULTS A region of treatment-induced sparing (TIS) was defined as the areas where GM was spared in estriol- as compared to placebo-treated groups, localized primarily within the frontal and parietal cortices. We observed that TIS volume was directly correlated with improvement on the PASAT. Next, a longitudinal cognitive disability-specific atlas (DSA) was defined by correlating voxelwise GM volumes with PASAT scores, that is, areas where less GM correlated with less improvement in PASAT scores. Finally, overlap between the TIS and the longitudinal cognitive DSA revealed a specific region of cortical GM that was preserved in estriol-treated subjects that was associated with better performance on the PASAT. CONCLUSIONS Discovery of this region of overlap was biology driven, not based on an a priori structure of interest. It included the medial frontal cortex, an area previously implicated in problem solving and attention. These findings indicate that localized GM sparing during estriol treatment was associated with improvement in cognitive testing, suggesting a clinically relevant, disability-specific biomarker for clinical trials of candidate neuroprotective treatments in MS.
Collapse
Affiliation(s)
- Allan MacKenzie‐Graham
- Department of NeurologyAhmanson‐Lovelace Brain Mapping CenterDavid Geffen School of Medicine at UCLALos AngelesCalifornia
- UCLA Multiple Sclerosis ProgramDepartment of NeurologyDavid Geffen School of Medicine at UCLALos AngelesCalifornia
| | - Jenny Brook
- Department of BiomathematicsDavid Geffen School of Medicine at UCLALos AngelesCalifornia
| | - Florian Kurth
- Department of NeurologyAhmanson‐Lovelace Brain Mapping CenterDavid Geffen School of Medicine at UCLALos AngelesCalifornia
- UCLA Multiple Sclerosis ProgramDepartment of NeurologyDavid Geffen School of Medicine at UCLALos AngelesCalifornia
| | - Yuichiro Itoh
- UCLA Multiple Sclerosis ProgramDepartment of NeurologyDavid Geffen School of Medicine at UCLALos AngelesCalifornia
| | - Cassandra Meyer
- Department of NeurologyAhmanson‐Lovelace Brain Mapping CenterDavid Geffen School of Medicine at UCLALos AngelesCalifornia
- UCLA Multiple Sclerosis ProgramDepartment of NeurologyDavid Geffen School of Medicine at UCLALos AngelesCalifornia
| | - Michael J. Montag
- UCLA Multiple Sclerosis ProgramDepartment of NeurologyDavid Geffen School of Medicine at UCLALos AngelesCalifornia
| | - He‐Jing Wang
- Department of BiomathematicsDavid Geffen School of Medicine at UCLALos AngelesCalifornia
| | - Robert Elashoff
- Department of BiomathematicsDavid Geffen School of Medicine at UCLALos AngelesCalifornia
| | - Rhonda R. Voskuhl
- UCLA Multiple Sclerosis ProgramDepartment of NeurologyDavid Geffen School of Medicine at UCLALos AngelesCalifornia
| |
Collapse
|
68
|
Pinares-Garcia P, Stratikopoulos M, Zagato A, Loke H, Lee J. Sex: A Significant Risk Factor for Neurodevelopmental and Neurodegenerative Disorders. Brain Sci 2018; 8:E154. [PMID: 30104506 PMCID: PMC6120011 DOI: 10.3390/brainsci8080154] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 08/08/2018] [Accepted: 08/10/2018] [Indexed: 12/11/2022] Open
Abstract
Males and females sometimes significantly differ in their propensity to develop neurological disorders. Females suffer more from mood disorders such as depression and anxiety, whereas males are more susceptible to deficits in the dopamine system including Parkinson's disease (PD), attention-deficit hyperactivity disorder (ADHD) and autism. Despite this, biological sex is rarely considered when making treatment decisions in neurological disorders. A better understanding of the molecular mechanism(s) underlying sex differences in the healthy and diseased brain will help to devise diagnostic and therapeutic strategies optimal for each sex. Thus, the aim of this review is to discuss the available evidence on sex differences in neuropsychiatric and neurodegenerative disorders regarding prevalence, progression, symptoms and response to therapy. We also discuss the sex-related factors such as gonadal sex hormones and sex chromosome genes and how these might help to explain some of the clinically observed sex differences in these disorders. In particular, we highlight the emerging role of the Y-chromosome gene, SRY, in the male brain and its potential role as a male-specific risk factor for disorders such as PD, autism, and ADHD in many individuals.
Collapse
Affiliation(s)
- Paulo Pinares-Garcia
- Brain and Gender laboratory, Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia.
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria 3168, Australia.
| | - Marielle Stratikopoulos
- Brain and Gender laboratory, Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia.
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria 3168, Australia.
| | - Alice Zagato
- Brain and Gender laboratory, Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia.
- School of Life and Environmental Sciences, Deakin University, Burwood, Victoria 3125, Australia.
| | - Hannah Loke
- Brain and Gender laboratory, Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia.
| | - Joohyung Lee
- Brain and Gender laboratory, Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia.
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria 3168, Australia.
| |
Collapse
|
69
|
Voskuhl R. It is time to conduct phase 3 clinical trials of sex hormones in MS - Yes. Mult Scler 2018; 24:1413-1415. [PMID: 30058469 DOI: 10.1177/1352458518768764] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Rhonda Voskuhl
- Multiple Sclerosis Program, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
70
|
Collongues N, Patte-Mensah C, De Seze J, Mensah-Nyagan AG, Derfuss T. Testosterone and estrogen in multiple sclerosis: from pathophysiology to therapeutics. Expert Rev Neurother 2018; 18:515-522. [PMID: 29799288 DOI: 10.1080/14737175.2018.1481390] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Neuroprotection and remyelination are two unmet needs in the treatment of multiple sclerosis (MS). Therapeutic potential has been identified with sexual hormones, supported in women by a decrease in MS activity during the pregnancy, in men by a greater severity of symptoms and a faster progression than in women. Areas covered: The therapeutic effect of testosterone and estrogens is reviewed. Both hormones have demonstrated an anti-inflammatory effect. Testosterone has an effect in protecting neurons in culture against glutamate-induced toxicity and oxidative stress, and stimulates myelin formation and regeneration mediated through the neural androgen receptor. In experimental autoimmune encephalomyelitis model, estrogens significantly decrease inflammation in the central nervous system via ERα, while its action on ERβ leads to myelin and axon reparation. Estriol therapy in two phase 2 trials showed a decrease in clinical disease activity and inflammatory parameters in MRI. However, evidence of a therapeutic effect of testosterone is scarce. Expert commentary: Phase 3 trials with estriol as an add-on supplementation are now mandatory. Testosterone is another candidate to be tested in phase 2 trials. These hormones should be considered as an adjunctive therapy. New validated tools are needed to assess their effect on neuroprotection and remyelination.
Collapse
Affiliation(s)
- Nicolas Collongues
- a Biopathology of Myelin, Neuroprotection and Therapeutic Strategies , INSERM U1119, University Hospital of Strasbourg , Strasbourg , France.,b Department of Neurology , University Hospital of Strasbourg , Strasbourg , France.,c Clinical Investigation Center , INSERM U1434, University Hospital of Strasbourg , Strasbourg , France
| | - Christine Patte-Mensah
- a Biopathology of Myelin, Neuroprotection and Therapeutic Strategies , INSERM U1119, University Hospital of Strasbourg , Strasbourg , France
| | - Jérôme De Seze
- a Biopathology of Myelin, Neuroprotection and Therapeutic Strategies , INSERM U1119, University Hospital of Strasbourg , Strasbourg , France.,b Department of Neurology , University Hospital of Strasbourg , Strasbourg , France.,c Clinical Investigation Center , INSERM U1434, University Hospital of Strasbourg , Strasbourg , France
| | - Ayikoe-Guy Mensah-Nyagan
- a Biopathology of Myelin, Neuroprotection and Therapeutic Strategies , INSERM U1119, University Hospital of Strasbourg , Strasbourg , France
| | - Tobias Derfuss
- d Departments of Neurology and Biomedicine , University Hospital Basel , Basel , Switzerland
| |
Collapse
|
71
|
Increase in chemokine CXCL1 by ERβ ligand treatment is a key mediator in promoting axon myelination. Proc Natl Acad Sci U S A 2018; 115:6291-6296. [PMID: 29844175 PMCID: PMC6004485 DOI: 10.1073/pnas.1721732115] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Estrogen receptor β (ERβ) ligands promote remyelination in mouse models of multiple sclerosis. Recent work using experimental autoimmune encephalomyelitis (EAE) has shown that ERβ ligands induce axon remyelination, but impact peripheral inflammation to varying degrees. To identify if ERβ ligands initiate a common immune mechanism in remyelination, central and peripheral immunity and pathology in mice given ERβ ligands at peak EAE were assessed. All ERβ ligands induced differential expression of cytokines and chemokines, but increased levels of CXCL1 in the periphery and in astrocytes. Oligodendrocyte CXCR2 binds CXCL1 and has been implicated in normal myelination. In addition, despite extensive immune cell accumulation in the CNS, all ERβ ligands promoted extensive remyelination in mice at peak EAE. This finding highlights a component of the mechanism by which ERβ ligands mediate remyelination. Hence, interplay between the immune system and central nervous system may be responsible for the remyelinating effects of ERβ ligands. Our findings of potential neuroprotective benefits arising from the presence of CXCL1 could have implications for improved therapies for multiple sclerosis.
Collapse
|
72
|
Abstract
OBJECTIVE Estriol is the main estrogen in pregnancy, but has received less attention outside gestation. It is well known that pregnancy has an immunosuppressive effect on many autoimmune diseases such as multiple sclerosis, psoriasis, thyroiditis, uveitis, and rheumatoid arthritis. Emerging evidence indicates that estriol has potential immunomodulatory benefits for many disease states including autoimmune, inflammatory, and neurodegenerative conditions. In this review, we discuss emerging roles for estriol in the treatment of menopausal symptoms, osteoporosis, cancer, hyperlipidemia, vascular disease, and multiple sclerosis. Estriol appears to offer a potentially cost-effective approach to a variety of conditions and may offer a wide range of health benefits. METHODS We reviewed the English language MEDLINE literature with estriol in the title with emphasis on publications including nonpregnant females between January 1974 and August 2016. Approximately 393 such articles were considered and 72 articles have been referenced in this review. RESULTS Estriol offers considerable benefits for postmenopausal women with reduced risks that are normally associated with traditional hormone therapies. These benefits include improved control of menopausal symptoms and better urogenital health. Moreover, the immunomodulatory role of estriol in reducing proinflammatory cytokines may be an important new therapeutic option for chronic autoimmune and neurodegenerative illnesses. Since it is a relatively weak estrogen, there is potential for use in men for conditions such as multiple sclerosis. CONCLUSIONS We conclude transvaginal estriol potentially offers a suitable physiologic delivery and cost-effective alternative to currently available estrogen regimens in selected patients. Additional studies on mode of delivery, safety, and efficacy merit further investigation.
Collapse
|
73
|
Kaisey M, Sicotte N, Giesser B. Multiple sclerosis management and reproductive changes: A guide for general neurologists. Neurol Clin Pract 2018; 8:142-147. [PMID: 29708217 DOI: 10.1212/cpj.0000000000000436] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 01/17/2018] [Indexed: 11/15/2022]
Abstract
Purpose of review Multiple sclerosis (MS) disease activity and symptoms are tied to hormonal changes. This review explains the current standard of care in MS at various stages of a woman's reproductive life and helps neurologists answer patients' most common questions surrounding MS care and fertility, pregnancy, and menopause. Recent findings Recent work has focused on MS risk and exacerbation with variables related to reproductive health. Management of disease-modifying therapies prenatally and postnatally is also a focus. Summary This review is a concise, practical guide for general neurologists caring for women with MS. MS is a disease that requires adaptation of management as a woman moves through reproductive stages. With proper planning and management, pregnancy is safe for women with MS. We describe the current standard of care based on trials, when available, and on expert opinion.
Collapse
Affiliation(s)
- Marwa Kaisey
- Department of Neurology (MK, MS), Cedars-Sinai Medical Center, Los Angeles, CA; and Department of Neurology (BG), University of California, Los Angeles
| | - Nancy Sicotte
- Department of Neurology (MK, MS), Cedars-Sinai Medical Center, Los Angeles, CA; and Department of Neurology (BG), University of California, Los Angeles
| | - Barbara Giesser
- Department of Neurology (MK, MS), Cedars-Sinai Medical Center, Los Angeles, CA; and Department of Neurology (BG), University of California, Los Angeles
| |
Collapse
|
74
|
Kempe P, Eklund D, Hallin A, Hammar M, Olsson T, Brynhildsen J, Ernerudh J. Immune profile in relation to sex steroid cyclicity in healthy women and women with multiple sclerosis. J Reprod Immunol 2018; 126:53-59. [PMID: 29501895 DOI: 10.1016/j.jri.2018.02.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2017] [Revised: 02/11/2018] [Accepted: 02/24/2018] [Indexed: 10/17/2022]
Abstract
To prospectively study systemic in vivo immunological effects of sex hormones, using different phases of oral combined hormonal contraceptives (CHC), and the natural menstrual cycles in both healthy women and in women with multiple sclerosis (MS), blood samples from sixty female MS patients and healthy controls with and without CHC were drawn in high and low estrogenic/progestogenic phases. Expression of Th-associated genes in blood cells was determined by qPCR and a panel of cytokines and chemokines was measured in plasma. High hormone level phases were associated with increases in Th1 (TBX21) and Th2 (GATA3) associated markers, as well as the B cell-associated chemokine CXCL13, while the inhibitory regulator CTLA-4 was decreased. These changes were not observed in MS patients, of whom most were treated with immunomodulatory drugs. Our data indicate immune activating properties in vivo of high steroid sex hormone levels during both CHC and normal menstrual cyclicity.
Collapse
Affiliation(s)
- Per Kempe
- Obstetrics and Gynaecology, Department of Clinical and Experimental Medicine, Linköping University, SE-58185, Linköping, Sweden; Department of Obstetrics & Gynaecology, County Hospital Sundsvall, SE-85643, Sundsvall, Sweden
| | - Daniel Eklund
- Department of Clinical and Experimental Medicine, Linköping University, SE-58185, Linköping, Sweden
| | - Agnes Hallin
- Department of Clinical and Experimental Medicine, Linköping University, SE-58185, Linköping, Sweden
| | - Mats Hammar
- Obstetrics and Gynaecology, Department of Clinical and Experimental Medicine, Linköping University, SE-58185, Linköping, Sweden
| | - Tomas Olsson
- Department of Clinical Neuroscience, Karolinska Institutet, SE-17177, Stockholm, Sweden
| | - Jan Brynhildsen
- Obstetrics and Gynaecology, Department of Clinical and Experimental Medicine, Linköping University, SE-58185, Linköping, Sweden.
| | - Jan Ernerudh
- Clinical Immunology and Transfusion Medicine, Department of Clinical and Experimental Medicine, Linköping University, SE-58185, Linköping, Sweden
| |
Collapse
|
75
|
Salpietro V, Polizzi A, Recca G, Ruggieri M. The role of puberty and adolescence in the pathobiology of pediatric multiple sclerosis. ACTA ACUST UNITED AC 2018. [DOI: 10.1186/s40893-017-0032-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
76
|
Cell-specific and region-specific transcriptomics in the multiple sclerosis model: Focus on astrocytes. Proc Natl Acad Sci U S A 2018; 115:E302-E309. [PMID: 29279367 PMCID: PMC5777065 DOI: 10.1073/pnas.1716032115] [Citation(s) in RCA: 184] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Changes in gene expression that occur across the central nervous system (CNS) during neurological diseases do not address the heterogeneity of cell types from one CNS region to another and are complicated by alterations in cellular composition during disease. Multiple sclerosis (MS) is multifocal by definition. Here, a cell-specific and region-specific transcriptomics approach was used to determine gene expression changes in astrocytes in the most widely used MS model, experimental autoimmune encephalomyelitis (EAE). Astrocyte-specific RNAs from various neuroanatomic regions were attained using RiboTag technology. Sequencing and bioinformatics analyses showed that EAE-induced gene expression changes differed between neuroanatomic regions when comparing astrocytes from spinal cord, cerebellum, cerebral cortex, and hippocampus. The top gene pathways that were changed in astrocytes from spinal cord during chronic EAE involved decreases in expression of cholesterol synthesis genes while immune pathway gene expression in astrocytes was increased. Optic nerve from EAE and optic chiasm from MS also showed decreased cholesterol synthesis gene expression. The potential role of cholesterol synthesized by astrocytes during EAE and MS is discussed. Together, this provides proof-of-concept that a cell-specific and region-specific gene expression approach can provide potential treatment targets in distinct neuroanatomic regions during multifocal neurological diseases.
Collapse
|
77
|
|
78
|
Golden LC, Voskuhl R. The importance of studying sex differences in disease: The example of multiple sclerosis. J Neurosci Res 2017; 95:633-643. [PMID: 27870415 DOI: 10.1002/jnr.23955] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 08/19/2016] [Accepted: 09/06/2016] [Indexed: 12/20/2022]
Abstract
To date, scientific research has often focused on one sex, with assumptions that study of the other sex would yield similar results. However, many diseases affect males and females differently. The sex of a patient can affect the risk for both disease susceptibility and progression. Such differences can be brought to the laboratory bench to be investigated, potentially bringing new treatments back to the clinic. This method of research, known as a "bedside to bench to bedside" approach, has been applied to studying sex differences in multiple sclerosis (MS). Females have greater susceptibly to MS, while males have worse disease progression. These two characteristics of the disease are influenced by the immune system and the nervous system, respectively. Thus, sex differences in each system must be studied. Personalized medicine has been at the forefront of research recently, and studying sex differences in disease fits with this initiative. This review will discuss the known sex differences in MS and highlight how investigating them can lead to new insights and potential treatments for both men and women. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Lisa C Golden
- Department of Neurology, University of California Los Angeles, Los Angeles, California.,Molecular Biology IDP, University of California Los Angeles, Los Angeles, California
| | - Rhonda Voskuhl
- Department of Neurology, University of California Los Angeles, Los Angeles, California
| |
Collapse
|
79
|
Massa MG, David C, Jörg S, Berg J, Gisevius B, Hirschberg S, Linker RA, Gold R, Haghikia A. Testosterone Differentially Affects T Cells and Neurons in Murine and Human Models of Neuroinflammation and Neurodegeneration. THE AMERICAN JOURNAL OF PATHOLOGY 2017. [PMID: 28634006 DOI: 10.1016/j.ajpath.2017.03.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The high female-to-male sex ratio of multiple sclerosis (MS) prevalence has continuously confounded researchers, especially in light of male patients' accelerated disease course at later stages of MS. Although multiple studies have concentrated on estrogenic mechanisms of disease modulation, fairly little attention has been paid to androgenic effects in a female system, and even fewer studies have attempted to dissociate hormonal effects on the neurodegenerative and neuroinflammatory processes of MS. Herein, we demonstrate the differential effects of hormone treatment on the acute inflammatory and chronic neurodegenerative phases of murine experimental autoimmune encephalomyelitis. Although s.c. treatment with testosterone and aromatase inhibitor applied beginning on the day of immunization ameliorated initial course of disease, similar treatment administered therapeutically exacerbated chronic disease course. Spinal cord analyses of axonal densities reflected the clinical scores of the chronic phase. In vitro, testosterone treatment not only decreased Th1 and Th17 differentiation in an aromatase-independent fashion, but also exacerbated cell death in induced pluripotent stem cell-derived primary human neurons under oxidative stress conditions in an aromatase inhibitor-dependent manner. Thus, through the alleviation of inflammatory processes and the exacerbation of neurodegenerative processes, androgens may contribute to the epidemiologic sex differentials observed in MS prevalence and course.
Collapse
Affiliation(s)
- Megan G Massa
- Department of Neurology, Ruhr University-Bochum, Bochum, Germany
| | - Christina David
- Department of Neurology, Ruhr University-Bochum, Bochum, Germany
| | - Stefanie Jörg
- Department of Neurology, Friedrich-Alexander University-Erlangen-Nuremberg, Erlangen, Germany
| | - Johannes Berg
- Department of Neurology, Ruhr University-Bochum, Bochum, Germany
| | - Barbara Gisevius
- Department of Neurology, Ruhr University-Bochum, Bochum, Germany
| | - Sarah Hirschberg
- Department of Neurology, Ruhr University-Bochum, Bochum, Germany
| | - Ralf A Linker
- Department of Neurology, Friedrich-Alexander University-Erlangen-Nuremberg, Erlangen, Germany
| | - Ralf Gold
- Department of Neurology, Ruhr University-Bochum, Bochum, Germany
| | - Aiden Haghikia
- Department of Neurology, Ruhr University-Bochum, Bochum, Germany.
| |
Collapse
|
80
|
Voskuhl R, Momtazee C. Pregnancy: Effect on Multiple Sclerosis, Treatment Considerations, and Breastfeeding. Neurotherapeutics 2017; 14:974-984. [PMID: 28766273 PMCID: PMC5722767 DOI: 10.1007/s13311-017-0562-7] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Multiple sclerosis (MS) commonly affects women in childbearing years making pregnancy issues important for patients with MS and their families. Pregnancy is a naturally occurring disease modifier of MS associated with a 70% reduction in relapse rates in the third trimester. This relapse rate reduction during the last trimester is roughly equal to the most effective disease-modifying treatments for MS. Given this efficacy, various pregnancy factors have been tested to determine which play a part in pregnancy's protection, and some have been translated to completed and ongoing phase II clinical trials. In contrast to protective effects during pregnancy, the postpartum period entails increased relapse risk, which may be due to either abrupt removal of protective pregnancy factors after delivery or to unique deleterious factors inherent to the postpartum period. The effect of breastfeeding on MS remains unclear. The best predictor for whether a patient will have a postpartum relapse is the incidence of her having active relapsing MS prior to pregnancy. The medical management of MS during pregnancy and the postpartum period is challenging given the risks of medication exposure to the fetus in utero and to the infant through breast milk. This review will focus on clinical aspects of pregnancy, including the effects of pregnancy on MS disease activity, as well as the medical management of MS during pregnancy and postpartum.
Collapse
Affiliation(s)
- Rhonda Voskuhl
- Multiple Sclerosis Program, UCLA Department of Neurology, David Geffen School of Medicine, University of Los Angeles, Los Angeles, CA, 90095, USA.
| | - Callene Momtazee
- Multiple Sclerosis Program, UCLA Department of Neurology, David Geffen School of Medicine, University of Los Angeles, Los Angeles, CA, 90095, USA
| |
Collapse
|
81
|
Glucocorticoid receptor in T cells mediates protection from autoimmunity in pregnancy. Proc Natl Acad Sci U S A 2017; 114:E181-E190. [PMID: 28049829 DOI: 10.1073/pnas.1617115114] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Pregnancy is one of the strongest inducers of immunological tolerance. Disease activity of many autoimmune diseases including multiple sclerosis (MS) is temporarily suppressed by pregnancy, but little is known about the underlying molecular mechanisms. Here, we investigated the endocrine regulation of conventional and regulatory T cells (Tregs) during reproduction. In vitro, we found the pregnancy hormone progesterone to robustly increase Treg frequencies via promiscuous binding to the glucocorticoid receptor (GR) in T cells. In vivo, T-cell-specific GR deletion in pregnant animals undergoing experimental autoimmune encephalomyelitis (EAE), the animal model of MS, resulted in a reduced Treg increase and a selective loss of pregnancy-induced protection, whereas reproductive success was unaffected. Our data imply that steroid hormones can shift the immunological balance in favor of Tregs via differential engagement of the GR in T cells. This newly defined mechanism confers protection from autoimmunity during pregnancy and represents a potential target for future therapy.
Collapse
|
82
|
Itoh N, Kim R, Peng M, DiFilippo E, Johnsonbaugh H, MacKenzie-Graham A, Voskuhl RR. Bedside to bench to bedside research: Estrogen receptor beta ligand as a candidate neuroprotective treatment for multiple sclerosis. J Neuroimmunol 2016; 304:63-71. [PMID: 27771018 DOI: 10.1016/j.jneuroim.2016.09.017] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 09/28/2016] [Indexed: 12/16/2022]
Abstract
Protective effects of pregnancy during MS have led to clinical trials of estriol, the pregnancy estrogen, in MS. Since estriol binds to estrogen receptor (ER) beta, ER beta ligand could represent a "next generation estriol" treatment. Here, ER beta ligand treatment was protective in EAE in both sexes and across genetic backgrounds. Neuroprotection was shown in spinal cord, sparing myelin and axons, and in brain, sparing neurons and synapses. Longitudinal in vivo MRIs showed decreased brain atrophy in cerebral cortex gray matter and cerebellum during EAE. Investigation of ER beta ligand as a neuroprotective treatment for MS is warranted.
Collapse
Affiliation(s)
- Noriko Itoh
- Department of Neurology, University of California, Los Angeles, David Geffen School of Medicine, USA
| | - Roy Kim
- Department of Neurology, University of California, Los Angeles, David Geffen School of Medicine, USA
| | - Mavis Peng
- Department of Neurology, University of California, Los Angeles, David Geffen School of Medicine, USA
| | - Emma DiFilippo
- Department of Neurology, University of California, Los Angeles, David Geffen School of Medicine, USA
| | - Hadley Johnsonbaugh
- Department of Neurology, University of California, Los Angeles, David Geffen School of Medicine, USA
| | - Allan MacKenzie-Graham
- Department of Neurology, University of California, Los Angeles, David Geffen School of Medicine, USA
| | - Rhonda R Voskuhl
- Department of Neurology, University of California, Los Angeles, David Geffen School of Medicine, USA.
| |
Collapse
|
83
|
Development of a high throughput drug screening assay to identify compounds that protect oligodendrocyte viability and differentiation under inflammatory conditions. BMC Res Notes 2016; 9:444. [PMID: 27629829 PMCID: PMC5024459 DOI: 10.1186/s13104-016-2219-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 08/15/2016] [Indexed: 11/29/2022] Open
Abstract
Background Newly proliferated oligodendrocyte precursor cells (OPCs) migrate and surround lesions of patients with multiple sclerosis (MS) and other demyelinating diseases, but fail to differentiate into oligodendrocytes (OLs) and remyelinate remaining viable axons. The abundance of secreted inflammatory factors within and surrounding these lesions likely plays a major inhibitory role, promoting cell death and preventing OL differentiation and axon remyelination. To identify clinical candidate compounds that may protect existing and differentiating OLs in patients, we have developed a high throughput screening (HTS) assay that utilizes purified rat OPCs. Results Using a fluorescent indicator of cell viability coupled with image quantification, we developed an assay to allow the identification of compounds that promote OL viability and differentiation in the presence of the synergistic inflammatory cytokines, tumor necrosis factor α and interferon-γ. We have utilized this assay to screen the NIH clinical collection library and identify compounds that protect OLs and promote OL differentiation in the presence of these inflammatory cytokines. Conclusion This primary OL-based cytokine protection assay is adaptable for HTS and may be easily modified for profiling of compounds in the presence of other potentially inhibitory molecules found in MS lesions. This assay should be of use to those interested in identifying drugs for the treatment of MS and other demyelinating diseases. Electronic supplementary material The online version of this article (doi:10.1186/s13104-016-2219-8) contains supplementary material, which is available to authorized users.
Collapse
|
84
|
Lariosa-Willingham KD, Rosler ES, Tung JS, Dugas JC, Collins TL, Leonoudakis D. A high throughput drug screening assay to identify compounds that promote oligodendrocyte differentiation using acutely dissociated and purified oligodendrocyte precursor cells. BMC Res Notes 2016; 9:419. [PMID: 27592856 PMCID: PMC5011342 DOI: 10.1186/s13104-016-2220-2] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 08/15/2016] [Indexed: 12/20/2022] Open
Abstract
Background Multiple sclerosis is caused by an autoimmune response resulting in demyelination and neural degeneration. The adult central nervous system has the capacity to remyelinate axons in part through the generation of new oligodendrocytes (OLs). To identify clinical candidate compounds that may promote remyelination, we have developed a high throughput screening (HTS) assay to identify compounds that promote the differentiation of oligodendrocyte precursor cells (OPCs) into OLs. Results Using acutely dissociated and purified rat OPCs coupled with immunofluorescent image quantification, we have developed an OL differentiation assay. We have validated this assay with a known promoter of differentiation, thyroid hormone, and subsequently used the assay to screen the NIH clinical collection library. We have identified twenty-seven hit compounds which were validated by dose response analysis and the generation of half maximal effective concentration (EC50) values allowed for the ranking of efficacy. The assay identified novel promoters of OL differentiation which we attribute to (1) the incorporation of an OL toxicity pre-screen to allow lowering the concentrations of toxic compounds and (2) the utilization of freshly purified, non-passaged OPCs. These features set our assay apart from other OL differentiation assays used for drug discovery efforts. Conclusions This acute primary OL-based differentiation assay should be of use to those interested in screening large compound libraries for the identification of drugs for the treatment of MS and other demyelinating diseases. Electronic supplementary material The online version of this article (doi:10.1186/s13104-016-2220-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Karen D Lariosa-Willingham
- Translational Medicine Center, Myelin Repair Foundation, Sunnyvale, CA, 94085, USA.,Teva Pharmaceuticals, Biologics and CNS Discovery, Redwood City, CA, 94063, USA
| | - Elen S Rosler
- Translational Medicine Center, Myelin Repair Foundation, Sunnyvale, CA, 94085, USA.,Alios BioPharma, South San Francisco, CA, 94080, USA
| | - Jay S Tung
- Translational Medicine Center, Myelin Repair Foundation, Sunnyvale, CA, 94085, USA
| | - Jason C Dugas
- Translational Medicine Center, Myelin Repair Foundation, Sunnyvale, CA, 94085, USA.,Rigel Pharmaceuticals, South San Francisco, CA, 94080, USA
| | - Tassie L Collins
- Translational Medicine Center, Myelin Repair Foundation, Sunnyvale, CA, 94085, USA.,NGM Biopharmaceuticals, Inc., South San Francisco, CA, 94080, USA
| | - Dmitri Leonoudakis
- Translational Medicine Center, Myelin Repair Foundation, Sunnyvale, CA, 94085, USA. .,Teva Pharmaceuticals, Biologics and CNS Discovery, Redwood City, CA, 94063, USA.
| |
Collapse
|
85
|
Gonzalez GA, Hofer MP, Syed YA, Amaral AI, Rundle J, Rahman S, Zhao C, Kotter MRN. Tamoxifen accelerates the repair of demyelinated lesions in the central nervous system. Sci Rep 2016; 6:31599. [PMID: 27554391 PMCID: PMC4995517 DOI: 10.1038/srep31599] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 06/15/2016] [Indexed: 01/04/2023] Open
Abstract
Enhancing central nervous system (CNS) myelin regeneration is recognized as an important strategy to ameliorate the devastating consequences of demyelinating diseases such as multiple sclerosis. Previous findings have indicated that myelin proteins, which accumulate following demyelination, inhibit remyelination by blocking the differentiation of rat oligodendrocyte progenitor cells (OPCs) via modulation of PKCα. We therefore screened drugs for their potential to overcome this differentiation block. From our screening, tamoxifen emerges as a potent inducer of OPC differentiation in vitro. We show that the effects of tamoxifen rely on modulation of the estrogen receptors ERα, ERβ, and GPR30. Furthermore, we demonstrate that administration of tamoxifen to demyelinated rats in vivo accelerates remyelination. Tamoxifen is a well-established drug and is thus a promising candidate for a drug to regenerate myelin, as it will not require extensive safety testing. In addition, Tamoxifen plays an important role in biomedical research as an activator of inducible genetic models. Our results highlight the importance of appropriate controls when using such models.
Collapse
Affiliation(s)
- Ginez A Gonzalez
- Anne McLaren Laboratory for Regenerative Medicine, Department of Clinical Neurosciences, Wellcome Trust and MRC Cambridge Stem Cell Institute, University of Cambridge, West Forvie Building, Forvie Site, Robinson Way, Cambridge CB2 0SZ, UK
| | - Matthias P Hofer
- Anne McLaren Laboratory for Regenerative Medicine, Department of Clinical Neurosciences, Wellcome Trust and MRC Cambridge Stem Cell Institute, University of Cambridge, West Forvie Building, Forvie Site, Robinson Way, Cambridge CB2 0SZ, UK
| | - Yasir A Syed
- Anne McLaren Laboratory for Regenerative Medicine, Department of Clinical Neurosciences, Wellcome Trust and MRC Cambridge Stem Cell Institute, University of Cambridge, West Forvie Building, Forvie Site, Robinson Way, Cambridge CB2 0SZ, UK
| | - Ana I Amaral
- Anne McLaren Laboratory for Regenerative Medicine, Department of Clinical Neurosciences, Wellcome Trust and MRC Cambridge Stem Cell Institute, University of Cambridge, West Forvie Building, Forvie Site, Robinson Way, Cambridge CB2 0SZ, UK
| | - Jon Rundle
- Anne McLaren Laboratory for Regenerative Medicine, Department of Clinical Neurosciences, Wellcome Trust and MRC Cambridge Stem Cell Institute, University of Cambridge, West Forvie Building, Forvie Site, Robinson Way, Cambridge CB2 0SZ, UK
| | - Saifur Rahman
- Anne McLaren Laboratory for Regenerative Medicine, Department of Clinical Neurosciences, Wellcome Trust and MRC Cambridge Stem Cell Institute, University of Cambridge, West Forvie Building, Forvie Site, Robinson Way, Cambridge CB2 0SZ, UK
| | - Chao Zhao
- Anne McLaren Laboratory for Regenerative Medicine, Department of Clinical Neurosciences, Wellcome Trust and MRC Cambridge Stem Cell Institute, University of Cambridge, West Forvie Building, Forvie Site, Robinson Way, Cambridge CB2 0SZ, UK
| | - Mark R N Kotter
- Anne McLaren Laboratory for Regenerative Medicine, Department of Clinical Neurosciences, Wellcome Trust and MRC Cambridge Stem Cell Institute, University of Cambridge, West Forvie Building, Forvie Site, Robinson Way, Cambridge CB2 0SZ, UK
| |
Collapse
|
86
|
Mendibe Bilbao M, Boyero Durán S, Bárcena Llona J, Rodriguez-Antigüedad A. Multiple sclerosis: Pregnancy and women's health issues. Neurologia 2016; 34:259-269. [PMID: 27546613 DOI: 10.1016/j.nrl.2016.06.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 06/06/2016] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND The course of multiple sclerosis (MS) is influenced by sex, pregnancy and hormonal factors. AIMS To analyse the influence of the above factors in order to clarify the aetiopathogenic mechanisms involved in the disease. METHODS We conducted a comprehensive review of scientific publications in the PubMed database using a keyword search for 'multiple sclerosis', 'MS', 'EAE', 'pregnancy', 'hormonal factors', 'treatment', and related terms. We reviewed the advances presented at the meeting held by the European Committee for Treatment and Research in Multiple Sclerosis (ECTRIMS) in March 2013 in London, as well as recommendations by international experts. RESULTS AND CONCLUSIONS We provide recommendations for counselling and treating women with MS prior to and during pregnancy and after delivery. Current findings on the effects of treatment on the mother, fetus, and newborn are also presented. We issue recommendations for future research in order to address knowledge gaps and clarify any inconsistencies in currently available data.
Collapse
Affiliation(s)
- M Mendibe Bilbao
- Departamento de Neurología, Hospital Universitario Cruces, Baracaldo, España.
| | - S Boyero Durán
- Departamento de Neurología, Hospital Universitario Cruces, Baracaldo, España
| | - J Bárcena Llona
- Departamento de Neurología, Hospital Universitario Cruces, Baracaldo, España
| | | |
Collapse
|
87
|
Pregnancy and multiple sclerosis: from molecular mechanisms to clinical application. Semin Immunopathol 2016; 38:709-718. [PMID: 27501960 DOI: 10.1007/s00281-016-0584-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 07/19/2016] [Indexed: 12/14/2022]
Abstract
Translational research generally refers to a "bench to bedside" approach where basic science discoveries in models move to clinical trials in humans. However, a "bedside to bench to bedside" approach may be more promising with respect to clinical relevance, since it starts with a clinical observation that can serve as a research paradigm to elucidate mechanisms and translate them back into novel therapeutic approaches. The effect of pregnancy on human autoimmune disorders in general, and multiple sclerosis (MS) in particular, serves as an intriguing example of how this can be used to understand disease pathobiology and discover new therapeutic targets. Disease activity in MS undergoes pronounced shifts in the time before, during, and after pregnancy. The most well-known and established example is a reduction in relapse rates in the last trimester by 70-80 %. However, disease activity reappears in the first few months after delivery, temporarily overshooting pre-pregnancy levels. This phenomenon has since its first description served as a model for investigating novel treatment options in animal models and has cumulated in successful phase 2a and 2b trials in female MS patients. However, recently, a number of other clinical observations have been made that might be similarly suitable to offer additional insights into pathobiological mechanisms of MS activity, progression, and possibly even incidence. Here, we outline the various changes in the clinical course of MS that have been described in relation to pregnancy, both short term and long term, and discuss how these may inform the development of novel treatments for autoimmune diseases.
Collapse
|
88
|
Sex effects on inflammatory and neurodegenerative processes in multiple sclerosis. Neurosci Biobehav Rev 2016; 67:137-46. [DOI: 10.1016/j.neubiorev.2015.12.015] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 12/09/2015] [Accepted: 12/21/2015] [Indexed: 01/14/2023]
|
89
|
Zapata LB, Oduyebo T, Whiteman MK, Houtchens MK, Marchbanks PA, Curtis KM. Contraceptive use among women with multiple sclerosis: a systematic review. Contraception 2016; 94:612-620. [PMID: 27452316 DOI: 10.1016/j.contraception.2016.07.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 07/13/2016] [Accepted: 07/18/2016] [Indexed: 11/30/2022]
Abstract
BACKGROUND Contraception is an important consideration for women with multiple sclerosis (MS); however, little is known about the possible effects of hormonal contraception on disease progression or other adverse outcomes (e.g., thrombosis, low bone mineral density). OBJECTIVE To evaluate the evidence on the safety of contraceptive use among women with MS. SEARCH STRATEGY We searched the PubMed database for peer-reviewed articles published in any language from database inception through July 2015. SELECTION CRITERIA We included studies that examined health outcomes among women diagnosed with MS initiating or continuing a contraceptive method. We excluded case reports and case series but included all other study designs. RESULTS From 111 articles, we identified four studies (from 5 articles) that met our inclusion criteria. Evidence from one randomized controlled trial, two retrospective cohort studies, and one cross-sectional study suggests that use of combined oral contraceptives (COCs) or oral contraceptives (OCs) (type not specified) among women with MS does not worsen the clinical course of disease, defined as disability level, disease severity or progression, relapse or number of new brain lesions on magnetic resonance imaging (body of evidence grading Level I, fair to Level II-3, poor). No studies were identified that examined the safety of other contraceptive methods or examined other outcomes of interest (venous thromboembolism, changes in bone mineral density) related to contraceptive use among women with MS. CONCLUSIONS Limited evidence suggests that COC or OC use after MS onset does not worsen the clinical course of disease.
Collapse
Affiliation(s)
- Lauren B Zapata
- Division of Reproductive Health, US Centers for Disease Control and Prevention, Chamblee, Georgia, 30341-3717.
| | - Titilope Oduyebo
- Division of Reproductive Health, US Centers for Disease Control and Prevention, Chamblee, Georgia, 30341-3717
| | - Maura K Whiteman
- Division of Reproductive Health, US Centers for Disease Control and Prevention, Chamblee, Georgia, 30341-3717
| | - Maria K Houtchens
- Department of Neurology, Partners Multiple Sclerosis Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| | - Polly A Marchbanks
- Division of Reproductive Health, US Centers for Disease Control and Prevention, Chamblee, Georgia, 30341-3717
| | - Kathryn M Curtis
- Division of Reproductive Health, US Centers for Disease Control and Prevention, Chamblee, Georgia, 30341-3717
| |
Collapse
|
90
|
Zhu WH, Lu CZ, Huang YM, Link H, Xiao BG. A putative mechanism on remission of multiple sclerosis during pregnancy: estrogen-induced indoleamine 2,3-dioxygenase by dendritic cells. Mult Scler 2016; 13:33-40. [PMID: 17294609 DOI: 10.1177/1352458506071171] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The basis for the reduced relapse rate of multiple sclerosis (MS) during pregnancy remains unexplained but, if defined, could create novel treatment options. Estrogen constitutes one candidate molecule, but the mechanism by which estrogen may affect MS during pregnancy is unclear. In this study, we used monocyte-derived dendritic cells (DCs) from MS patients to explore the estrogen (17-b-estradiol)-related pathway of immune modulation. Estrogen induced the expression of indoleamine 2,3-dioxygenase (IDO) on DCs, limiting T-cell proliferation and both Th1 and Th2 cytokine production. The suppression of T-cell proliferation mediated by estrogenexposed DCs was partly abolished by the IDO-inhibitor, 1-methyl-dl-tryptophan, indicating that estrogen-exposed DCs induced IDO-dependent T-cell suppression. Our data support the hypothesis that the change in the clinical course of MS observed in pregnancy may be related to the estrogen DC-IDO axis, which could represent a novel target for MS therapy.
Collapse
Affiliation(s)
- W H Zhu
- Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | | | | | | | | |
Collapse
|
91
|
van den Broek HHLP, Damoiseaux JGMC, De Baets MH, Hupperts RMM. The influence of sex hormones on cytokines in multiple sclerosis and experimental autoimmune encephalomyelitis: a review. Mult Scler 2016; 11:349-59. [PMID: 15957520 DOI: 10.1191/1352458505ms1174rr] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The female predominance of multiple sclerosis (MS) has suggested that hormonal differences between the sexes must confer some protective effect on males or enhance the susceptibility of females to this disease. There has been evidence that gonadal hormones can modulate the immune response regulated by antigen presenting cells and T cells. These cells control the immune response by the production of interacting pro- and anti-inflammatory cytokines. The first include the acute phase pro-inflammatory cytokines of the innate immune response as well as the T-helper 1 (Th1) cytokines, while the later contain the Th2 cytokines as well as the suppressor cytokines. There is some evidence that MS and experimental autoimmune encephalitis (EAE) are Th1 cell-mediated diseases. For this reason many studies have been done to influence the pro-inflammatory cytokine production of these Th1 cells in favour of an anti-inflammatory immune response as mediated by Th2 cells. However the role of the regulatory T cells in this context is not clearly understood. Here we review the studies concerning the role of sex hormones on the cytokine production in relation to the disease course of MS and EAE and in particular in the light of the recent revival of the regulatory T cells and their suppressive cytokines.
Collapse
Affiliation(s)
- Hans H L P van den Broek
- Department of Neurology, University Hospital Maastricht, P Debyelaan 25, PO Box 5800, 6202 AZ Maastricht, The Netherlands.
| | | | | | | |
Collapse
|
92
|
Pozzilli C, Pugliatti M. An overview of pregnancy-related issues in patients with multiple sclerosis. Eur J Neurol 2016; 22 Suppl 2:34-9. [PMID: 26374512 DOI: 10.1111/ene.12797] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 06/05/2015] [Indexed: 11/28/2022]
Abstract
Although pregnancy in women with multiple sclerosis (MS) is not generally considered high risk, there are some associated therapeutic challenges. The pregnancy-associated reduction in the relapse rate, especially in the third trimester, is followed by a sharp increase in the first few months postpartum. Nevertheless, retrospective evidence for pregnant women with and without MS followed for up to 10 years indicates that pregnancy has no perceptible effect on long-term disease course or disability progression. Likewise, MS has no apparent effects on the pregnancy course or fetal outcomes. All disease-modifying therapies (DMTs) have potential adverse effects on fertility and pregnancy outcomes, but the level of risk varies amongst agents. There is some support for continued use of interferon-β and glatiramer acetate throughout pregnancy to reduce the risk of relapse. Use of DMTs during breastfeeding is best avoided if possible. Close evaluation of drug safety information is imperative when managing women with MS who are pregnant or wish to become pregnant. Decision-making should be a shared experience between patient and physician, and the approach must be individualized for each patient.
Collapse
Affiliation(s)
- C Pozzilli
- Department of Neurology and Psychiatry, Sapienza University, Rome, Italy
| | - M Pugliatti
- Department of Biomedical and Specialty Surgical Sciences, University of Ferrara, Italy
| | | |
Collapse
|
93
|
Khalaj AJ, Hasselmann J, Augello C, Moore S, Tiwari-Woodruff SK. Nudging oligodendrocyte intrinsic signaling to remyelinate and repair: Estrogen receptor ligand effects. J Steroid Biochem Mol Biol 2016; 160:43-52. [PMID: 26776441 PMCID: PMC5233753 DOI: 10.1016/j.jsbmb.2016.01.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 01/08/2016] [Accepted: 01/11/2016] [Indexed: 01/06/2023]
Abstract
Demyelination in multiple sclerosis (MS) leads to significant, progressive axonal and neuronal degeneration. Currently existing immunosuppressive and immunomodulatory therapies alleviate MS symptoms and slow, but fail to prevent or reverse, disease progression. Restoration of damaged myelin sheath by replenishment of mature oligodendrocytes (OLs) should not only restore saltatory axon conduction, but also provide a major boost to axon survival. Our previous work has shown that therapeutic treatment with the modestly selective generic estrogen receptor (ER) β agonist diarylpropionitrile (DPN) confers functional neuroprotection in a chronic experimental autoimmune encephalomyelitis (EAE) mouse model of MS by stimulating endogenous remyelination. Recently, we found that the more potent, selective ERβ agonist indazole-chloride (Ind-Cl) improves clinical disease and motor performance. Importantly, electrophysiological measures revealed improved corpus callosal conduction and reduced axon refractoriness. This Ind-Cl treatment-induced functional remyelination was attributable to increased OL progenitor cell (OPC) and mature OL numbers. At the intracellular signaling level, transition of early to late OPCs requires ERK1/2 signaling, and transition of immature to mature OLs requires mTOR signaling; thus, the PI3K/Akt/mTOR pathway plays a major role in the late stages of OL differentiation and myelination. Indeed, therapeutic treatment of EAE mice with various ERβ agonists results in increased brain-derived neurotrophic factor (BDNF) and phosphorylated (p) Akt and p-mTOR levels. It is notable that while DPN's neuroprotective effects occur in the presence of peripheral and central inflammation, Ind-Cl is directly neuroprotective, as demonstrated by remyelination effects in the cuprizone-induced demyelination model, as well as immunomodulatory. Elucidating the mechanisms by which ER agonists and other directly remyelinating agents modulate endogenous OPC and OL regulatory signaling is critical to the development of effective remyelinating drugs. The discovery of signaling targets to induce functional remyelination will valuably contribute to the treatment of demyelinating neurological diseases, including MS, stroke, and traumatic brain and spinal cord injury.
Collapse
Affiliation(s)
- Anna J Khalaj
- Division of Biomedical Sciences, School of Medicine at the University of California, Riverside, United States
| | - Jonathan Hasselmann
- Division of Biomedical Sciences, School of Medicine at the University of California, Riverside, United States
| | - Catherine Augello
- Division of Biomedical Sciences, School of Medicine at the University of California, Riverside, United States
| | - Spencer Moore
- Division of Biomedical Sciences, School of Medicine at the University of California, Riverside, United States
| | - Seema K Tiwari-Woodruff
- Division of Biomedical Sciences, School of Medicine at the University of California, Riverside, United States; Neuroscience Graduate Program, University of California, Riverside, United States.
| |
Collapse
|
94
|
Siddiqui AN, Siddiqui N, Khan RA, Kalam A, Jabir NR, Kamal MA, Firoz CK, Tabrez S. Neuroprotective Role of Steroidal Sex Hormones: An Overview. CNS Neurosci Ther 2016; 22:342-50. [PMID: 27012165 PMCID: PMC6492877 DOI: 10.1111/cns.12538] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Revised: 02/21/2016] [Accepted: 02/21/2016] [Indexed: 12/11/2022] Open
Abstract
Progesterone, estrogens, and testosterone are the well-known steroidal sex hormones, which have been reported to have "nonreproductive "effects in the brain, specifically in the neuroprotection and neurotrophy. In the last one decade, there has been a surge in the research on the role of these hormones in neuroprotection and their positive impact on different brain injuries. The said interest has been sparked by a desire to understand the action and mechanisms of these steroidal sex hormones throughout the body. The aim of this article was to highlight the potential outcome of the steroidal hormones, viz. progesterone, estrogens, and testosterone in terms of their role in neuroprotection and other brain injuries. Their possible mechanism of action at both genomic and nongenomic level will be also discussed. As far as our knowledge goes, we are for the first time reporting neuroprotective effect and possible mechanism of action of these hormones in a single article.
Collapse
Affiliation(s)
- Ali Nasir Siddiqui
- Department of Pharmaceutical Medicine, Faculty of Pharmacy, Jamia Hamdard, New Delhi, India
| | - Nahida Siddiqui
- Department of Pharmacognosy and Phytochemistry, Faculty of Pharmacy, Jamia Hamdard, New Delhi, India
| | - Rashid Ali Khan
- Department of Pharmaceutical Medicine, Faculty of Pharmacy, Jamia Hamdard, New Delhi, India
| | - Abul Kalam
- Department of Pharmacology, Faculty of Pharmacy, Jamia Hamdard, New Delhi, India
| | - Nasimudeen R Jabir
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammad Amjad Kamal
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Enzymoics, 7 Peterlee Place, Hebersham, NSW, Australia
| | | | - Shams Tabrez
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
95
|
Coyle PK. Management of women with multiple sclerosis through pregnancy and after childbirth. Ther Adv Neurol Disord 2016; 9:198-210. [PMID: 27134675 DOI: 10.1177/1756285616631897] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Multiple sclerosis (MS) is a major acquired neurologic disease of young adults. The prototypic patient is a young woman of reproductive age. Gender preference is becoming more pronounced, since MS is increasing specifically among women. Any healthcare provider who deals with MS must be prepared to discuss pregnancy issues, and provide appropriate counseling. This is now complicated by the availability of multiple treatment options. There is growing literature on which to base recommendations, particularly regarding washout periods. After a brief background introduction, this review will discuss state-of-the-art family planning counseling in the treatment era, divided into prepregnancy, pregnancy, and postpartum MS issues.
Collapse
Affiliation(s)
- Patricia K Coyle
- Dept. of Neurology HSC, T12 Stony Brook University Medical Center Stony Brook, NY 11794, USA
| |
Collapse
|
96
|
D'Amico E, Leone C, Patti F. Offspring Number Does Not Influence Reaching the Disability's Milestones in Multiple Sclerosis: A Seven-Year Follow-Up Study. Int J Mol Sci 2016; 17:234. [PMID: 26907250 PMCID: PMC4783965 DOI: 10.3390/ijms17020234] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 01/20/2016] [Accepted: 01/21/2016] [Indexed: 01/09/2023] Open
Abstract
OBJECTIVES Data on pregnancy long-term effects on multiple sclerosis (MS) course are still controversial; whether experiencing more than one pregnancy exposes one to risk of the disability's accrual is still unknown. We investigated differences existing in terms of disability progression among women with MS (wwMS) who had one or more children after their MS onset. METHODS Monoparous and multiparous wwMS were enrolled from the Catania MS Center, Italy, in a monocenter retrospective study. A Cox proportional hazards model was used to examine the effect of the number of parities on time from MS disease onset to EDSS 4.0 and 6.0. The study protocol was approved by the local Ethical Committee. RESULTS During the seven years of observation, 32.1% and 23.2% of the monoparous group reached expanded disability disease status (EDSS) 4.0 and 6.0 respectively, compared to 13.3% and 3.3% of the multiparous group (p = 0.057 and p = 0.017; respectively). The Kaplan-Meier curve analysis showed no statistically-significant differences between the two groups in reaching the two milestones. The multiparous group showed a longer time to reach the EDSS 4.0 (3.5 vs. 2.6 years, log-rank 0.57, p = 0.45). The Cox regression analysis showed that the EDSS at the time of first pregnancy (Exp(B) 9.4, CI 4.5-19.7, p< 0.001) and the time from MS onset to first pregnancy (Exp(B) 0.96, CI = 0.93-0.98, p < 0.05) were significant predictors of reaching the EDSS 4.0, whereas a model including only the EDSS one year after the first pregnancy significantly predicted (Exp(B) value of 6.4, CI 2.6-15.4, p < 0.001) the reaching of EDSS 6.0. CONCLUSIONS Our results suggest that experiencing more than one pregnancy could not convey a different clinical outcome in wwMS. Further research is needed to confirm our results.
Collapse
Affiliation(s)
- Emanuele D'Amico
- Department of Neurology, University of Catania, Multiple Sclerosis Center, Policlinico G. Rodolico, Via Santa Sofia, 78 Catania 95123, Italy.
| | - Carmela Leone
- Department of Neurology, University of Catania, Multiple Sclerosis Center, Policlinico G. Rodolico, Via Santa Sofia, 78 Catania 95123, Italy.
| | - Francesco Patti
- Department of Neurology, University of Catania, Multiple Sclerosis Center, Policlinico G. Rodolico, Via Santa Sofia, 78 Catania 95123, Italy.
| |
Collapse
|
97
|
Gourdy P, Bourgeois EA, Levescot A, Pham L, Riant E, Ahui ML, Damotte D, Gombert JM, Bayard F, Ohlsson C, Arnal JF, Herbelin A. Estrogen Therapy Delays Autoimmune Diabetes and Promotes the Protective Efficiency of Natural Killer T-Cell Activation in Female Nonobese Diabetic Mice. Endocrinology 2016; 157:258-67. [PMID: 26485613 DOI: 10.1210/en.2015-1313] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Therapeutic strategies focused on restoring immune tolerance remain the main avenue to prevent type 1 diabetes (T1D). Because estrogens potentiate FoxP3+ regulatory T cells (Treg) and invariant natural killer T (iNKT) cells, two regulatory lymphocyte populations that are functionally deficient in nonobese diabetic (NOD) mice, we investigated whether estradiol (E2) therapy influences the course of T1D in this model. To this end, female NOD mice were sc implanted with E2- or placebo-delivering pellets to explore the course of spontaneous and cyclophosphamide-induced diabetes. Treg-depleted and iNKT-cell-deficient (Jα18(-/-)) NOD mice were used to assess the respective involvement of these lymphocyte populations in E2 effects. Early E2 administration (from 4 wk of age) was found to preserve NOD mice from both spontaneous and cyclophosphamide-induced diabetes, and a complete protection was also observed throughout treatment when E2 treatment was initiated after the onset of insulitis (from 12 wk of age). This delayed E2 treatment remained fully effective in Treg-depleted mice but failed to entirely protect Jα18(-/-) mice. Accordingly, E2 administration was shown to restore the cytokine production of iNKT cells in response to in vivo challenge with the cognate ligand α-galactosylceramide. Finally, transient E2 administration potentiated the previously described protective action of α-galactosylceramide treatment in NOD females. This study provides original evidence that E2 therapy strongly protects NOD mice from T1D and reveals the estrogen/iNKT cell axis as a new effective target to counteract diabetes onset at the stage of insulitis. Estrogen-based therapy should thus be considered for T1D prevention.
Collapse
MESH Headings
- Animals
- Autoimmune Diseases/drug therapy
- Autoimmune Diseases/immunology
- Autoimmune Diseases/metabolism
- Autoimmune Diseases/prevention & control
- Cytokines/blood
- Cytokines/metabolism
- Diabetes Mellitus, Type 1/drug therapy
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/prevention & control
- Drug Implants
- Estradiol/administration & dosage
- Estradiol/therapeutic use
- Estrogen Replacement Therapy
- Estrogens/administration & dosage
- Estrogens/therapeutic use
- Female
- Galactosylceramides/agonists
- Galactosylceramides/pharmacology
- Galactosylceramides/therapeutic use
- Immune Tolerance/drug effects
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Lymphocyte Activation/drug effects
- Lymphocyte Depletion/adverse effects
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Mice, Mutant Strains
- Ovariectomy/adverse effects
- Prediabetic State/drug therapy
- Prediabetic State/immunology
- Prediabetic State/metabolism
- Prediabetic State/prevention & control
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
Collapse
Affiliation(s)
- Pierre Gourdy
- INSERM Unité 1048 (P.G., E.R., F.B., J.-F.A.), Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; Toulouse University (P.G., J.-F.A.), 31059 Toulouse, France; Department of Diabetology (P.G.), Toulouse University Hospital, 31403 Toulouse, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 8147 (E.A.B., L.P., M.-L.A.), Necker Hospital, 75015 Paris, France; Paris Descartes University, Necker Hospital (E.A.B., L.P., M.-L.A., A.H.), 75014 Paris, France; INSERM Unité 1082 (A.L., A.H.), 86022 Poitiers, France; Paris-Sud-11 University (A.L.), 91405 Orsay, France; Department of Anatomy and Cytology (A.L., D.D.), Hôtel Dieu, 49033 Paris, France; Laboratory of Immunology (J.-M.G.), Poitiers, and Poitiers University (J.-M.G., A.H.), 86000 Poitiers, France; Centre Hospitalo-Universitaire de Poitiers (J.-M.G., A.H.), 86021 Poitiers, France; and Centre for Bone and Arthritis Research (C.O.), University of Gothenburg, S-405 30 Gothenburg, Sweden
| | - Elvire A Bourgeois
- INSERM Unité 1048 (P.G., E.R., F.B., J.-F.A.), Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; Toulouse University (P.G., J.-F.A.), 31059 Toulouse, France; Department of Diabetology (P.G.), Toulouse University Hospital, 31403 Toulouse, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 8147 (E.A.B., L.P., M.-L.A.), Necker Hospital, 75015 Paris, France; Paris Descartes University, Necker Hospital (E.A.B., L.P., M.-L.A., A.H.), 75014 Paris, France; INSERM Unité 1082 (A.L., A.H.), 86022 Poitiers, France; Paris-Sud-11 University (A.L.), 91405 Orsay, France; Department of Anatomy and Cytology (A.L., D.D.), Hôtel Dieu, 49033 Paris, France; Laboratory of Immunology (J.-M.G.), Poitiers, and Poitiers University (J.-M.G., A.H.), 86000 Poitiers, France; Centre Hospitalo-Universitaire de Poitiers (J.-M.G., A.H.), 86021 Poitiers, France; and Centre for Bone and Arthritis Research (C.O.), University of Gothenburg, S-405 30 Gothenburg, Sweden
| | - Anaïs Levescot
- INSERM Unité 1048 (P.G., E.R., F.B., J.-F.A.), Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; Toulouse University (P.G., J.-F.A.), 31059 Toulouse, France; Department of Diabetology (P.G.), Toulouse University Hospital, 31403 Toulouse, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 8147 (E.A.B., L.P., M.-L.A.), Necker Hospital, 75015 Paris, France; Paris Descartes University, Necker Hospital (E.A.B., L.P., M.-L.A., A.H.), 75014 Paris, France; INSERM Unité 1082 (A.L., A.H.), 86022 Poitiers, France; Paris-Sud-11 University (A.L.), 91405 Orsay, France; Department of Anatomy and Cytology (A.L., D.D.), Hôtel Dieu, 49033 Paris, France; Laboratory of Immunology (J.-M.G.), Poitiers, and Poitiers University (J.-M.G., A.H.), 86000 Poitiers, France; Centre Hospitalo-Universitaire de Poitiers (J.-M.G., A.H.), 86021 Poitiers, France; and Centre for Bone and Arthritis Research (C.O.), University of Gothenburg, S-405 30 Gothenburg, Sweden
| | - Linh Pham
- INSERM Unité 1048 (P.G., E.R., F.B., J.-F.A.), Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; Toulouse University (P.G., J.-F.A.), 31059 Toulouse, France; Department of Diabetology (P.G.), Toulouse University Hospital, 31403 Toulouse, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 8147 (E.A.B., L.P., M.-L.A.), Necker Hospital, 75015 Paris, France; Paris Descartes University, Necker Hospital (E.A.B., L.P., M.-L.A., A.H.), 75014 Paris, France; INSERM Unité 1082 (A.L., A.H.), 86022 Poitiers, France; Paris-Sud-11 University (A.L.), 91405 Orsay, France; Department of Anatomy and Cytology (A.L., D.D.), Hôtel Dieu, 49033 Paris, France; Laboratory of Immunology (J.-M.G.), Poitiers, and Poitiers University (J.-M.G., A.H.), 86000 Poitiers, France; Centre Hospitalo-Universitaire de Poitiers (J.-M.G., A.H.), 86021 Poitiers, France; and Centre for Bone and Arthritis Research (C.O.), University of Gothenburg, S-405 30 Gothenburg, Sweden
| | - Elodie Riant
- INSERM Unité 1048 (P.G., E.R., F.B., J.-F.A.), Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; Toulouse University (P.G., J.-F.A.), 31059 Toulouse, France; Department of Diabetology (P.G.), Toulouse University Hospital, 31403 Toulouse, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 8147 (E.A.B., L.P., M.-L.A.), Necker Hospital, 75015 Paris, France; Paris Descartes University, Necker Hospital (E.A.B., L.P., M.-L.A., A.H.), 75014 Paris, France; INSERM Unité 1082 (A.L., A.H.), 86022 Poitiers, France; Paris-Sud-11 University (A.L.), 91405 Orsay, France; Department of Anatomy and Cytology (A.L., D.D.), Hôtel Dieu, 49033 Paris, France; Laboratory of Immunology (J.-M.G.), Poitiers, and Poitiers University (J.-M.G., A.H.), 86000 Poitiers, France; Centre Hospitalo-Universitaire de Poitiers (J.-M.G., A.H.), 86021 Poitiers, France; and Centre for Bone and Arthritis Research (C.O.), University of Gothenburg, S-405 30 Gothenburg, Sweden
| | - Marie-Louise Ahui
- INSERM Unité 1048 (P.G., E.R., F.B., J.-F.A.), Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; Toulouse University (P.G., J.-F.A.), 31059 Toulouse, France; Department of Diabetology (P.G.), Toulouse University Hospital, 31403 Toulouse, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 8147 (E.A.B., L.P., M.-L.A.), Necker Hospital, 75015 Paris, France; Paris Descartes University, Necker Hospital (E.A.B., L.P., M.-L.A., A.H.), 75014 Paris, France; INSERM Unité 1082 (A.L., A.H.), 86022 Poitiers, France; Paris-Sud-11 University (A.L.), 91405 Orsay, France; Department of Anatomy and Cytology (A.L., D.D.), Hôtel Dieu, 49033 Paris, France; Laboratory of Immunology (J.-M.G.), Poitiers, and Poitiers University (J.-M.G., A.H.), 86000 Poitiers, France; Centre Hospitalo-Universitaire de Poitiers (J.-M.G., A.H.), 86021 Poitiers, France; and Centre for Bone and Arthritis Research (C.O.), University of Gothenburg, S-405 30 Gothenburg, Sweden
| | - Diane Damotte
- INSERM Unité 1048 (P.G., E.R., F.B., J.-F.A.), Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; Toulouse University (P.G., J.-F.A.), 31059 Toulouse, France; Department of Diabetology (P.G.), Toulouse University Hospital, 31403 Toulouse, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 8147 (E.A.B., L.P., M.-L.A.), Necker Hospital, 75015 Paris, France; Paris Descartes University, Necker Hospital (E.A.B., L.P., M.-L.A., A.H.), 75014 Paris, France; INSERM Unité 1082 (A.L., A.H.), 86022 Poitiers, France; Paris-Sud-11 University (A.L.), 91405 Orsay, France; Department of Anatomy and Cytology (A.L., D.D.), Hôtel Dieu, 49033 Paris, France; Laboratory of Immunology (J.-M.G.), Poitiers, and Poitiers University (J.-M.G., A.H.), 86000 Poitiers, France; Centre Hospitalo-Universitaire de Poitiers (J.-M.G., A.H.), 86021 Poitiers, France; and Centre for Bone and Arthritis Research (C.O.), University of Gothenburg, S-405 30 Gothenburg, Sweden
| | - Jean-Marc Gombert
- INSERM Unité 1048 (P.G., E.R., F.B., J.-F.A.), Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; Toulouse University (P.G., J.-F.A.), 31059 Toulouse, France; Department of Diabetology (P.G.), Toulouse University Hospital, 31403 Toulouse, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 8147 (E.A.B., L.P., M.-L.A.), Necker Hospital, 75015 Paris, France; Paris Descartes University, Necker Hospital (E.A.B., L.P., M.-L.A., A.H.), 75014 Paris, France; INSERM Unité 1082 (A.L., A.H.), 86022 Poitiers, France; Paris-Sud-11 University (A.L.), 91405 Orsay, France; Department of Anatomy and Cytology (A.L., D.D.), Hôtel Dieu, 49033 Paris, France; Laboratory of Immunology (J.-M.G.), Poitiers, and Poitiers University (J.-M.G., A.H.), 86000 Poitiers, France; Centre Hospitalo-Universitaire de Poitiers (J.-M.G., A.H.), 86021 Poitiers, France; and Centre for Bone and Arthritis Research (C.O.), University of Gothenburg, S-405 30 Gothenburg, Sweden
| | - Francis Bayard
- INSERM Unité 1048 (P.G., E.R., F.B., J.-F.A.), Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; Toulouse University (P.G., J.-F.A.), 31059 Toulouse, France; Department of Diabetology (P.G.), Toulouse University Hospital, 31403 Toulouse, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 8147 (E.A.B., L.P., M.-L.A.), Necker Hospital, 75015 Paris, France; Paris Descartes University, Necker Hospital (E.A.B., L.P., M.-L.A., A.H.), 75014 Paris, France; INSERM Unité 1082 (A.L., A.H.), 86022 Poitiers, France; Paris-Sud-11 University (A.L.), 91405 Orsay, France; Department of Anatomy and Cytology (A.L., D.D.), Hôtel Dieu, 49033 Paris, France; Laboratory of Immunology (J.-M.G.), Poitiers, and Poitiers University (J.-M.G., A.H.), 86000 Poitiers, France; Centre Hospitalo-Universitaire de Poitiers (J.-M.G., A.H.), 86021 Poitiers, France; and Centre for Bone and Arthritis Research (C.O.), University of Gothenburg, S-405 30 Gothenburg, Sweden
| | - Claes Ohlsson
- INSERM Unité 1048 (P.G., E.R., F.B., J.-F.A.), Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; Toulouse University (P.G., J.-F.A.), 31059 Toulouse, France; Department of Diabetology (P.G.), Toulouse University Hospital, 31403 Toulouse, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 8147 (E.A.B., L.P., M.-L.A.), Necker Hospital, 75015 Paris, France; Paris Descartes University, Necker Hospital (E.A.B., L.P., M.-L.A., A.H.), 75014 Paris, France; INSERM Unité 1082 (A.L., A.H.), 86022 Poitiers, France; Paris-Sud-11 University (A.L.), 91405 Orsay, France; Department of Anatomy and Cytology (A.L., D.D.), Hôtel Dieu, 49033 Paris, France; Laboratory of Immunology (J.-M.G.), Poitiers, and Poitiers University (J.-M.G., A.H.), 86000 Poitiers, France; Centre Hospitalo-Universitaire de Poitiers (J.-M.G., A.H.), 86021 Poitiers, France; and Centre for Bone and Arthritis Research (C.O.), University of Gothenburg, S-405 30 Gothenburg, Sweden
| | - Jean-François Arnal
- INSERM Unité 1048 (P.G., E.R., F.B., J.-F.A.), Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; Toulouse University (P.G., J.-F.A.), 31059 Toulouse, France; Department of Diabetology (P.G.), Toulouse University Hospital, 31403 Toulouse, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 8147 (E.A.B., L.P., M.-L.A.), Necker Hospital, 75015 Paris, France; Paris Descartes University, Necker Hospital (E.A.B., L.P., M.-L.A., A.H.), 75014 Paris, France; INSERM Unité 1082 (A.L., A.H.), 86022 Poitiers, France; Paris-Sud-11 University (A.L.), 91405 Orsay, France; Department of Anatomy and Cytology (A.L., D.D.), Hôtel Dieu, 49033 Paris, France; Laboratory of Immunology (J.-M.G.), Poitiers, and Poitiers University (J.-M.G., A.H.), 86000 Poitiers, France; Centre Hospitalo-Universitaire de Poitiers (J.-M.G., A.H.), 86021 Poitiers, France; and Centre for Bone and Arthritis Research (C.O.), University of Gothenburg, S-405 30 Gothenburg, Sweden
| | - André Herbelin
- INSERM Unité 1048 (P.G., E.R., F.B., J.-F.A.), Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; Toulouse University (P.G., J.-F.A.), 31059 Toulouse, France; Department of Diabetology (P.G.), Toulouse University Hospital, 31403 Toulouse, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 8147 (E.A.B., L.P., M.-L.A.), Necker Hospital, 75015 Paris, France; Paris Descartes University, Necker Hospital (E.A.B., L.P., M.-L.A., A.H.), 75014 Paris, France; INSERM Unité 1082 (A.L., A.H.), 86022 Poitiers, France; Paris-Sud-11 University (A.L.), 91405 Orsay, France; Department of Anatomy and Cytology (A.L., D.D.), Hôtel Dieu, 49033 Paris, France; Laboratory of Immunology (J.-M.G.), Poitiers, and Poitiers University (J.-M.G., A.H.), 86000 Poitiers, France; Centre Hospitalo-Universitaire de Poitiers (J.-M.G., A.H.), 86021 Poitiers, France; and Centre for Bone and Arthritis Research (C.O.), University of Gothenburg, S-405 30 Gothenburg, Sweden
| |
Collapse
|
98
|
Voskuhl RR, Wang H, Wu TCJ, Sicotte NL, Nakamura K, Kurth F, Itoh N, Bardens J, Bernard JT, Corboy JR, Cross AH, Dhib-Jalbut S, Ford CC, Frohman EM, Giesser B, Jacobs D, Kasper LH, Lynch S, Parry G, Racke MK, Reder AT, Rose J, Wingerchuk DM, MacKenzie-Graham AJ, Arnold DL, Tseng CH, Elashoff R. Estriol combined with glatiramer acetate for women with relapsing-remitting multiple sclerosis: a randomised, placebo-controlled, phase 2 trial. Lancet Neurol 2015; 15:35-46. [PMID: 26621682 DOI: 10.1016/s1474-4422(15)00322-1] [Citation(s) in RCA: 136] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 10/09/2015] [Accepted: 10/28/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND Relapses of multiple sclerosis decrease during pregnancy, when the hormone estriol is increased. Estriol treatment is anti-inflammatory and neuroprotective in preclinical studies. In a small single-arm study of people with multiple sclerosis estriol reduced gadolinium-enhancing lesions and was favourably immunomodulatory. We assessed whether estriol treatment reduces multiple sclerosis relapses in women. METHODS We did a randomised, double-blind, placebo-controlled phase 2 trial at 16 academic neurology centres in the USA, between June 28, 2007, and Jan 9, 2014. Women aged 18-50 years with relapsing-remitting multiple sclerosis were randomly assigned (1:1) with a random permuted block design to either daily oral estriol (8 mg) or placebo, each in combination with injectable glatiramer acetate 20 mg daily. Patients and all study personnel, except for pharmacists and statisticians, were masked to treatment assignment. The primary endpoint was annualised relapse rate after 24 months, with a significance level of p=0.10. Relapses were confirmed by an increase in Expanded Disability Status Scale score assessed by an independent physician. Analysis was by intention to treat. The trial is registered with ClinicalTrials.gov, number NCT00451204. FINDINGS We enrolled 164 patients: 83 were allocated to the estriol group and 81 were allocated to the placebo group. The annualised confirmed relapse rate was 0.25 relapses per year (95% CI 0.17-0.37) in the estriol group versus 0.37 relapses per year (0.25-0.53) in the placebo group (adjusted rate ratio 0.63, 95% CI 0.37-1.05; p=0.077). The proportion of patients with serious adverse events did not differ substantially between the estriol group and the placebo group (eight [10%] of 82 patients vs ten [13%] of 76 patients). Irregular menses were more common in the estriol group than in the placebo group (19 [23%] vs three [4%], p=0.0005), but vaginal infections were less common (one [1%] vs eight [11%], p=0.0117). There were no differences in breast fibrocystic disease, uterine fibroids, or endometrial lining thickness as assessed by clinical examination, mammogram, uterine ultrasound, or endometrial lining biopsy. INTERPRETATION Estriol plus glatiramer acetate met our criteria for reducing relapse rates, and treatment was well tolerated over 24 months. These results warrant further investigation in a phase 3 trial. FUNDING National Institutes of Health, National Multiple Sclerosis Society, Conrad N Hilton Foundation, Jack H Skirball Foundation, Sherak Family Foundation, and the California Community Foundation.
Collapse
Affiliation(s)
- Rhonda R Voskuhl
- David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA, USA.
| | - HeJing Wang
- David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA, USA
| | - T C Jackson Wu
- David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA, USA
| | | | | | - Florian Kurth
- David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA, USA
| | - Noriko Itoh
- David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA, USA
| | - Jenny Bardens
- David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA, USA
| | | | | | - Anne H Cross
- Washington University School of Medicine, St Louis, MO, USA
| | | | - Corey C Ford
- University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | | | - Barbara Giesser
- David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA, USA
| | - Dina Jacobs
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Lloyd H Kasper
- Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | - Sharon Lynch
- University of Kansas Medical Center, Kansas City, KS, USA
| | | | - Michael K Racke
- Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | | | - John Rose
- Salt Lake City VA Medical Center, Salt Lake City, UT, USA
| | | | - Allan J MacKenzie-Graham
- David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA, USA
| | | | - Chi Hong Tseng
- David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA, USA
| | - Robert Elashoff
- David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
99
|
Langer-Gould A. Sex hormones and multiple sclerosis: another informative failure. Lancet Neurol 2015; 15:22-3. [PMID: 26621681 DOI: 10.1016/s1474-4422(15)00348-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 11/18/2015] [Indexed: 10/22/2022]
Affiliation(s)
- Annette Langer-Gould
- Kaiser Permanente, Research & Evaluation, 100 S Los Robles Avenue, Pasadena, California 91101, USA.
| |
Collapse
|
100
|
Sternberg Z. Genetic, Epigenetic, and Environmental Factors Influencing Neurovisceral Integration of Cardiovascular Modulation: Focus on Multiple Sclerosis. Neuromolecular Med 2015; 18:16-36. [PMID: 26502224 DOI: 10.1007/s12017-015-8375-5] [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] [Received: 04/01/2015] [Accepted: 10/19/2015] [Indexed: 12/31/2022]
Abstract
Thought to be an autoimmune inflammatory CNS disease, multiple sclerosis (MS) involves multiple pathologies with heterogeneous clinical presentations. An impaired neurovisceral integration of cardiovascular modulation, indicated by sympathetic and parasympathetic autonomic nervous system (ANS) dysfunction, is among common MS clinical presentations. ANS dysfunction could not only enhance MS inflammatory and neurodegenerative processes, but can also lead to clinical symptoms such as depression, fatigue, sleep disorder, migraine, osteoporosis, and cerebral hemodynamic impairments. Therefore, factors influencing ANS functional activities, in one way or another, will have a significant impact on MS disease course. This review describes the genetic and epigenetic factors, and their interactions with a number of environmental factors contributing to the neurovisceral integration of cardiovascular modulation, with a focus on MS. Future studies should investigate the improvement in cardiovascular ANS function, as a strategy for preventing and minimizing MS-related morbidities, and improving patients' quality of life.
Collapse
|