1
|
Floreani A, Gabbia D, De Martin S. Are Gender Differences Important for Autoimmune Liver Diseases? Life (Basel) 2024; 14:500. [PMID: 38672770 PMCID: PMC11050899 DOI: 10.3390/life14040500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/04/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
Gender Medicine has had an enormous expansion over the last ten years. Autoimmune liver diseases include several conditions, i.e., autoimmune hepatitis (AIH), primary biliary cholangitis (PBC), primary sclerosing cholangitis (PSC), and conditions involving the liver or biliary tree overlapping with AIH, as well as IgG4-related disease. However, little is known about the impact of sex in the pathogenesis and natural history of these conditions. The purpose of this review is to provide an update of the gender disparities among the autoimmune liver diseases by reviewing the data published from 1999 to 2023. The epidemiology of these diseases has been changing over the last years, due to the amelioration of knowledge in their diagnosis, pathogenesis, and treatment. The clinical data collected so far support the existence of sex differences in the natural history of autoimmune liver diseases. Notably, their history could be longer than that which is now known, with problems being initiated even at a pediatric age. Moreover, gender disparity has been observed during the onset of complications related to end-stage liver disease, including cancer incidence. However, there is still an important debate among researchers about the impact of sex and the pathogenesis of these conditions. With this review, we would like to emphasize the urgency of basic science and clinical research to increase our understanding of the sex differences in autoimmune liver diseases.
Collapse
Affiliation(s)
- Annarosa Floreani
- Scientific Consultant IRCCS Negrar, 37024 Verona, Italy
- University of Padova, 35122 Padova, Italy
| | - Daniela Gabbia
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35122 Padova, Italy; (D.G.); (S.D.M.)
| | - Sara De Martin
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35122 Padova, Italy; (D.G.); (S.D.M.)
| |
Collapse
|
2
|
Dunn SE, Perry WA, Klein SL. Mechanisms and consequences of sex differences in immune responses. Nat Rev Nephrol 2024; 20:37-55. [PMID: 37993681 DOI: 10.1038/s41581-023-00787-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/26/2023] [Indexed: 11/24/2023]
Abstract
Biological sex differences refer to differences between males and females caused by the sex chromosome complement (that is, XY or XX), reproductive tissues (that is, the presence of testes or ovaries), and concentrations of sex steroids (that is, testosterone or oestrogens and progesterone). Although these sex differences are binary for most human individuals and mice, transgender individuals receiving hormone therapy, individuals with genetic syndromes (for example, Klinefelter and Turner syndromes) and people with disorders of sexual development reflect the diversity in sex-based biology. The broad distribution of sex steroid hormone receptors across diverse cell types and the differential expression of X-linked and autosomal genes means that sex is a biological variable that can affect the function of all physiological systems, including the immune system. Sex differences in immune cell function and immune responses to foreign and self antigens affect the development and outcome of diverse diseases and immune responses.
Collapse
Affiliation(s)
- Shannon E Dunn
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
- Women's College Research Institute, Women's College Hospital, Toronto, Ontario, Canada
| | - Whitney A Perry
- Division of Geographic Medicine and Infectious Diseases, Tufts Medical Center, Boston, MA, USA
| | - Sabra L Klein
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| |
Collapse
|
3
|
Hoffmann JP, Liu JA, Seddu K, Klein SL. Sex hormone signaling and regulation of immune function. Immunity 2023; 56:2472-2491. [PMID: 37967530 DOI: 10.1016/j.immuni.2023.10.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/25/2023] [Accepted: 10/14/2023] [Indexed: 11/17/2023]
Abstract
Immune responses to antigens, including innocuous, self, tumor, microbial, and vaccine antigens, differ between males and females. The quest to uncover the mechanisms for biological sex differences in the immune system has intensified, with considerable literature pointing toward sex hormonal influences on immune cell function. Sex steroids, including estrogens, androgens, and progestins, have profound effects on immune function. As such, drastic changes in sex steroid concentrations that occur with aging (e.g., after puberty or during the menopause transition) or pregnancy impact immune responses and the pathogenesis of immune-related diseases. The effect of sex steroids on immunity involves both the concentration of the ligand and the density and distribution of genomic and nongenomic receptors that serve as transcriptional regulators of immune cellular responses to affect autoimmunity, allergy, infectious diseases, cancers, and responses to vaccines. The next frontier will be harnessing these effects of sex steroids to improve therapeutic outcomes.
Collapse
Affiliation(s)
- Joseph P Hoffmann
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Jennifer A Liu
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Kumba Seddu
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Sabra L Klein
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA; Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA; Department of Medicine, Division of Infectious Diseases, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
4
|
Dodd KC, Menon M. Sex bias in lymphocytes: Implications for autoimmune diseases. Front Immunol 2022; 13:945762. [PMID: 36505451 PMCID: PMC9730535 DOI: 10.3389/fimmu.2022.945762] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 11/03/2022] [Indexed: 11/25/2022] Open
Abstract
Autoimmune diseases are characterized by a significant sex dimorphism, with women showing increased susceptibility to disease. This is, at least in part, due to sex-dependent differences in the immune system that are influenced by the complex interplay between sex hormones and sex chromosomes, with contribution from sociological factors, diet and gut microbiota. Sex differences are evident in the number and function of lymphocyte populations. Women mount a stronger pro-inflammatory response than males, with increased lymphocyte proliferation, activation and pro-inflammatory cytokine production, whereas men display expanded regulatory cell subsets. Ageing alters the immune landscape of men and women in differing ways, resulting in changes in autoimmune disease susceptibility. Here we review the current literature on sex differences in lymphocyte function, the factors that influence this, and the implications for autoimmune disease. We propose that improved understanding of sex bias in lymphocyte function can provide sex-specific tailoring of treatment strategies for better management of autoimmune diseases.
Collapse
Affiliation(s)
- Katherine C. Dodd
- Lydia Becker Institute of Immunology and Inflammation, Division of Immunology, Immunity to Infection and Respiratory Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom,Manchester Centre for Clinical Neurosciences, Salford Royal Hospital, Salford, United Kingdom
| | - Madhvi Menon
- Lydia Becker Institute of Immunology and Inflammation, Division of Immunology, Immunity to Infection and Respiratory Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom,*Correspondence: Madhvi Menon,
| |
Collapse
|
5
|
Xu L, Huang G, Cong Y, Yu Y, Li Y. Sex-related Differences in Inflammatory Bowel Diseases: The Potential Role of Sex Hormones. Inflamm Bowel Dis 2022; 28:1766-1775. [PMID: 35486387 DOI: 10.1093/ibd/izac094] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Indexed: 12/13/2022]
Abstract
Inflammatory bowel disease (IBD), characterized by chronic inflammation of the gastrointestinal tract, is a global health care problem. Compelling evidence shows sex differences regarding the prevalence, pathophysiology, clinical presentation, and treatment outcome of IBD. Sex hormones, including estrogen, progesterone, and androgen, have been proposed to have a role in the pathogenesis of sexual dimorphism in IBD. Clinical and experimental data support the modulatory effects of sex hormones on various clinical characteristics of the disease, including intestinal barrier dysfunction and mucosal immune activation. Additionally, the potential role of sex hormones in the modulation of gut microbiota is attracting increasing attention. Here, we discuss the sex dimorphic disease profile and address the potential mechanisms involved in the sex-specific pathogenesis of IBD. Improved understanding of these sex differences in the clinic could improve the knowledge of patients with IBD with heterogeneous disease profiles.
Collapse
Affiliation(s)
- Leiqi Xu
- Department of Gastroenterology, Qilu Hospital, Shandong University, Jinan, Shandong, P.R. China
| | - Gang Huang
- Department of Gastroenterology, Qilu Hospital, Shandong University, Jinan, Shandong, P.R. China
| | - Yingzi Cong
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA.,Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Yanbo Yu
- Department of Gastroenterology, Qilu Hospital, Shandong University, Jinan, Shandong, P.R. China
| | - Yanqing Li
- Department of Gastroenterology, Qilu Hospital, Shandong University, Jinan, Shandong, P.R. China
| |
Collapse
|
6
|
Bi Z, Cao Y, Liu C, Gui M, Lin J, Zhang Q, Li Y, Ji S, Bu B. Remission and relapses of myasthenia gravis on long-term tacrolimus: a retrospective cross-sectional study of a Chinese cohort. Ther Adv Chronic Dis 2022; 13:20406223221122538. [PMID: 36093262 PMCID: PMC9459458 DOI: 10.1177/20406223221122538] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 08/02/2022] [Indexed: 11/23/2022] Open
Abstract
Objective: To identify the factors that predict the remission and relapses in myasthenia
gravis (MG) patients improved by prednisone and tacrolimus treatment. Methods: A retrospective, observational cohort analysis of MG patients who achieved
remission after receiving prednisone and tacrolimus were performed at Tongji
Hospital. The main outcome measures were the time to remission, prednisone
discontinuation, tacrolimus reduction–associated relapse, and treatment
outcome. Results: After adding tacrolimus, 256 patients were able to achieve remission with a
mean time to remission of 2.1 ± 1.4 months. After a median follow-up of
2.9 years, 167 patients (65.2%) discontinued prednisone, and 20 patients
(7.8%) achieved complete stable remission. Moreover, 53 of the 109 patients
who were tapering tacrolimus experienced relapses. In multivariable
analysis, female sex, low tacrolimus concentrations, and quantitative
myasthenia gravis (QMG) scores have a positive correlation with the time to
remission; concomitant additional autoimmune disease (AID) and high
anti-acetylcholine receptor antibody (AChR-ab) levels were significantly
associated with low probabilities of prednisone discontinuation [odds ratio
(OR) = 0.312–0.912, respectively]; rapid tacrolimus decrement speed
(⩾0.76 mg/year) was an independent predictor for the development of relapse
during tapering tacrolimus (OR = 5.662). Conclusion: Sex, tacrolimus concentrations, and QMG scores can be used as potential
predictors of the time to remission in MG patients treated with tacrolimus
and prednisone. Prednisone should be tapered slowly, especially in patients
with additional AID or high serum titers of AChR-ab. To avoid symptoms
recurrence, the dose of tacrolimus should reduce slowly, not exceeding
0.76 mg/year.
Collapse
Affiliation(s)
- Zhuajin Bi
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yayun Cao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Chenchen Liu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mengcui Gui
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Lin
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qing Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yue Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Suqiong Ji
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bitao Bu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, Hubei Province, China
| |
Collapse
|
7
|
Immune Cell Contributors to the Female Sex Bias in Multiple Sclerosis and Experimental Autoimmune Encephalomyelitis. Curr Top Behav Neurosci 2022; 62:333-373. [PMID: 35467295 DOI: 10.1007/7854_2022_324] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Multiple sclerosis (MS) is a chronic, autoimmune, demyelinating disease of the central nervous system (CNS) that leads to axonal damage and accumulation of disability. Relapsing-remitting MS (RR-MS) is the most frequent presentation of MS and this form of MS is three times more prevalent in females than in males. This female bias in MS is apparent only after puberty, suggesting a role for sex hormones in this regulation; however, very little is known of the biological mechanisms that underpin the sex difference in MS onset. Experimental autoimmune encephalomyelitis (EAE) is an animal model of RR-MS that presents more severely in females in certain mouse strains and thus has been useful to study sex differences in CNS autoimmunity. Here, we overview the immunopathogenesis of MS and EAE and how immune mechanisms in these diseases differ between a male and female. We further describe how females exhibit more robust myelin-specific T helper (Th) 1 immunity in MS and EAE and how this sex bias in Th cells is conveyed by sex hormone effects on the T cells, antigen presenting cells, regulatory T cells, and innate lymphoid cell populations.
Collapse
|
8
|
Su M, Liu X, Wang L, Song J, Zhou Z, Luo S, Zhao C. Risk factors for pregnancy-related clinical outcome in myasthenia gravis: a systemic review and meta-analysis. Orphanet J Rare Dis 2022; 17:52. [PMID: 35172854 PMCID: PMC8848664 DOI: 10.1186/s13023-022-02205-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 02/06/2022] [Indexed: 11/28/2022] Open
Abstract
OBJECTIVE Myasthenia gravis (MG) is an autoimmune disorder that frequently affects females at reproductive age. Herein, we aimed to assess the associations of clinical factors with pregnancy-related outcome in MG. METHODS We searched PubMed and EMBASE for case-control and cohort studies that reported the MG status during or after pregnancy and relevant clinical variables. The data was extracted in proportions and odds ratios (ORs) with 95% confidence intervals (CIs) in subsequent meta-analysis. RESULTS Fifteen eligible articles reporting on 734 pregnancies with 193 worsening and 51 improved episodes were included out of 1765 records. The estimated worsening proportions in total, antepartum and postpartum periods were 0.36 (95% CI 0.25-0.40), 0.23 (95% CI 0.14-0.34) and 0.11 (95% CI 0.04-0.22) respectively. The proportion of pregnancy-related improvement in enrolled patients was 0.28 (95% CI 0.17-0.40), with 0.07 (95% CI 0.00-0.28) during pregnancy and 0.14 (95% CI 0.02-0.34) after pregnancy. No significant associations were disclosed between the clinical factors and MG worsening. Thymectomy before delivery is a strong predictor for MG improvement in postpartum period (OR 4.85, 95% CI 1.88-12.50, p = 0.001). CONCLUSION The total proportion of pregnancy-related MG worsening and improvement in MG was 0.36 (95% CI 0.25-0.40) and 0.28 (95% CI 0.17-0.40), respectively. Thymectomy before the delivery may aid in clinical improvements associated with pregnancy. Future prospective cohort studies are required to determine more relevant factors.
Collapse
Affiliation(s)
- Manqiqige Su
- Huashan Rare Disease Center, Department of Neurology, Huashan Hospital, Fudan University, 200040, Shanghai, China
- National Center for Neurological Disorders, Shanghai, 200040, China
| | - Xiaoqing Liu
- Inner Mongolia Medical University, 010110, Inner Mongolia, China
| | - Liang Wang
- Huashan Rare Disease Center, Department of Neurology, Huashan Hospital, Fudan University, 200040, Shanghai, China
- National Center for Neurological Disorders, Shanghai, 200040, China
| | - Jie Song
- Huashan Rare Disease Center, Department of Neurology, Huashan Hospital, Fudan University, 200040, Shanghai, China
- National Center for Neurological Disorders, Shanghai, 200040, China
| | - Zhirui Zhou
- Radiation Oncology Center, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Sushan Luo
- Huashan Rare Disease Center, Department of Neurology, Huashan Hospital, Fudan University, 200040, Shanghai, China.
- National Center for Neurological Disorders, Shanghai, 200040, China.
| | - Chongbo Zhao
- Huashan Rare Disease Center, Department of Neurology, Huashan Hospital, Fudan University, 200040, Shanghai, China.
- National Center for Neurological Disorders, Shanghai, 200040, China.
| |
Collapse
|
9
|
Forgash JT, Chang YM, Mittelman NS, Petesch S, Benedicenti L, Galban E, Hammond JJ, Glass EN, Barker JR, Shelton GD, Luo J, Garden OA. Clinical features and outcome of acquired myasthenia gravis in 94 dogs. J Vet Intern Med 2021; 35:2315-2326. [PMID: 34331481 PMCID: PMC8478050 DOI: 10.1111/jvim.16223] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 06/25/2021] [Accepted: 07/12/2021] [Indexed: 12/22/2022] Open
Abstract
Background Factors known to be associated with outcome of acquired myasthenia gravis (MG) in dogs are limited. Hypothesis/Objectives Of dogs with MG, advancing age and comorbid neoplasia are associated with poor long‐term prognosis and low rates of remission. Animals Ninety‐four client‐owned dogs with MG diagnosed by acetylcholine receptor antibody (AChR Ab) assay between 2001 and 2019 from a university clinic and 3 private clinics in the United States. Methods Cases were retrospectively evaluated and data were collected to determine clinical signs, treatment, and response to therapy defined by means of a clinical scoring rubric. Immunological remission was defined as a return of the AChR Ab concentration to <0.6 nmol/L. Multivariable binary logistic regression analysis was used to identify clinical criteria predicting remission. Results An anticholinesterase drug was used to treat 90/94 (96%) dogs, which in 63/94 (67%) was the sole treatment; other drugs included immune modulators. Clinical remission (lack of clinical signs ≥4 weeks after treatment cessation) was observed in 29 (31% [95% confidence interval (CI): 22.4‐40.8%]) dogs, clinical response (lack of clinical signs on treatment) in 14 (15% [95% CI: 9.0‐23.6%]) dogs, clinical improvement (on treatment) in 24 (26% [95% CI: 17.8‐35.2%]) dogs, and no clinical improvement in 27 (29% [95% CI: 20.5‐38.6%]) dogs. Immunological remission was observed in 27/46 (59%) dogs, with clinical remission in all 27. Younger age (P = .04) and comorbid endocrine disease (P = .04) were associated with clinical remission. Initial AChR Ab concentration (P = .02) and regurgitation (P = .04) were negatively associated with clinical remission. Conclusions and Clinical Importance Clinical remission in MG is less likely in older dogs and dogs presenting with regurgitation or high initial AChR Ab concentration, but more likely in younger dogs and dogs with comorbid endocrine disease.
Collapse
Affiliation(s)
- Jennifer T Forgash
- Department of Clinical Sciences & Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Yu-Mei Chang
- Research Support Office, Royal Veterinary College, University of London, London, United Kingdom
| | - Neil S Mittelman
- Department of Clinical Sciences & Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Scott Petesch
- Department of Clinical Sciences & Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Leontine Benedicenti
- Department of Clinical Sciences & Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Evelyn Galban
- Department of Clinical Sciences & Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - James J Hammond
- Department of Neurology and Neurosurgery, Pieper Memorial Veterinary Center, Middletown, Connecticut, USA
| | - Eric N Glass
- Section of Neurology and Neurosurgery, Red Bank Veterinary Hospital, Tinton Falls, New Jersey, USA
| | - Jessica R Barker
- Department of Neurology and Neurosurgery, Bush Veterinary Neurology Service, Springfield, Virginia, USA
| | - G Diane Shelton
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, California, USA
| | - Jie Luo
- Department of Clinical Sciences & Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Oliver A Garden
- Department of Clinical Sciences & Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
10
|
Belimezi M, Kalliaropoulos A, Jiménez J, Garcia I, Mentis AFA, Chrousos GP. Age at sampling and sex distribution of AChRAb vs. MuSKAb myasthenia gravis in a large Greek population. Clin Neurol Neurosurg 2021; 208:106847. [PMID: 34343914 DOI: 10.1016/j.clineuro.2021.106847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 07/22/2021] [Accepted: 07/24/2021] [Indexed: 10/20/2022]
Abstract
OBJECTIVE Myasthenia gravis (MG) is a typical B-cell-mediated neuromuscular junction disease that can be classified into seropositive and seronegative subtypes. Association of patients' age at sampling and sex with the two major seropositive MG subcategories, i.e., MGs linked to antibodies directed against the acetylcholine receptor (AChRAb) and against the muscle-specific kinase (MuSKAb), has not been compared in a large population. METHODS We performed a retrospective analysis of samples from patients with MG in Greece who underwent neurochemical diagnostic evaluation between January 2, 2013, and August 31, 2016. RESULTS Overall, 1620 adult (623 male and 997 female patients; male-to-female ratio = 0.62) and 51 pediatric patients were found to be seropositive for MG. The distributions in both male and female patients were bimodal in the total and AChRAb MG cases but not in the total MuSKAb MG cases. Significant differences in the age at sampling distribution between the male and female adult patients were observed only in the AChRAb MG subtype. Significant differences between the AChRAb and MuSKAb MG categories were noted in the mean age values (60.10 and 51.49 years, respectively, for female and 65.69 and 56.19 years, respectively, for male adult patients). CONCLUSION Our findings confirm an uneven profile of age at sampling and sex between the AChRAb and MuSKAb MG cases in a large population. Future mechanistic studies can elucidate the cause of these differences. Moreover, clinical studies can explore how such differences can affect MG treatment and prognosis.
Collapse
Affiliation(s)
- Maria Belimezi
- Diagnostic Services Laboratory, Hellenic Pasteur Institute, Athens, Greece
| | | | - Juan Jiménez
- ADEMA Universitary School, University of the Balearic Islands (UIB), Palma, Balearic Islands, Spain
| | - Irene Garcia
- Department of Mathematical Sciences and Informatics, and Health Research Institute (IdISBa), University of the Balearic Islands (UIB), Palma, Balearic Islands, Spain
| | - Alexios-Fotios A Mentis
- Diagnostic Services Laboratory, Hellenic Pasteur Institute, Athens, Greece; University Research Institute of Maternal and Child Health and Precision Medicine, National and Kapodistrian University of Athens, Levadias, Athens, Greece.
| | - George P Chrousos
- University Research Institute of Maternal and Child Health and Precision Medicine, National and Kapodistrian University of Athens, Levadias, Athens, Greece
| |
Collapse
|
11
|
Sheikh S, Alvi U, Soliven B, Rezania K. Drugs That Induce or Cause Deterioration of Myasthenia Gravis: An Update. J Clin Med 2021; 10:jcm10071537. [PMID: 33917535 PMCID: PMC8038781 DOI: 10.3390/jcm10071537] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/24/2021] [Accepted: 03/31/2021] [Indexed: 12/20/2022] Open
Abstract
Myasthenia gravis (MG) is an autoimmune neuromuscular disorder which is characterized by presence of antibodies against acetylcholine receptors (AChRs) or other proteins of the postsynaptic membrane resulting in damage to postsynaptic membrane, decreased number of AChRs or blocking of the receptors by autoantibodies. A number of drugs such as immune checkpoint inhibitors, penicillamine, tyrosine kinase inhibitors and interferons may induce de novo MG by altering the immune homeostasis mechanisms which prevent emergence of autoimmune diseases such as MG. Other drugs, especially certain antibiotics, antiarrhythmics, anesthetics and neuromuscular blockers, have deleterious effects on neuromuscular transmission, resulting in increased weakness in MG or MG-like symptoms in patients who do not have MG, with the latter usually being under medical circumstances such as kidney failure. This review summarizes the drugs which can cause de novo MG, MG exacerbation or MG-like symptoms in nonmyasthenic patients.
Collapse
|
12
|
Construction of a TF-miRNA-gene feed-forward loop network predicts biomarkers and potential drugs for myasthenia gravis. Sci Rep 2021; 11:2416. [PMID: 33510225 PMCID: PMC7843995 DOI: 10.1038/s41598-021-81962-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 01/07/2021] [Indexed: 01/07/2023] Open
Abstract
Myasthenia gravis (MG) is an autoimmune disease and the most common type of neuromuscular disease. Genes and miRNAs associated with MG have been widely studied; however, the molecular mechanisms of transcription factors (TFs) and the relationship among them remain unclear. A TF–miRNA–gene network (TMGN) of MG was constructed by extracting six regulatory pairs (TF–miRNA, miRNA–gene, TF–gene, miRNA–TF, gene–gene and miRNA–miRNA). Then, 3/4/5-node regulatory motifs were detected in the TMGN. Then, the motifs with the highest Z-score, occurring as 3/4/5-node composite feed-forward loops (FFLs), were selected as statistically significant motifs. By merging these motifs together, we constructed a 3/4/5-node composite FFL motif-specific subnetwork (CFMSN). Then, pathway and GO enrichment analyses were performed to further elucidate the mechanism of MG. In addition, the genes, TFs and miRNAs in the CFMSN were also utilized to identify potential drugs. Five related genes, 3 TFs and 13 miRNAs, were extracted from the CFMSN. As the most important TF in the CFMSN, MYC was inferred to play a critical role in MG. Pathway enrichment analysis showed that the genes and miRNAs in the CFMSN were mainly enriched in pathways related to cancer and infections. Furthermore, 21 drugs were identified through the CFMSN, of which estradiol, estramustine, raloxifene and tamoxifen have the potential to be novel drugs to treat MG. The present study provides MG-related TFs by constructing the CFMSN for further experimental studies and provides a novel perspective for new biomarkers and potential drugs for MG.
Collapse
|
13
|
Bubuioc AM, Kudebayeva A, Turuspekova S, Lisnic V, Leone MA. The epidemiology of myasthenia gravis. J Med Life 2021; 14:7-16. [PMID: 33767779 PMCID: PMC7982252 DOI: 10.25122/jml-2020-0145] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 10/14/2020] [Indexed: 11/17/2022] Open
Abstract
Neuromuscular junction (NMJ) disorders include several dysfunctions that ultimately lead to muscle weakness. Myasthenia gravis (MG) is the most prevalent NMJ disorder with a highly polymorphic clinical presentation and many different faces. Being an autoimmune disease, MG correlates with the presence of detectable antibodies directed against the acetylcholine receptor, muscle-specific kinase, lipoprotein-related protein 4, agrin, titin, and ryanodine in the postsynaptic membrane at the NMJ. MG has become a prototype serving to understand both autoimmunity and the function of the NMJ better. The aim of this review is to synthesize some of the epidemiological data available. Epidemiological data regarding MG are important for postulating hypotheses regarding its etiology and facilitating the description of MG subtypes. Thus, adequate documentation through broad databases is essential. The incidence and prevalence of MG reported around the globe have been rising steadily and consistently over the past decades. Ethnic aspects, gender-related differences, and environmental risk factors have been described, implying that these might contribute to a specific phenotype, further suggesting that MG may be considered an umbrella term that covers several clinical entities.
Collapse
Affiliation(s)
- Ana-Maria Bubuioc
- Department of Neurology, Nicolae Testemitanu State University of Medicine and Pharmacy Chisinau, the Republic of Moldova
| | - Aigerim Kudebayeva
- Department of Neurology, Kazakh Medical University of Continuing Education, Almaty, Kazakhstan
| | - Saule Turuspekova
- Department of Nervous Diseases with course of Neurosurgery, Asfendiyarov Kazakh National Medical University, Almaty, Kazakhstan
| | - Vitalie Lisnic
- Department of Neurology, Nicolae Testemitanu State University of Medicine and Pharmacy Chisinau, the Republic of Moldova
| | - Maurizio Angelo Leone
- Neurology Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| |
Collapse
|
14
|
Priyanka HP, Nair RS. Neuroimmunomodulation by estrogen in health and disease. AIMS Neurosci 2020; 7:401-417. [PMID: 33263078 PMCID: PMC7701372 DOI: 10.3934/neuroscience.2020025] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/27/2020] [Indexed: 12/18/2022] Open
Abstract
Systemic homeostasis is maintained by the robust bidirectional regulation of the neuroendocrine-immune network by the active involvement of neural, endocrine and immune mediators. Throughout female reproductive life, gonadal hormones undergo cyclic variations and mediate concomitant modulations of the neuroendocrine-immune network. Dysregulation of the neuroendocrine-immune network occurs during aging as a cumulative effect of declining neural, endocrine and immune functions and loss of compensatory mechanisms including antioxidant enzymes, growth factors and co-factors. This leads to disruption of homeostasis and sets the stage for the development of female-specific age-associated diseases such as autoimmunity, osteoporosis, cardiovascular diseases and hormone-dependent cancers. Ovarian hormones especially estrogen, play a key role in the maintenance of health and homeostasis by modulating the nervous, endocrine and immune functions and thereby altering neuroendocrine-immune homeostasis. Immunologically estrogen's role in the modulation of Th1/Th2 immune functions and contributing to pro-inflammatory conditions and autoimmunity has been widely studied. Centrally, hypothalamic and pituitary hormones influence gonadal hormone secretion in murine models during onset of estrous cycles and are implicated in reproductive aging-associated acyclicity. Loss of estrogen affects neuronal plasticity and the ensuing decline in cognitive functions during reproductive aging in females implicates estrogen in the incidence and progression of neurodegenerative diseases. Peripherally, sympathetic noradrenergic (NA) innervations of lymphoid organs and the presence of both adrenergic (AR) and estrogen receptors (ER) on lymphocytes poise estrogen as a potent neuroimmunomodulator during health and disease. Cyclic variations in estrogen levels throughout reproductive life, perimenopausal surge in estrogen levels followed by its precipitous decline, concomitant with decline in central hypothalamic catecholaminergic activity, peripheral sympathetic NA innervation and associated immunosuppression present an interesting study to explore female-specific age-associated diseases in a new light.
Collapse
Affiliation(s)
- Hannah P Priyanka
- Inspire Laboratory, Institute of Advanced Research in Health Sciences, Tamil Nadu Government Multi Super Speciality Hospital, Omandurar Government Estate, Chennai-600002, India
| | | |
Collapse
|
15
|
Krenn M, Grisold A, Wohlfarth P, Rath J, Cetin H, Koneczny I, Zimprich F. Pathomechanisms and Clinical Implications of Myasthenic Syndromes Exacerbated and Induced by Medical Treatments. Front Mol Neurosci 2020; 13:156. [PMID: 32922263 PMCID: PMC7457047 DOI: 10.3389/fnmol.2020.00156] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 07/28/2020] [Indexed: 12/21/2022] Open
Abstract
Myasthenic syndromes are typically characterized by muscle weakness and increased fatigability due to an impaired transmission at the neuromuscular junction (NMJ). Most cases are caused by acquired autoimmune conditions such as myasthenia gravis (MG), typically with antibodies against the acetylcholine receptor (AChR). Different drugs are among the major factors that may complicate pre-existing autoimmune myasthenic conditions by further impairing transmission at the NMJ. Some clinical observations are substantiated by experimental data, indicating that presynaptic, postsynaptic or more complex pathomechanisms at the NMJ may be involved, depending on the individual compound. Most robust data exist for the risks associated with some antibiotics (e.g., aminoglycosides, ketolides, fluoroquinolones) and cardiovascular medications (e.g., class Ia antiarrhythmics, beta blockers). Apart from primarily autoimmune-mediated disorders of the NMJ, de novo myasthenic manifestations may also be triggered by medical treatments that induce an autoimmune reaction. Most notably, there is growing evidence that the immune checkpoint inhibitors (ICI), a modern class of drugs to treat various malignancies, represent a relevant risk factor to develop severe and progressive medication-induced myasthenia via an immune-mediated mechanism. From a clinical perspective, it is of utmost importance for the treating physicians to be aware of such adverse treatment effects and their consequences. In this article, we aim to summarize existing evidence regarding the key molecular and immunological mechanisms as well as the clinical implications of medication-aggravated and medication-induced myasthenic syndromes.
Collapse
Affiliation(s)
- Martin Krenn
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Anna Grisold
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Philipp Wohlfarth
- Division of Blood and Marrow Transplantation, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Jakob Rath
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Hakan Cetin
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Inga Koneczny
- Department of Neurology, Medical University of Vienna, Vienna, Austria.,Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Fritz Zimprich
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
16
|
Jiao W, Hu F, Li J, Song J, Liang J, Li L, Song Y, Chen Z, Li Q, Ke L. Qiangji Jianli Decoction promotes mitochondrial biogenesis in skeletal muscle of myasthenia gravis rats via AMPK/PGC-1α signaling pathway. Biomed Pharmacother 2020; 129:110482. [PMID: 32768964 DOI: 10.1016/j.biopha.2020.110482] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 05/29/2020] [Accepted: 06/30/2020] [Indexed: 11/30/2022] Open
Abstract
The Qiangji Jianli Decoction (QJJLD) is an effective Chinese medicine formula for treating Myasthenia gravis (MG) in the clinic. QJJLD has been proven to regulate mitochondrial fusion and fission of skeletal muscle in myasthenia gravis. In this study, we investigated whether QJJLD plays a therapeutic role in regulating mitochondrial biogenesis in MG and explored the underlying mechanism. Rats were experimentally induced to establish autoimmune myasthenia gravis (EAMG) by subcutaneous immunization with R97-116 peptides. The treatment groups were administered three different dosages of QJJLD respectively. After the intervention of QJJLD, the pathological changes of gastrocnemius muscle in MG rats were significantly improved; SOD, GSH-Px, Na+-K+ ATPase and Ca2+-Mg2+ ATPase activities were increased; and MDA content was decreased in the gastrocnemius muscle. Moreover, AMPK, p38MAPK, PGC-1α, NRF-1, Tfam and COX IV mRNA and protein expression levels were also reversed by QJJLD. These results implied that QJJLD may provide a potential therapeutic strategy through promoting mitochondrial biogenesis to alleviate MG via activating the AMPK/PGC-1α signaling pathway.
Collapse
Affiliation(s)
- Wei Jiao
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China; Institute of Pi-Wei, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
| | - Fangyu Hu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China; Institute of Pi-Wei, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
| | - Jinqiu Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China; Institute of Pi-Wei, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
| | - Jingwei Song
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China; Institute of Pi-Wei, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
| | - Jian Liang
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
| | - Lanqi Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China; Institute of Pi-Wei, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
| | - Yafang Song
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China; Institute of Pi-Wei, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China.
| | - Zhiwei Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China; Institute of Pi-Wei, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
| | - Qing Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China; Institute of Pi-Wei, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
| | - Lingling Ke
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China; Institute of Pi-Wei, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
| |
Collapse
|
17
|
Yee Mon KJ, Goldsmith E, Watson NB, Wang J, Smith NL, Rudd BD. Differential Sensitivity to IL-12 Drives Sex-Specific Differences in the CD8+ T Cell Response to Infection. Immunohorizons 2020; 3:121-132. [PMID: 31317126 PMCID: PMC6636834 DOI: 10.4049/immunohorizons.1800066] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
It is well known that males and females respond differently to intracellular pathogens. Females mount a more robust immune response than males, which decreases their susceptibility to infection but comes at the cost of increasing immunopathology. However, the underlying basis for sex-specific differences in the CD8+ T cell response to infection remains poorly understood. In this study, we show that female CD8+ T cells have an intrinsic propensity to become short-lived effectors, whereas male CD8+ T cells give rise to more memory precursor effector cells after murine infection with either a virus (vaccinia virus) or bacteria (Listeria monocytogenes). Interestingly, we found that the propensity of female CD8+ T cells to form short-lived effectors is not because they respond to lower amounts of cognate Ag but rather because they have an enhanced capacity to respond to IL-12, which facilitates more effector cell differentiation at each round of cell division. Our findings provide key insights into the sex-based immunological differences that underlie variations in the susceptibility to infection in males and females. ImmunoHorizons, 2019, 3: 121–132.
Collapse
Affiliation(s)
- Kristel Joy Yee Mon
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853; and
| | - Elizabeth Goldsmith
- College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523
| | - Neva B Watson
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853; and
| | - Jocelyn Wang
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853; and
| | - Norah L Smith
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853; and
| | - Brian D Rudd
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853; and
| |
Collapse
|
18
|
Merrheim J, Villegas J, Van Wassenhove J, Khansa R, Berrih-Aknin S, le Panse R, Dragin N. Estrogen, estrogen-like molecules and autoimmune diseases. Autoimmun Rev 2020; 19:102468. [PMID: 31927086 DOI: 10.1016/j.autrev.2020.102468] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 09/23/2019] [Indexed: 12/13/2022]
Abstract
In western countries, the slope of autoimmune disease (AD) incidence is increasing and affects 5-8% of the population. Mainly prevalent in women, these pathologies are due to thymic tolerance processes breakdown. The female sex hormone, estrogen, is involved in this AD female susceptibility. However, predisposition factors have to act in concert with unknown triggering environmental factors (virus, microbiota, pollution) to initiate AD. Individuals are exposed to various environmental compounds that display endocrine disruption abilities. The cellular effects of some of these molecules may be mediated through the aryl hydrocarbon receptor (AhR). Here, we review the effects of these molecules on the homeostasis of the thymic cells, the immune tolerance intrinsic factors (transcription factors, epigenetic marks) and on the immune tolerance extrinsic factors (microbiota, virus sensibility). This review highlights the contribution of estrogen and endocrine disruptors on the dysregulation of mechanisms sustaining AD development.
Collapse
Affiliation(s)
- Judith Merrheim
- Sorbonne Université, Paris, France; Inserm UMRS 974, Paris, France; AIM, Institute of Myology, Paris, France; Centre de Recherche en Myologie, Sorbonne Université, Inserm UMRS 974, Hôpital La Pitié- Salpêtrière, 105 Bd de l'hôpital, 75013 Paris, France
| | - José Villegas
- Sorbonne Université, Paris, France; Inserm UMRS 974, Paris, France; AIM, Institute of Myology, Paris, France; Centre de Recherche en Myologie, Sorbonne Université, Inserm UMRS 974, Hôpital La Pitié- Salpêtrière, 105 Bd de l'hôpital, 75013 Paris, France
| | - Jérôme Van Wassenhove
- Sorbonne Université, Paris, France; Inserm UMRS 974, Paris, France; AIM, Institute of Myology, Paris, France; Centre de Recherche en Myologie, Sorbonne Université, Inserm UMRS 974, Hôpital La Pitié- Salpêtrière, 105 Bd de l'hôpital, 75013 Paris, France
| | - Rémi Khansa
- Sorbonne Université, Paris, France; Inserm UMRS 974, Paris, France; AIM, Institute of Myology, Paris, France; Centre de Recherche en Myologie, Sorbonne Université, Inserm UMRS 974, Hôpital La Pitié- Salpêtrière, 105 Bd de l'hôpital, 75013 Paris, France
| | - Sonia Berrih-Aknin
- Sorbonne Université, Paris, France; Inserm UMRS 974, Paris, France; AIM, Institute of Myology, Paris, France; Centre de Recherche en Myologie, Sorbonne Université, Inserm UMRS 974, Hôpital La Pitié- Salpêtrière, 105 Bd de l'hôpital, 75013 Paris, France
| | - Rozen le Panse
- Sorbonne Université, Paris, France; Inserm UMRS 974, Paris, France; AIM, Institute of Myology, Paris, France; Centre de Recherche en Myologie, Sorbonne Université, Inserm UMRS 974, Hôpital La Pitié- Salpêtrière, 105 Bd de l'hôpital, 75013 Paris, France
| | - Nadine Dragin
- Sorbonne Université, Paris, France; Inserm UMRS 974, Paris, France; Inovarion, Paris, France; Centre de Recherche en Myologie, Sorbonne Université, Inserm UMRS 974, Hôpital La Pitié- Salpêtrière, 105 Bd de l'hôpital, 75013 Paris, France.
| |
Collapse
|
19
|
Fang W, Li Y, Mo R, Wang J, Qiu L, Ou C, Lin Z, Huang Z, Feng H, He X, Wang W, Xu P, Wang L, Ran H, Liu W. Hospital and healthcare insurance system record-based epidemiological study of myasthenia gravis in southern and northern China. Neurol Sci 2020; 41:1211-1223. [PMID: 31897952 DOI: 10.1007/s10072-019-04146-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 11/07/2019] [Indexed: 12/31/2022]
Abstract
OBJECTIVE This is the first cross-region epidemiological study of myasthenia gravis (MG) in China. We estimated the incidence, prevalence, and medical costs of MG in southern China and explored the differences between the southern and northern Chinese populations. METHODS We collected and analyzed records from 20 hospitals in the southern city, Guangzhou, 13 hospitals in the northern city, Harbin, and two healthcare insurance systems: job based and residence based in Guangzhou during 2000-2017. RESULTS (1) The estimated annual incidence of MG was 1.55-3.66 per 100,000, and the estimated prevalence of MG was 2.19-11.07 per 100,000 in southern China based on insurance records. (2) The proportion of hospitalized MG patients in the south-based hospital records was three times as high as that in the north-based hospital records. (3) Female MG prevalence was significantly higher than male MG prevalence in Guangzhou, while the similar gender difference in Harbin was not statistically significant due to higher variation in earlier years. (4) The average expense was $35-42 for each outpatient service and $2526-2673 for each hospitalization expense in the south. (5) Contrary to the increase of insurance-based estimate of MG prevalence, the proportion of hospitalized MG patients did not increase over the years, suggesting rising awareness and utilization of health insurance. CONCLUSIONS The southern MG population had a significantly higher prevalence and a lower response threshold to medication than the northern MG population. These results are calling for further investigations on the genetic, cultural, and environmental variations of the Chinese MG populations between north and south.
Collapse
Affiliation(s)
- Wei Fang
- School of Earth and Environmental Sciences, Queens College, City University of New York, Queens, NY, USA
| | - Yan Li
- Department of Neurology, The First Affiliated Hospital of Sun Yat-sen University, #58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, People's Republic of China.,Department of Neurosurgical Intensive Care Unit, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Rong Mo
- Department of Neurology, The First Affiliated Hospital of Sun Yat-sen University, #58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, People's Republic of China
| | - Jianjian Wang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Li Qiu
- Department of Neurology, The First Affiliated Hospital of Sun Yat-sen University, #58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, People's Republic of China
| | - Chuangyi Ou
- Department of Neurology, The First Affiliated Hospital of Sun Yat-sen University, #58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, People's Republic of China
| | - Zhongqiang Lin
- Department of Neurology, The First Affiliated Hospital of Sun Yat-sen University, #58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, People's Republic of China
| | - Zhidong Huang
- Department of Neurology, The First Affiliated Hospital of Sun Yat-sen University, #58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, People's Republic of China
| | - Huiyu Feng
- Department of Neurology, The First Affiliated Hospital of Sun Yat-sen University, #58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, People's Republic of China
| | - Xuetao He
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, People's Republic of China
| | - Weizhi Wang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Peipei Xu
- Faculty of Geographical Science, Beijing Normal University, Beijing, People's Republic of China
| | - Lihua Wang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China.
| | - Hao Ran
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, People's Republic of China.
| | - Weibin Liu
- Department of Neurology, The First Affiliated Hospital of Sun Yat-sen University, #58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, People's Republic of China.
| |
Collapse
|
20
|
Cao L, Liu W, Zhu Z. Clinical characteristics and relationship between myasthenia gravis and premature ovarian failure: report of two cases. J Int Med Res 2019; 47:3992-3997. [PMID: 31342860 PMCID: PMC6726818 DOI: 10.1177/0300060519863525] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Myasthenia gravis (MG) and premature ovarian failure (POF) are rare. MF and POF greatly affect patients’ health. Combined occurrence of MF and POF in young women can have serious consequences. We report two cases of MG with POF. Case 1 was a 20-year-old woman who presented with myasthenic crisis and menopause in September 2015 and November 2015, respectively. The patient’s estradiol and follicle-stimulating hormone levels were abnormal. She was administered plasmapheresis and methylprednisolone pulse therapy. She improved and was discharged with normal restoration of menstruation after 3 months. Case 2 was a 21-year-old woman who had right eyelid droop and double vision in June 2014. She presented with menstrual disorder and menopause in August 2014 and September 2014, respectively. Estradiol and follicle-stimulating hormone levels were abnormal. She underwent progesterone therapy. She was admitted to hospital again in March 2016 with a myasthenic crisis. She received methylprednisolone pulse therapy and underwent thymectomy, but menstruation was not restored. In conclusion, there is comorbidity of POF in MG, and there is a close relationship between these two diseases. MG may subsequently lead to development of POF, and timely immunotherapy for MG may normalize POF.
Collapse
Affiliation(s)
- Liming Cao
- Department of Neurology, The Third Affiliated Hospital of Shenzhen University, Shenzhen City, China
- Liming Cao, Department of Neurology, The Third Affiliated Hospital of Shenzhen University, 47 Friendship Road, Luohu District, Shenzhen City, China, 518000.
| | - Weibin Liu
- Department of Neurology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou City, China
| | - Zhishan Zhu
- Department of Neurology, The Third Affiliated Hospital of Shenzhen University, Shenzhen City, China
| |
Collapse
|
21
|
Abstract
PURPOSE OF REVIEW To give an overview of recently published articles addressing the mechanisms underlying sex bias in autoimmune disease. RECENT FINDINGS Recent studies investigating the origins of sex bias in autoimmune disease have revealed an extensive and interconnected network of genetic, hormonal, microbial, and environmental influences. Investigation of sex hormones has moved beyond profiling the effects of hormones on activity and prevalence of immune cell types to defining the specific immunity-related genes driving these changes. Deeper examination of the genetic content of the X and Y chromosomes and genetic escapees of X chromosome inactivation has revealed some key drivers of female-biased autoimmunity. Animal studies are offering further insights into the connections among microbiota, particularly that of the gut, and the immune system. SUMMARY Sex bias in autoimmune disease is the manifestation of a complex interplay of the sex chromosomes, sex hormones, the microbiota, and additional environmental and sociological factors.
Collapse
|
22
|
Asmail A, Kesler A, Kolb H, Drory VE, Karni A. A tri-modal distribution of age-of-onset in female patients with myasthenia gravis is associated with the gender-related clinical differences. Int J Neurosci 2018; 129:313-319. [DOI: 10.1080/00207454.2018.1529669] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Ali Asmail
- Neuroimmunology Service, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Anat Kesler
- Neuro-ophthalmology Unit of the Department of Ophthalmology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- The Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Hadar Kolb
- Neuroimmunology Service, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Vivian E. Drory
- The Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Neuromuscular Service of the Department of Neurology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Arnon Karni
- Neuroimmunology Service, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- The Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Segol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
23
|
Bernard I, Sacquin A, Kassem S, Benamar M, Colacios C, Gador M, Pérals C, Fazilleau N, Saoudi A. A Natural Variant of the Signaling Molecule Vav1 Enhances Susceptibility to Myasthenia Gravis and Influences the T Cell Receptor Repertoire. Front Immunol 2018; 9:2399. [PMID: 30410484 PMCID: PMC6210741 DOI: 10.3389/fimmu.2018.02399] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 09/27/2018] [Indexed: 01/01/2023] Open
Abstract
The guanine nucleotide exchange factor Vav1 is essential for transducing T cell receptor (TCR) signals and plays an important role in T cell development and activation. Previous genetic studies identified a natural variant of Vav1 characterized by the substitution of an arginine (R) residue by a tryptophane (W) at position 63 (Vav1R63W). This variant impacts Vav1 adaptor functions and controls susceptibility to T cell-mediated neuroinflammation. To assess the implication of this Vav1 variant on the susceptibility to antibody-mediated diseases, we used the animal model of myasthenia gravis, experimental autoimmune myasthenia gravis (EAMG). To this end, we generated a knock-in (KI) mouse model bearing a R to W substitution in the Vav1 gene (Vav1R63W) and immunized it with either torpedo acetylcholine receptor (tAChR) or the α146-162 immunodominant peptide. We observed that the Vav1R63W conferred increased susceptibility to EAMG, revealed by a higher AChR loss together with an increased production of effector cytokines (IFN-γ, IL-17A, GM-CSF) by antigen-specific CD4+ T cells, as well as an increased frequency of antigen-specific CD4+ T cells. This correlated with the emergence of a dominant antigen-specific T cell clone in KI mice that was not present in wild-type mice, suggesting an impact on thymic selection and/or a different clonal selection threshold following antigen encounter. Our results highlight the key role of Vav1 in the pathophysiology of EAMG and this was associated with an impact on the TCR repertoire of AChR reactive T lymphocytes.
Collapse
Affiliation(s)
- Isabelle Bernard
- Centre de Physiopathologie de Toulouse Purpan, Université de Toulouse, UPS, Inserm, CNRS, Toulouse, France
| | - Antoine Sacquin
- Centre de Physiopathologie de Toulouse Purpan, Université de Toulouse, UPS, Inserm, CNRS, Toulouse, France
| | - Sahar Kassem
- Centre de Physiopathologie de Toulouse Purpan, Université de Toulouse, UPS, Inserm, CNRS, Toulouse, France
| | - Mehdi Benamar
- Centre de Physiopathologie de Toulouse Purpan, Université de Toulouse, UPS, Inserm, CNRS, Toulouse, France
| | - Céline Colacios
- Centre de Physiopathologie de Toulouse Purpan, Université de Toulouse, UPS, Inserm, CNRS, Toulouse, France
| | - Mylène Gador
- Centre de Physiopathologie de Toulouse Purpan, Université de Toulouse, UPS, Inserm, CNRS, Toulouse, France
| | - Corine Pérals
- Centre de Physiopathologie de Toulouse Purpan, Université de Toulouse, UPS, Inserm, CNRS, Toulouse, France
| | - Nicolas Fazilleau
- Centre de Physiopathologie de Toulouse Purpan, Université de Toulouse, UPS, Inserm, CNRS, Toulouse, France
| | - Abdelhadi Saoudi
- Centre de Physiopathologie de Toulouse Purpan, Université de Toulouse, UPS, Inserm, CNRS, Toulouse, France
| |
Collapse
|
24
|
Schwinge D, Schramm C. Sex-related factors in autoimmune liver diseases. Semin Immunopathol 2018; 41:165-175. [DOI: 10.1007/s00281-018-0715-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 09/18/2018] [Indexed: 12/13/2022]
|
25
|
Bereshchenko O, Bruscoli S, Riccardi C. Glucocorticoids, Sex Hormones, and Immunity. Front Immunol 2018; 9:1332. [PMID: 29946321 PMCID: PMC6006719 DOI: 10.3389/fimmu.2018.01332] [Citation(s) in RCA: 160] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 05/29/2018] [Indexed: 12/15/2022] Open
Abstract
Glucocorticoid hormones regulate essential body functions in mammals, control cell metabolism, growth, differentiation, and apoptosis. Importantly, they are potent suppressors of inflammation, and multiple immune-modulatory mechanisms involving leukocyte apoptosis, differentiation, and cytokine production have been described. Due to their potent anti-inflammatory and immune-suppressive activity, synthetic glucocorticoids (GCs) are the most prescribed drugs used for treatment of autoimmune and inflammatory diseases. It is long been noted that males and females exhibit differences in the prevalence in several autoimmune diseases (AD). This can be due to the role of sexual hormones in regulation of the immune responses, acting through their endogenous nuclear receptors to mediate gene expression and generate unique gender-specific cellular environments. Given the fact that GCs are the primary physiological anti-inflammatory hormones, and that sex hormones may also exert immune-modulatory functions, the link between GCs and sex hormones may exist. Understanding the nature of this possible crosstalk is important to unravel the reason of sexual disparity in AD and to carefully prescribe these drugs for the treatment of inflammatory diseases. In this review, we discuss similarities and differences between the effects of sex hormones and GCs on the immune system, to highlight possible axes of functional interaction.
Collapse
Affiliation(s)
- Oxana Bereshchenko
- Section of Pharmacology, Department of Medicine, University of Perugia, Perugia, Italy.,Department of Surgery and Biomedical Sciences, University of Perugia, Perugia, Italy
| | - Stefano Bruscoli
- Section of Pharmacology, Department of Medicine, University of Perugia, Perugia, Italy
| | - Carlo Riccardi
- Section of Pharmacology, Department of Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
26
|
Pedersen AL, Brownrout JL, Saldanha CJ. Neuroinflammation and neurosteroidogenesis: Reciprocal modulation during injury to the adult zebra finch brain. Physiol Behav 2018; 187:51-56. [DOI: 10.1016/j.physbeh.2017.10.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 10/11/2017] [Accepted: 10/11/2017] [Indexed: 01/10/2023]
|
27
|
Wu X, Tong B, Yang Y, Luo J, Yuan X, Wei Z, Yue M, Xia Y, Dai Y. Arctigenin functions as a selective agonist of estrogen receptor β to restrict mTORC1 activation and consequent Th17 differentiation. Oncotarget 2018; 7:83893-83906. [PMID: 27863380 PMCID: PMC5356633 DOI: 10.18632/oncotarget.13338] [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: 08/26/2016] [Accepted: 11/08/2016] [Indexed: 12/13/2022] Open
Abstract
Arctigenin was previously proven to inhibit Th17 cell differentiation and thereby attenuate colitis in mice by down-regulating the activation of mechanistic target of rapamycin complex 1 (mTORC1). The present study was performed to address its underlying mechanism in view of estrogen receptor (ER). The specific antagonist PHTPP or siRNA of ERβ largely diminished the inhibitory effect of arctigenin on the mTORC1 activation in T cell lines and primary CD4+ T cells under Th17-polarization condition, suggesting that arctigenin functioned in an ERβ-dependent manner. Moreover, arctigenin was recognized to be an agonist of ERβ, which could bind to ERβ with a moderate affinity, promote dissociation of ERβ/HSP90 complex and nuclear translocation and phosphorylation of ERβ, and increase the transcription activity. Following activation of ERβ, arctigenin inhibited the activity of mTORC1 by disruption of ERβ-raptor-mTOR complex assembly. Deficiency of ERβ markedly abolished arctigenin-mediated inhibition of Th17 cell differentiation. In colitis mice, the activation of ERβ, inhibition of mTORC1 activation and Th17 response by arctigenin were abolished by PHTPP treatment. In conclusion, ERβ might be the target protein of arctigenin responsible for inhibition of mTORC1 activation and resultant prevention of Th17 cell differentiation and colitis development.
Collapse
Affiliation(s)
- Xin Wu
- Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Tong Jia Xiang, Nanjing, China.,Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Bei Tong
- Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Tong Jia Xiang, Nanjing, China
| | - Yan Yang
- Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Tong Jia Xiang, Nanjing, China
| | - Jinque Luo
- Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Tong Jia Xiang, Nanjing, China
| | - Xusheng Yuan
- Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Tong Jia Xiang, Nanjing, China
| | - Zhifeng Wei
- Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Tong Jia Xiang, Nanjing, China
| | - Mengfan Yue
- Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Tong Jia Xiang, Nanjing, China
| | - Yufeng Xia
- Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Tong Jia Xiang, Nanjing, China
| | - Yue Dai
- Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Tong Jia Xiang, Nanjing, China
| |
Collapse
|
28
|
Santos E, Braga A, Gabriel D, Duarte S, Martins da Silva A, Matos I, Freijo M, Martins J, Silveira F, Nadais G, Sousa F, Fraga C, Santos Silva R, Lopes C, Gonçalves G, Pinto C, Sousa Braga J, Leite MI. MuSK myasthenia gravis and pregnancy. Neuromuscul Disord 2017; 28:150-153. [PMID: 29305138 DOI: 10.1016/j.nmd.2017.11.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 10/19/2017] [Accepted: 11/21/2017] [Indexed: 01/15/2023]
Abstract
Muscle specific kinase (MuSK) myasthenia gravis (MG, MuSK-MG) is a rare subgroup of MG affecting mainly women during childbearing years. We investigated the influence of pregnancy in the course of MuSK-MG and pregnancy outcomes in females with MuSK-MG. A multicentre cohort of 17 women with MuSK-MG was studied retrospectively; 13 of them with ≥1 pregnancy. MuSK-MG onset age was 35,4 years; 23,0% had other autoimmune disorder; 46,2% were treatment refractory. Thirteen women experienced 27 pregnancies, either after MG onset (group I) (n = 4; maternal age at conception = 29.8 years) or before MG onset (group II) (n = 23; maternal age at conception = 26.2 years). In group I pregnancy occurred in average 9.8 years after the MG onset; it occurred in average 17.0 years before MG in group II. In group I, all were on steroids at time of conception, one on azathioprine and another receiving IVIG regularly. There were mild exacerbations that responded to treatment adjustments. There were no relapses in the 12 months following the delivery. There was no pre-eclampsia, birth defects or stillbirths in either group; 3 miscarriages in group II. One case of neonatal MG was recorded. In this small series, pregnancy did not seem to precipitate MuSK-MG or to have a major influence in the MuSK-MG course, and there was no apparent negative impact in pregnancy outcomes in those where pregnancy followed the MG onset. The weight was lower in the newborn of the group I mothers, although none had low birth weight.
Collapse
Affiliation(s)
- Ernestina Santos
- Neurology Department, Hospital Santo Antonio, Centro Hospitalar Porto, Porto, Portugal; Unit for Multidisciplinary Research in Biomedicine, Instituto de Ciencias Biomedicas de Abel Salazar, Universidade do Porto, Porto, Portugal.
| | - Antonio Braga
- Obstetrics Department, Centro Materno-Infantil do Norte, Centro Hospitalar Porto, Porto, Portugal
| | - Denis Gabriel
- Neurology Department, Hospital Santo Antonio, Centro Hospitalar Porto, Porto, Portugal
| | - Sara Duarte
- Neurology Department, Hospital Santo Antonio, Centro Hospitalar Porto, Porto, Portugal
| | - Ana Martins da Silva
- Neurology Department, Hospital Santo Antonio, Centro Hospitalar Porto, Porto, Portugal; Unit for Multidisciplinary Research in Biomedicine, Instituto de Ciencias Biomedicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Ilda Matos
- Neurology Department, Centro Hospitalar do Nordeste, Mirandela, Portugal
| | - Marta Freijo
- Neurology Department, Centro Hospitalar do Nordeste, Mirandela, Portugal
| | - Joao Martins
- Neurology Department, Hospital de Pedro Hispano, Matosinhos, Portugal
| | | | - Goreti Nadais
- Neurology Department, Hospital Sao Joao, Porto, Portugal
| | - Filipa Sousa
- Neurology Department, Hospital de Braga, Braga, Portugal
| | - Carla Fraga
- Centro Hospitalar do Vale do Sousa, Penafiel, Portugal
| | - Rosa Santos Silva
- Neurology Department, Centro Hospitalar do Alto Minho, Viana do Castelo, Portugal
| | - Carlos Lopes
- Instituto de Ciencias Biomedicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Guilherme Gonçalves
- Unit for Multidisciplinary Research in Biomedicine, Instituto de Ciencias Biomedicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Clara Pinto
- Obstetrics Department, Centro Materno-Infantil do Norte, Centro Hospitalar Porto, Porto, Portugal
| | - Jorge Sousa Braga
- Obstetrics Department, Centro Materno-Infantil do Norte, Centro Hospitalar Porto, Porto, Portugal
| | - Maria Isabel Leite
- Nuffield Department of Clinical Neurosciences, Oxford University Hospitals, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
29
|
Activation of the peripheral immune system regulates neuronal aromatase in the adult zebra finch brain. Sci Rep 2017; 7:10191. [PMID: 28860515 PMCID: PMC5579002 DOI: 10.1038/s41598-017-10573-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 08/10/2017] [Indexed: 11/23/2022] Open
Abstract
Estradiol provision via neural aromatization decreases neuro-inflammation and –degeneration, but almost nothing is known about the interactions between the peripheral immune system and brain aromatase. Given the vulnerability of the CNS we reasoned that brain aromatization may protect circuits from the threats of peripheral infection; perhaps shielding cells that are less resilient from the degeneration associated with peripheral infection or trauma. Lipopolysaccharide (LPS) or vehicle was administered peripherally to adult zebra finches and sickness behavior was recorded 2 or 24 hours later. The central transcription of cytokines and aromatase was measured, as were telencephalic aromatase activity and immunoreactive aromatase (24 hour time point only). Two hours post LPS, sickness-like behaviors increased, the transcription of IL-1β was higher in both sexes, and TNFα was elevated in females. 24 hours post-LPS, the behavior of LPS birds was similar to controls, and cytokines had returned to baseline, but aromatase mRNA and activity were elevated in both sexes. Immunocytochemistry revealed greater numbers of aromatase-expressing neurons in LPS birds. These data suggest that the activation of the immune system via peripheral endotoxin increases neuronal aromatase; a mechanism that may rapidly generate a potent anti-neuroinflammatory steroid in response to peripheral activation of the immune system.
Collapse
|
30
|
Dragin N, Nancy P, Villegas J, Roussin R, Le Panse R, Berrih-Aknin S. Balance between Estrogens and Proinflammatory Cytokines Regulates Chemokine Production Involved in Thymic Germinal Center Formation. Sci Rep 2017; 7:7970. [PMID: 28801669 PMCID: PMC5554297 DOI: 10.1038/s41598-017-08631-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 07/12/2017] [Indexed: 12/11/2022] Open
Abstract
The early-onset form of Myasthenia Gravis (MG) is prevalent in women and associates with ectopic germinal centers (GCs) development and inflammation in the thymus. we aimed to investigate the contribution of estrogens in the molecular processes involved in thymic GCs formation. We examined expression of genes involved in anti-acetylcholine receptor (AChR) response in MG, MHC class II and α-AChR subunit as well as chemokines involved in GC development (CXCL13, CCL21and CXCL12). In resting conditions, estrogens have strong regulatory effects on thymic epithelial cells (TECs), inducing a decreased protein expression of the above molecules. In knockout mouse models for estrogen receptor or aromatase, we observed that perturbation in estrogen transduction pathway altered MHC Class II, α-AChR, and CXCL13 expression. However, in inflammatory conditions, estrogen effects were partially overwhelmed by pro-inflammatory cytokines. Interestingly, estrogens were able to control production of type I interferon and therefore play dual roles during inflammatory events. In conclusion, we showed that estrogens inhibited expression of α-AChR and HLA-DR in TECs, suggesting that estrogens may alter the tolerization process and favor environment for an autoimmune response. By contrast, under inflammatory conditions, estrogen effects depend upon strength of the partner molecules with which it is confronted to.
Collapse
Affiliation(s)
- Nadine Dragin
- Inovarion, Paris, France. .,Sorbonne Universités, UPMC Univ Paris 06, Paris, France. .,INSERM U974, Paris, France.
| | - Patrice Nancy
- Department of Pathology, New York University, School of Medicine, New York, USA
| | - José Villegas
- Sorbonne Universités, UPMC Univ Paris 06, Paris, France.,INSERM U974, Paris, France.,AIM, institute of myology, Paris, France
| | | | - Rozen Le Panse
- Sorbonne Universités, UPMC Univ Paris 06, Paris, France.,INSERM U974, Paris, France.,AIM, institute of myology, Paris, France
| | - Sonia Berrih-Aknin
- Sorbonne Universités, UPMC Univ Paris 06, Paris, France.,INSERM U974, Paris, France.,AIM, institute of myology, Paris, France
| |
Collapse
|
31
|
Mazzoli M, Ariatti A, Valzania F, Kaleci S, Tondelli M, Nichelli PF, Galassi G. Factors affecting outcome in ocular myasthenia gravis. Int J Neurosci 2017. [PMID: 28625092 DOI: 10.1080/00207454.2017.1344237] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
AIM OF THE STUDY 50%-60% of patients with ocular myasthenia gravis (OMG) progress to generalized myasthenia gravis (GMG) within two years. The aim of our study was to explore factors affecting prognosis of OMG and to test the predictive role of several independent clinical variables. MATERIALS AND METHODS We reviewed a cohort of 168 Caucasian patients followed from September 2000 to January 2016. Several independent variables were considered as prognostic factors: gender, age of onset, results on electrophysiological tests, presence and level of antibodies against acetylcholine receptors (AChR Abs), treatments, thymic abnormalities. The primary outcome was the progression to GMG and/or the presence of bulbar symptoms. Secondary outcomes were either achievement of sustained minimal manifestation status or worsening in ocular quantitative MG subscore (O-QMGS) or worsening in total QMG score (T-QMGS), assessed by Myasthenia Gravis Foundation of America (MGFA) quantitative scores. Changes in mental and physical subscores of health-related quality of life (HRQoL) were assessed with SF-36 questionnaire. Variance analysis was used to interpret the differences between AChR Ab titers at different times of follow up among the generalized and non-generalized patients. RESULTS Conversion to GMG occurred in 18.4% of patients; it was significantly associated with sex, later onset of disease and anti-AChR Ab positivity. Antibody titer above the mean value of 25.8 pmol/mL showed no significant effect on generalization. Sex and late onset of disease significantly affected T-QMGS worsening. None of the other independent variables significantly affected O-QMGS and HRQoL. CONCLUSIONS Sex, later onset and anti-AChR Ab positivity were significantly associated with clinical worsening.
Collapse
Affiliation(s)
- Marco Mazzoli
- a Department of Biomedical, Metabolic and Neural Sciences , University Hospital , Modena , Italy
| | - Alessandra Ariatti
- a Department of Biomedical, Metabolic and Neural Sciences , University Hospital , Modena , Italy
| | - Franco Valzania
- a Department of Biomedical, Metabolic and Neural Sciences , University Hospital , Modena , Italy
| | - Shaniko Kaleci
- b Department of Diagnostic Clinical Medicine and Public Health , University of Modena and Reggio Emilia , Modena , Italy
| | - Manuela Tondelli
- a Department of Biomedical, Metabolic and Neural Sciences , University Hospital , Modena , Italy
| | - Paolo F Nichelli
- a Department of Biomedical, Metabolic and Neural Sciences , University Hospital , Modena , Italy
| | - Giuliana Galassi
- a Department of Biomedical, Metabolic and Neural Sciences , University Hospital , Modena , Italy
| |
Collapse
|
32
|
Association of early onset myasthenia gravis in Newfoundland dogs with the canine major histocompatibility complex class I. Neuromuscul Disord 2017; 27:409-416. [DOI: 10.1016/j.nmd.2017.01.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 01/08/2017] [Accepted: 01/27/2017] [Indexed: 12/19/2022]
|
33
|
Laffont S, Seillet C, Guéry JC. Estrogen Receptor-Dependent Regulation of Dendritic Cell Development and Function. Front Immunol 2017; 8:108. [PMID: 28239379 PMCID: PMC5300975 DOI: 10.3389/fimmu.2017.00108] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 01/23/2017] [Indexed: 12/23/2022] Open
Abstract
Autoimmunity, infectious diseases and cancer affect women and men differently. Because they tend to develop more vigorous adaptive immune responses than men, women are less susceptible to some infectious diseases but also at higher risk of autoimmunity. The regulation of immune responses by sex-dependent factors probably involves several non-redundant mechanisms. A privileged area of study, however, concerns the role of sex steroid hormones in the biology of innate immune cells, especially dendritic cells (DCs). In recent years, our understanding of the lineage origin of DC populations has expanded, and the lineage-committing transcription factors shaping peripheral DC subsets have been identified. Both progenitor cells and mature DC subsets express estrogen receptors (ERs), which are ligand-dependent transcription factors. This suggests that estrogens may contribute to the reported sex differences in immunity by regulating DC biology. Here, we review the recent literature and highlight evidence that estrogen-dependent activation of ERα regulates the development or the functional responses of particular DC subsets. The in vitro model of GM-CSF-induced DC differentiation shows that CD11c+ CD11bint Ly6cneg cells depend on ERα activation by estrogen for their development, and for the acquisition of competence to activate naive CD4+ T lymphocytes and mount a robust pro-inflammatory cytokine response to CD40 stimulation. In this model, estrogen signaling in conjunction with GM-CSF is necessary to promote early interferon regulatory factor (Irf)-4 expression in macrophage-DC progenitors and their subsequent differentiation into IRF-4hi CD11c+ CD11bint Ly6cneg cells, closely related to the cDC2 subset. The Flt3L-induced model of DC differentiation in turn shows that ERα signaling promotes the development of conventional DC (cDC) and plasmacytoid DC (pDC) with higher capability of pro-inflammatory cytokine production in response to TLR stimulation. Likewise, cell-intrinsic ER signaling positively regulates the TLR-driven production of type I interferons (IFNs) in mouse pDCs in vivo. This effect of estrogens likely contributes to the greater proficiency of women's pDCs than men's as regards the production of type I IFNs elicited by TLR7 ligands. In summary, evidence is emerging in support of the notion that estrogen signaling regulates important aspects of cDC and pDC development and/or effector functions, in both mice and humans.
Collapse
Affiliation(s)
- Sophie Laffont
- Centre de Physiopathologie de Toulouse Purpan (CPTP), Université de Toulouse, INSERM, CNRS, UPS , Toulouse , France
| | - Cyril Seillet
- Division of Molecular Immunology, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia; Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Jean-Charles Guéry
- Centre de Physiopathologie de Toulouse Purpan (CPTP), Université de Toulouse, INSERM, CNRS, UPS , Toulouse , France
| |
Collapse
|
34
|
Boldingh MI, Maniaol AH, Brunborg C, Weedon-Fekjær H, Verschuuren JJGM, Tallaksen CME. Increased risk for clinical onset of myasthenia gravis during the postpartum period. Neurology 2016; 87:2139-2145. [PMID: 27770065 DOI: 10.1212/wnl.0000000000003339] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 08/03/2016] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE To study the risk of clinical onset of myasthenia gravis (MG) in pregnancy and during the first 6 months postpartum because an association between pregnancy or the postpartum period and the onset of autoimmune MG is widely assumed but not proven. METHODS The design was a cross-sectional population-based cohort study of 2 MG cohorts (Norway and the Netherlands) with 1,038 healthy controls from Norway. Data were obtained on 246 women with MG (age at onset 15-45 years). Data on pregnancy, hormonal factors, and clinical symptoms were collected by a previously validated environmental MG questionnaire. Relative risk of MG onset before, during, and after pregnancy was calculated by multinomial logistic regression for Norwegian women reaching 45 years of age, adjusted for the observed distribution of person-years in the corresponding control group. RESULTS Of the included women with MG, 13 (11.5%) of the Dutch and 24 (18.0%) of the Norwegian patients had their first myasthenia symptoms during the pregnancy or postpartum period. The postpartum period was confirmed to be significantly associated with the onset of symptoms of MG in Norwegian women with MG (relative risk 5.5, 95% confidence interval 2.6-11.6). The risk was highest after the first childbirth. CONCLUSIONS Women have a high-risk period for the onset of clinical symptoms of MG in the postpartum period, in particular after the first childbirth. Future studies should aim at elucidating the role of the hormonal-immunological-genetic interaction in the pathogenesis of MG.
Collapse
Affiliation(s)
- Marion I Boldingh
- From the Department of Neurology (M.I.B., A.H.M., C.M.E.T.) and Oslo Centre for Biostatistics and Epidemiology (C.B., H.W.-F.), Research Support Services, Oslo University Hospital, Norway; Department of Neurology (J.J.G.M.V.), Leiden University Medical Centre, the Netherlands; and Institute of Clinical Medicine (M.I.B., C.M.E.T.), University of Oslo, Norway.
| | - Angelina H Maniaol
- From the Department of Neurology (M.I.B., A.H.M., C.M.E.T.) and Oslo Centre for Biostatistics and Epidemiology (C.B., H.W.-F.), Research Support Services, Oslo University Hospital, Norway; Department of Neurology (J.J.G.M.V.), Leiden University Medical Centre, the Netherlands; and Institute of Clinical Medicine (M.I.B., C.M.E.T.), University of Oslo, Norway
| | - Cathrine Brunborg
- From the Department of Neurology (M.I.B., A.H.M., C.M.E.T.) and Oslo Centre for Biostatistics and Epidemiology (C.B., H.W.-F.), Research Support Services, Oslo University Hospital, Norway; Department of Neurology (J.J.G.M.V.), Leiden University Medical Centre, the Netherlands; and Institute of Clinical Medicine (M.I.B., C.M.E.T.), University of Oslo, Norway
| | - Harald Weedon-Fekjær
- From the Department of Neurology (M.I.B., A.H.M., C.M.E.T.) and Oslo Centre for Biostatistics and Epidemiology (C.B., H.W.-F.), Research Support Services, Oslo University Hospital, Norway; Department of Neurology (J.J.G.M.V.), Leiden University Medical Centre, the Netherlands; and Institute of Clinical Medicine (M.I.B., C.M.E.T.), University of Oslo, Norway
| | - Jan J G M Verschuuren
- From the Department of Neurology (M.I.B., A.H.M., C.M.E.T.) and Oslo Centre for Biostatistics and Epidemiology (C.B., H.W.-F.), Research Support Services, Oslo University Hospital, Norway; Department of Neurology (J.J.G.M.V.), Leiden University Medical Centre, the Netherlands; and Institute of Clinical Medicine (M.I.B., C.M.E.T.), University of Oslo, Norway
| | - Chantal M E Tallaksen
- From the Department of Neurology (M.I.B., A.H.M., C.M.E.T.) and Oslo Centre for Biostatistics and Epidemiology (C.B., H.W.-F.), Research Support Services, Oslo University Hospital, Norway; Department of Neurology (J.J.G.M.V.), Leiden University Medical Centre, the Netherlands; and Institute of Clinical Medicine (M.I.B., C.M.E.T.), University of Oslo, Norway
| |
Collapse
|
35
|
Imbalance of circulating CD4+CXCR5+FOXP3+ Tfr-like cells and CD4+CXCR5+FOXP3− Tfh-like cells in myasthenia gravis. Neurosci Lett 2016; 630:176-182. [DOI: 10.1016/j.neulet.2016.07.049] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 07/24/2016] [Accepted: 07/25/2016] [Indexed: 02/07/2023]
|
36
|
Lee HS, Lee HS, Shin HY, Choi YC, Kim SM. The Epidemiology of Myasthenia Gravis in Korea. Yonsei Med J 2016; 57:419-25. [PMID: 26847295 PMCID: PMC4740535 DOI: 10.3349/ymj.2016.57.2.419] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 11/02/2015] [Accepted: 11/20/2015] [Indexed: 11/27/2022] Open
Abstract
PURPOSE An epidemiological study of myasthenia gravis (MG) has not been performed in Korea. The purpose of this study was to estimate the prevalence and incidence of MG in Korea. MATERIALS AND METHODS Health Insurance Review and Assessment (HIRA) data from 2010 to 2014 were searched for MG codes as defined by the International Classification of Diseases, 10th revision. After identifying MG cases, we estimated the prevalence and annual incidence of MG based on the HIRA database and Korean population data. RESULTS During the study period, 10138 MG cases were identified. The prevalence of MG was 10.42 cases per 100000 people in 2010 and this increased every year to 12.99 cases per 100000 people in 2014. The average incidence of MG between 2011 and 2014 was 0.69 cases per 100000 person-years. The prevalence and incidence were higher in the older (≥ 50 years) age group than in the younger (<50 years) age group [prevalence: 9.26 vs. 19.24 per 100000, relative risk 2.077, 95% confidence interval (CI) 1.976-2.183, p<0.001; incidence: 0.47 vs. 1.18 per 100000, relative risk 2.490, 95% CI 2.006-3.091, p<0.001]. CONCLUSION This study was the first nationwide population-based epidemiological study of MG in Korea. The prevalence and incidence of MG were consistent with those of previous studies. We found an increase in the prevalence of MG and a predominance of elderly MG patients.
Collapse
Affiliation(s)
- Hyung Seok Lee
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| | - Hye Sun Lee
- Department of Biostatistics, Yonsei University College of Medicine, Seoul, Korea
| | - Ha Young Shin
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| | - Young Chul Choi
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| | - Seung Min Kim
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
37
|
Engelmann F, Rivera A, Park B, Messerle-Forbes M, Jensen JT, Messaoudi I. Impact of Estrogen Therapy on Lymphocyte Homeostasis and the Response to Seasonal Influenza Vaccine in Post-Menopausal Women. PLoS One 2016; 11:e0149045. [PMID: 26859566 PMCID: PMC4747494 DOI: 10.1371/journal.pone.0149045] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 01/25/2016] [Indexed: 01/01/2023] Open
Abstract
It is widely recognized that changes in levels of ovarian steroids modulate severity of autoimmune disease and immune function in young adult women. These observations suggest that the loss of ovarian steroids associated with menopause could affect the age-related decline in immune function, known as immune senescence. Therefore, in this study, we determined the impact of menopause and estrogen therapy (ET) on lymphocyte subset frequency as well as the immune response to seasonal influenza vaccine in three different groups: 1) young adult women (regular menstrual cycles, not on hormonal contraception); 2) post-menopausal (at least 2 years) women who are not receiving any form of hormone therapy (HT) and 3) post-menopausal hysterectomized women receiving ET. Although the numbers of circulating CD4 and CD20 B cells were reduced in the post-menopausal group receiving ET, we also detected a better preservation of naïve B cells, decreased CD4 T cell inflammatory cytokine production, and slightly lower circulating levels of the pro-inflammatory cytokine IL-6. Following vaccination, young adult women generated more robust antibody and T cell responses than both post-menopausal groups. Despite similar vaccine responses between the two post-menopausal groups, we observed a direct correlation between plasma 17β estradiol (E2) levels and fold increase in IgG titers within the ET group. These findings suggest that ET affects immune homeostasis and that higher plasma E2 levels may enhance humoral responses in post-menopausal women.
Collapse
Affiliation(s)
- Flora Engelmann
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California, United States of America
| | - Andrea Rivera
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California, United States of America
| | - Byung Park
- Division of Biostatistics, Department of Public Health and Preventive Medicine, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Marci Messerle-Forbes
- Department of Obstetrics and Gynecology, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Jeffrey T. Jensen
- Department of Obstetrics and Gynecology, Oregon Health and Science University, Portland, Oregon, United States of America
- Division of Reproductive Sciences, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
| | - Ilhem Messaoudi
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California, United States of America
- Division of Reproductive Sciences, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
- * E-mail:
| |
Collapse
|
38
|
Guidelines for standard preclinical experiments in the mouse model of myasthenia gravis induced by acetylcholine receptor immunization. Exp Neurol 2015; 270:11-7. [DOI: 10.1016/j.expneurol.2015.02.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 01/13/2015] [Accepted: 02/04/2015] [Indexed: 01/25/2023]
|
39
|
Kovats S. Estrogen receptors regulate innate immune cells and signaling pathways. Cell Immunol 2015; 294:63-9. [PMID: 25682174 PMCID: PMC4380804 DOI: 10.1016/j.cellimm.2015.01.018] [Citation(s) in RCA: 657] [Impact Index Per Article: 73.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 01/26/2015] [Accepted: 01/29/2015] [Indexed: 02/07/2023]
Abstract
Humans show strong sex differences in immunity to infection and autoimmunity,
suggesting sex hormones modulate immune responses. Indeed, receptors for estrogens (ER)
regulate cells and pathways in the innate and adaptive immune system, as well as immune
cell development. ERs are ligand-dependent transcription factors that mediate long-range
chromatin interactions and form complexes at gene regulatory elements, thus promoting
epigenetic changes and transcription. ERs also participate in membrane-initiated steroid
signaling to generate rapid responses. Estradiol and ER activity show profound dose- and
context-dependent effects on innate immune signaling pathways and myeloid cell
development. While estradiol most often promotes the production of type I interferon, innate pathways
leading to pro-inflammatory cytokine production may be enhanced or dampened by ER
activity. Regulation of innate immune cells and signaling by ERs may contribute to the
reported sex differences in innate immune pathways. Here we review the recent literature
and highlight several molecular mechanisms by which ERs regulate the development or
functional responses of innate immune cells.
Collapse
Affiliation(s)
- Susan Kovats
- Arthritis & Clinical Immunology Research Program, Oklahoma Medical Research Foundation, United States.
| |
Collapse
|
40
|
Estrogen modulates β2-adrenoceptor-induced cell-mediated and inflammatory immune responses through ER-α involving distinct intracellular signaling pathways, antioxidant enzymes, and nitric oxide. Cell Immunol 2014; 292:1-8. [DOI: 10.1016/j.cellimm.2014.08.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 08/06/2014] [Indexed: 11/16/2022]
|
41
|
Gilbert EL, Ryan MJ. Estrogen in cardiovascular disease during systemic lupus erythematosus. Clin Ther 2014; 36:1901-1912. [PMID: 25194860 DOI: 10.1016/j.clinthera.2014.07.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 07/25/2014] [Accepted: 07/28/2014] [Indexed: 01/11/2023]
Abstract
PURPOSE Systemic lupus erythematosus (SLE) is a chronic inflammatory autoimmune disease that disproportionately affects women during their childbearing years. Cardiovascular disease is the leading cause of mortality in this patient population at an age when women often have low cardiovascular risk. Hypertension is a major cardiovascular disease risk factor, and its prevalence is markedly increased in women with SLE. Estrogen has traditionally been implicated in SLE disease progression because of the prevalence of the disease in women; however, its role in cardiovascular risk factors such as hypertension is unclear. The objective of this review is to discuss evidence for the role of estrogen in both human and murine SLE with emphasis on the effect of estrogen on cardiovascular risk factors, including hypertension. METHODS PubMed was used to search for articles with terms related to estradiol and SLE. The references of retrieved publications were also reviewed. FINDINGS The potential permissive role of estrogen in SLE development is supported by studies from experimental animal models of lupus in which early removal of estrogen or its effects leads to attenuation of SLE disease parameters, including autoantibody production and renal injury. However, data about the role of estrogens in human SLE are much less clear, with most studies not reaching firm conclusions about positive or negative outcomes after hormonal manipulations involving estrogen during SLE (ie, oral contraceptives, hormone therapy). Significant gaps in knowledge remain about the effect of estrogen on cardiovascular risk factors during SLE. Studies in women with SLE were not designed to determine the effect of estrogen or hormone therapy on blood pressure even though hypertension is highly prevalent, and risk of premature ovarian failure could necessitate use of hormone therapy in women with SLE. Recent evidence from an experimental animal model of lupus found that estrogen may protect against cardiovascular risk factors in adulthood. In addition, increasing evidence suggests that estrogen may have distinct temporal effects on cardiovascular risk factors during SLE. IMPLICATIONS Data from experimental models of lupus suggest that estrogens may have an important permissive role for developing SLE early in life. However, their role in adulthood remains unclear, particularly for the effect on cardiovascular disease and its risk factors. Additional work is needed to understand the effect of estrogens in human SLE, and preclinical studies in experimental models of SLE may contribute important mechanistic insight to further advance the field.
Collapse
Affiliation(s)
- Emily L Gilbert
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| | - Michael J Ryan
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi.
| |
Collapse
|
42
|
Genetic basis of myasthenia gravis – A comprehensive review. J Autoimmun 2014; 52:146-53. [DOI: 10.1016/j.jaut.2013.12.001] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Accepted: 12/02/2013] [Indexed: 11/24/2022]
|
43
|
Berrih-Aknin S. Myasthenia Gravis: paradox versus paradigm in autoimmunity. J Autoimmun 2014; 52:1-28. [PMID: 24934596 DOI: 10.1016/j.jaut.2014.05.001] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 05/07/2014] [Indexed: 12/12/2022]
Abstract
Myasthenia Gravis (MG) is a paradigm of organ-specific autoimmune disease (AID). It is mediated by antibodies that target the neuromuscular junction. The purpose of this review is to place MG in the general context of autoimmunity, to summarize the common mechanisms between MG and other AIDs, and to describe the specific mechanisms of MG. We have chosen the most common organ-specific AIDs to compare with MG: type 1 diabetes mellitus (T1DM), autoimmune thyroid diseases (AITD), multiple sclerosis (MS), some systemic AIDs (systemic lupus erythematous (SLE), rheumatoid arthritis (RA), Sjogren's syndrome (SS)), as well as inflammatory diseases of the gut and liver (celiac disease (CeD), Crohn's disease (CD), and primary biliary cirrhosis (PBC)). Several features are similar between all AIDs, suggesting that common pathogenic mechanisms lead to their development. In this review, we address the predisposing factors (genetic, epigenetic, hormones, vitamin D, microbiota), the triggering components (infections, drugs) and their interactions with the immune system [1,2]. The dysregulation of the immune system is detailed and includes the role of B cells, Treg cells, Th17 and cytokines. We particularly focused on the role of TNF-α and interferon type I whose role in MG is very analogous to that in several other AIDS. The implication of AIRE, a key factor in central tolerance is also discussed. Finally, if MG is a prototype of AIDS, it has a clear specificity compared to the other AIDS, by the fact that the target organ, the muscle, is not the site of immune infiltration and B cell expansion, but exclusively that of antibody-mediated pathogenic mechanisms. By contrast, the thymus in the early onset subtype frequently undergoes tissue remodeling, resulting in the development of ectopic germinal centers surrounded by high endothelial venules (HEV), as observed in the target organs of many other AIDs.
Collapse
Affiliation(s)
- Sonia Berrih-Aknin
- Sorbonne Universités, UPMC Univ Paris 06, Myology Research Center UM76, F-75013 Paris, France; INSERM U974, F-75013 Paris, France; CNRS FRE 3617, F-75013 Paris, France; Institute of Myology, F-75013 Paris, France.
| |
Collapse
|
44
|
Berrih-Aknin S, Le Panse R. Myasthenia gravis: a comprehensive review of immune dysregulation and etiological mechanisms. J Autoimmun 2014; 52:90-100. [PMID: 24389034 DOI: 10.1016/j.jaut.2013.12.011] [Citation(s) in RCA: 238] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 12/12/2013] [Indexed: 12/31/2022]
Abstract
Autoimmune myasthenia gravis (MG) is characterized by muscle weakness caused by antibodies directed against proteins of the neuromuscular junction. The main antigenic target is the acetylcholine receptor (AChR), but the muscle Specific Kinase (MuSK) and the low-density lipoprotein receptor-related protein (LRP4) are also targets. This review summarizes the clinical and biological data available for different subgroups of patients, who are classified according to antigenic target, age of onset, and observed thymic abnormalities, such as follicular hyperplasia or thymoma. Here, we analyze in detail the role of the thymus in the physiopathology of MG and propose an explanation for the development of the thymic follicular hyperplasia that is commonly observed in young female patients with anti-AChR antibodies. The influence of the pro-inflammatory environment is discussed, particularly the role of TNF-α and Th17-related cytokines, which could explain the escape of thymic T cells from regulation and the chronic inflammation in the MG thymus. Together with this immune dysregulation, active angiogenic processes and the upregulation of chemokines could promote thymic follicular hyperplasia. MG is a multifactorial disease, and we review the etiological mechanisms that could lead to its onset. Recent global genetic analyses have highlighted potential susceptibility genes. In addition, miRNAs, which play a crucial role in immune function, have been implicated in MG by recent studies. We also discuss the role of sex hormones and the influence of environmental factors, such as the viral hypothesis. This hypothesis is supported by reports that type I interferon and molecules mimicking viral infection can induce thymic changes similar to those observed in MG patients with anti-AChR antibodies.
Collapse
Affiliation(s)
- Sonia Berrih-Aknin
- INSERM U974, Paris, France; CNRS UMR 7215, Paris, France; UPMC Univ Paris 6, Paris, France; AIM, Institute of myology, Paris, France.
| | - Rozen Le Panse
- INSERM U974, Paris, France; CNRS UMR 7215, Paris, France; UPMC Univ Paris 6, Paris, France; AIM, Institute of myology, Paris, France.
| |
Collapse
|
45
|
Selective modulation of lymphoproliferation and cytokine production via intracellular signaling targets by α1- and α2-adrenoceptors and estrogen in splenocytes. Int Immunopharmacol 2013; 17:774-84. [PMID: 24055020 DOI: 10.1016/j.intimp.2013.08.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 07/26/2013] [Accepted: 08/28/2013] [Indexed: 11/21/2022]
Abstract
UNLABELLED The mechanistic implications of the presence of sympathetic noradrenergic innervation in lymphoid organs in synaptic association with lymphocytes open to the influence of hormonal fluctuations throughout reproductive age in females has not been investigated yet. OBJECTIVES The aim of the present study is to investigate the role of alpha-adrenoceptors (α-ARs) and estrogen in modulating immune responses in the spleen through intracellular signaling targets such as ERK 1/2, CREB, Akt, NF-κB. METHODS Splenocytes from young Sprague-Dawley rats were incubated with α1- and α2- AR specific agonists, phenylephrine and clonidine, without and with 17b-estradiol or specific antagonists prazosin and idazoxan to examine their effects on proliferation, cytokine production, nitric oxide production, and intracellular signaling molecules. RESULTS α1-AR stimulation inhibited lymphocyte proliferation and IFN-g production and enhanced IL-2, p-ERK and p-CREB expression. Co-stimulation using estrogen enhanced cytokine production and suppressed p-Akt expression. α1-AR blockade reversed agonist-induced IL-2 production alone. α2-AR stimulation inhibited lymphocyte proliferation, p-ERK and p-CREB expression, and increased p-NF-kB and p-Akt expression. Co-stimulation with estrogen increased IL-2 and suppressed p-CREB expression. α2-AR Idazoxan prevented IL-2 production in the absence and presence of estrogen, and reversed clonidine-induced increase in NO production and p-ERK and p-Akt expression in the presence of estrogen. CONCLUSIONS These results suggest that the cell-mediated immune responses are selectively modulated depending upon the subtypes of α-AR and further, these effects are differentially regulated in the presence of estrogen mediated through selective alteration in the intracellular signaling pathways involving ERK, CREB, Akt, and NF-κB.
Collapse
|
46
|
Yakimchuk K, Jondal M, Okret S. Estrogen receptor α and β in the normal immune system and in lymphoid malignancies. Mol Cell Endocrinol 2013; 375:121-9. [PMID: 23707618 DOI: 10.1016/j.mce.2013.05.016] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Revised: 05/14/2013] [Accepted: 05/18/2013] [Indexed: 02/07/2023]
Abstract
Estrogens regulate various normal and pathophysiological processes including cancers. Cellular signaling by estrogens is mediated by estrogen receptor α (ERα) and β (ERβ), respectively. Binding of agonists to the ERs affects gene transcription. The main endogenous estrogen, 17β-estradiol (E2), binds to both ERα and ERβ with similar affinity. However, the ligand-binding pocket of ERα and ERβ are slightly different which has allowed the development of selective ER ligands. Importantly, while estrogens via ERα stimulate proliferation, signaling via ERβ inhibits proliferation and promotes apoptosis. In both normal and cancer cells the ERs are co-expressed with ER splice variants which may modify the transcriptional activity of the wild-type receptors. Estrogens have prominent effects on immune functions and both ERα and ERβ are expressed in immune cells and lymphoid malignancies. With regard to lymphoid malignancies, most show estrogen influence as several epidemiological studies of lymphoid cancers demonstrate gender differences in incidence and prognosis with males being more affected. In line with these findings, recent results generated by us have shown that ERβ selective agonists inhibit growth and induce apoptosis in human and murine lymphomas in vivo in xenograft experiments. This suggests that ERβ selective agonists in the future may be useful in the treatment of lymphomas.
Collapse
Affiliation(s)
- Konstantin Yakimchuk
- Department of Biosciences and Nutrition, Karolinska Institutet, Novum, SE-141 83 Huddinge, Sweden
| | | | | |
Collapse
|
47
|
Priyanka HP, Krishnan HC, Singh RV, Hima L, Thyagarajan S. Estrogen modulates in vitro T cell responses in a concentration- and receptor-dependent manner: effects on intracellular molecular targets and antioxidant enzymes. Mol Immunol 2013; 56:328-39. [PMID: 23911387 DOI: 10.1016/j.molimm.2013.05.226] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 04/24/2013] [Accepted: 05/20/2013] [Indexed: 01/21/2023]
Abstract
Estrogen is a key hormone in facilitating ovulation and maintenance of pregnancy in young females and subsequent decline in its production contributes to the development of age-associated disorders such as hormone-dependent cancer, osteoporosis, and cardiovascular diseases. The mechanisms through which estrogen promotes female-specific diseases with advancing age are unclear especially, its effects on immune system which is vital for the maintenance of homeostasis and health. Although the diverse effects of estrogen on Th immunity (Th1 vs. Th2) have been characterized in several cell-types and animal models, there is no direct mechanistic study to understand its immunomodulatory actions. The purpose of this study is to investigate whether the in vitro effects of 17β-estradiol on lymphocytes from the spleen influence cell-mediated immune responses based on its concentration and type of estrogen receptors (ERs) and to assess its mechanism of action at the cellular level. Lymphocytes from the spleens of young Sprague-Dawley rats were isolated and incubated with various concentrations of 17β-estradiol (10(-6)-10(-14)M) and specific ERα- and β-agonists (10(-6)M, 10(-8)M and 10(-10)M) without or with concanavalin A (Con A) to measure T lymphocyte proliferation, IFN-γ and IL-2 production, p-ERK 1/2, p-CREB, and p-Akt, activities of antioxidant enzymes[superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPx)], and nitric oxide (NO) production. The specificity of ER-mediated actions in lymphocytes was examined by coincubation with nonspecific ER antagonists ICI(182,780) or tamoxifen. Lower concentrations of 17β-estradiol enhanced proliferation of T lymphocytes and IFN-γ production without or with Con A stimulation but had no effect on IL-2 production. ERα and ERβ agonists induced an increase in T cell proliferation and IFN-γ production and these effects were inhibited by tamoxifen. ERβ agonist alone enhanced IL-2 production by the lymphocytes. Coincubation with 17β-estradiol and ERα- and β-agonists augmented p-ERK 1/2, p-CREB, and p-Akt expression in the lymphocytes and tamoxifen reversed the ER agonist-induced effects on these molecular targets. Estrogen increased the activities of SOD, CAT, and GPx in both non-stimulated and Con A-stimulated splenocytes in a concentration-dependent manner. Both ERα- and β-agonists enhanced CAT and GPx activity while ERα-agonist decreased SOD activity and ERβ-agonist increased SOD activity. The effects of ER agonists on the antioxidant enzymes were reversed by ICI(182,780). Coincubation of lower doses of 17β-estradiol with Con A and both ER agonists enhanced NO production while higher dose of estrogen with Con A and ERα agonist suppressed its production and these effects were reversed by tamoxifen. Taken together, these results suggest that the effects of estrogen on the cell-mediated immune responses are dependent upon its concentrations and mediated through specific estrogen receptors involving intracellular signaling pathways and antioxidant enzymes.
Collapse
Affiliation(s)
- Hannah P Priyanka
- Integrative Medicine Laboratory, Department of Biotechnology, School of Bioengineering, SRM University, Kattankulathur 603203 Tamil Nadu, India
| | | | | | | | | |
Collapse
|
48
|
Cavalcante P, Cufi P, Mantegazza R, Berrih-Aknin S, Bernasconi P, Le Panse R. Etiology of myasthenia gravis: Innate immunity signature in pathological thymus. Autoimmun Rev 2013; 12:863-74. [DOI: 10.1016/j.autrev.2013.03.010] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/12/2013] [Indexed: 01/09/2023]
|
49
|
Seillet C, Rouquié N, Foulon E, Douin-Echinard V, Krust A, Chambon P, Arnal JF, Guéry JC, Laffont S. Estradiol promotes functional responses in inflammatory and steady-state dendritic cells through differential requirement for activation function-1 of estrogen receptor α. THE JOURNAL OF IMMUNOLOGY 2013; 190:5459-70. [PMID: 23626011 DOI: 10.4049/jimmunol.1203312] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
17β-Estradiol (E2) has been shown to regulate GM-CSF- or Flt3 ligand-driven dendritic cell (DC) development through estrogen receptor (ER) α signaling in myeloid progenitors. ERα regulates transcription of target genes through two distinct activation functions (AFs), AF-1 and AF-2, whose respective involvement varies in a cell type- or tissue-specific manner. In this study, we investigated the role of ERα AFs in the development and effector functions of inflammatory DCs, steady-state conventional DCs, and plasmacytoid DCs (pDC), using mouse lacking either AF-1 or AF-2. In agreement with previous works, we showed that E2 fostered the differentiation and effector functions of inflammatory DCs through ERα-dependent upregulation of IFN regulatory factor (IRF)-4 in GM-CSF-stimulated myeloid progenitors. Interestingly, whereas AF-1 was required for early IRF-4 upregulation in DC precursors, it was dispensable to enhance IRF-4 expression in differentiated DCs to a level compatible with the development of the more functional Ly6C(-) CD11b(+) DC subset. Presence of E2 had no effect on progenitors from either knock-in mice with 7-aa deletion in helix 12 of ERα, lacking AF-2, or ERα(-/-) mice. By contrast, in Flt3 ligand-driven DC differentiation, activation of AF-1 domain was required to promote the development of more functionally competent conventional DCs and pDCs. Moreover, lack of ERα AF-1 blunted the TLR7-mediated IFN-α response of female pDCs in vivo. Thus, our study demonstrates that ERα uses AF-1 differently in steady-state and inflammatory DC lineages to regulate their innate functions, suggesting that selective ER modulators could be used to target specific DC subsets.
Collapse
|
50
|
Nguyen TG, Ward CM, Morris JM. To B or not to B cells-mediate a healthy start to life. Clin Exp Immunol 2013; 171:124-34. [PMID: 23286939 PMCID: PMC3573283 DOI: 10.1111/cei.12001] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/02/2012] [Indexed: 01/19/2023] Open
Abstract
Maternal immune responses during pregnancy are critical in programming the future health of a newborn. The maternal immune system is required to accommodate fetal immune tolerance as well as to provide a protective defence against infections for the immunocompromised mother and her baby during gestation and lactation. Natural immunity and antibody production by maternal B cells play a significant role in providing such immunoprotection. However, aberrations in the B cell compartment as a consequence of maternal autoimmunity can pose serious risks to both the mother and her baby. Despite their potential implication in shaping pregnancy outcomes, the role of B cells in human pregnancy has been poorly studied. This review focuses on the role of B cells and the implications of B cell depletion therapy in pregnancy. It highlights the evidence of an association between aberrant B cell compartment and obstetric conditions. It also alludes to the potential mechanisms that amplify these B cell aberrances and thereby contribute to exacerbation of some maternal autoimmune conditions and poor neonatal outcomes. Clinical and experimental evidence suggests strongly that maternal autoantibodies contribute directly to the pathologies of obstetric and neonatal conditions that have significant implications for the lifelong health of a newborn. The evidence for clinical benefit and safety of B cell depletion therapies in pregnancy is reviewed, and an argument is mounted for further clinical evaluation of B cell-targeted therapies in high-risk pregnancy, with an emphasis on improving neonatal outcomes and prevention of neonatal conditions such as congenital heart block and fetal/neonatal alloimmune thrombocytopenia.
Collapse
Affiliation(s)
- T G Nguyen
- Perinatal Research, Kolling Institute of Medical Research, North Shore Hospital, Sydney, Australia.
| | | | | |
Collapse
|