1
|
Fettig NM, Pu A, Osborne LC, Gommerman JL. The influence of aging and the microbiome in multiple sclerosis and other neurologic diseases. Immunol Rev 2024; 325:166-189. [PMID: 38890777 DOI: 10.1111/imr.13361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
The human gut microbiome is well-recognized as a key player in maintaining health. However, it is a dynamic entity that changes across the lifespan. How the microbial changes that occur in later decades of life shape host health or impact age-associated inflammatory neurological diseases such as multiple sclerosis (MS) is still unclear. Current understanding of the aging gut microbiome is largely limited to cross-sectional observational studies. Moreover, studies in humans are limited by confounding host-intrinsic and extrinsic factors that are not easily disentangled from aging. This review provides a comprehensive summary of existing literature on the aging gut microbiome and its known relationships with neurological diseases, with a specific focus on MS. We will also discuss preclinical animal models and human studies that shed light on the complex microbiota-host interactions that have the potential to influence disease pathology and progression in aging individuals. Lastly, we propose potential avenues of investigation to deconvolute features of an aging microbiota that contribute to disease, or alternatively promote health in advanced age.
Collapse
Affiliation(s)
- Naomi M Fettig
- Department of Microbiology & Immunology, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Annie Pu
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Lisa C Osborne
- Department of Microbiology & Immunology, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | | |
Collapse
|
2
|
Kalakh S, Mouihate A. The Effects of Neuroactive Steroids on Myelin in Health and Disease. Med Princ Pract 2024; 33:198-214. [PMID: 38350432 PMCID: PMC11175611 DOI: 10.1159/000537794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 02/12/2024] [Indexed: 02/15/2024] Open
Abstract
Myelin plays a pivotal role in the efficient transmission of nerve impulses. Disruptions in myelin integrity are associated with numerous neurological disorders, including multiple sclerosis. In the central nervous system (CNS), myelin is formed by oligodendrocytes. Remyelination refers to the re-formation of the damaged myelin sheath by newly formed oligodendrocytes. Steroids have gained attention for their potential modulatory effects on myelin in both health and disease. Steroids are traditionally associated with endocrine functions, but their local synthesis within the nervous system has generated significant interest. The term "neuroactive steroids" refers to steroids that can act on cells of the nervous system. In the healthy state, neuroactive steroids promote myelin formation, maintenance, and repair by enhancing oligodendrocyte differentiation and maturation. In pathological conditions, such as demyelination injury, multiple neuroactive steroids have shown promise in promoting remyelination. Understanding the effects of neuroactive steroids on myelin could lead to novel therapeutic approaches for demyelinating diseases and neurodegenerative disorders. This review highlights the potential therapeutic significance of neuroactive steroids in myelin-related health and diseases. We review the synthesis of steroids by neurons and glial cells and discuss the roles of neuroactive steroids on myelin structure and function in health and disease. We emphasize the potential promyelinating effects of the varying levels of neuroactive steroids during different female physiological states such as the menstrual cycle, pregnancy, lactation, and postmenopause.
Collapse
Affiliation(s)
- Samah Kalakh
- Department of Physiology, College of Medicine, Kuwait University, Kuwait City, Kuwait
- School of Engineering and Computing, American International University, Kuwait City, Kuwait
| | - Abdeslam Mouihate
- Department of Physiology, College of Medicine, Kuwait University, Kuwait City, Kuwait
| |
Collapse
|
3
|
Zahaf A, Kassoussi A, Hutteau-Hamel T, Mellouk A, Marie C, Zoupi L, Tsouki F, Mattern C, Bobé P, Schumacher M, Williams A, Parras C, Traiffort E. Androgens show sex-dependent differences in myelination in immune and non-immune murine models of CNS demyelination. Nat Commun 2023; 14:1592. [PMID: 36949062 PMCID: PMC10033728 DOI: 10.1038/s41467-023-36846-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 02/20/2023] [Indexed: 03/24/2023] Open
Abstract
Neuroprotective, anti-inflammatory, and remyelinating properties of androgens are well-characterized in demyelinated male mice and men suffering from multiple sclerosis. However, androgen effects mediated by the androgen receptor (AR), have been only poorly studied in females who make low androgen levels. Here, we show a predominant microglial AR expression in demyelinated lesions from female mice and women with multiple sclerosis, but virtually undetectable AR expression in lesions from male animals and men with multiple sclerosis. In female mice, androgens and estrogens act in a synergistic way while androgens drive microglia response towards regeneration. Transcriptomic comparisons of demyelinated mouse spinal cords indicate that, regardless of the sex, androgens up-regulate genes related to neuronal function integrity and myelin production. Depending on the sex, androgens down-regulate genes related to the immune system in females and lipid catabolism in males. Thus, androgens are required for proper myelin regeneration in females and therapeutic approaches of demyelinating diseases need to consider male-female differences.
Collapse
Affiliation(s)
- Amina Zahaf
- U1195 Inserm, Paris-Saclay University, Kremlin-Bicêtre, France
| | | | | | - Amine Mellouk
- UMR996 Inserm, Paris-Saclay University, Clamart, France
| | | | - Lida Zoupi
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh BioQuarter, Edinburgh, UK
| | - Foteini Tsouki
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh BioQuarter, Edinburgh, UK
| | | | - Pierre Bobé
- UMR996 Inserm, Paris-Saclay University, Clamart, France
| | | | - Anna Williams
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh BioQuarter, Edinburgh, UK
| | - Carlos Parras
- Paris Brain Institute, Sorbonne University, Paris, France
| | | |
Collapse
|
4
|
Petrušić M, Stojić-Vukanić Z, Pilipović I, Kosec D, Prijić I, Leposavić G. Thymic changes as a contributing factor in the increased susceptibility of old Albino Oxford rats to EAE development. Exp Gerontol 2023; 171:112009. [PMID: 36334894 DOI: 10.1016/j.exger.2022.112009] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/26/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022]
Abstract
The study was aimed to examine putative contribution of thymic involution to ageing-associated increase in susceptibility of Albino Oxford (AO) rats to the development of clinical EAE, and vice versa influence of the disease on the progression of thymic involution. To this end we examined (i) the parameters of thymocyte negative selection efficacy, the thymic generation of CD4+CD25+Foxp3+ T regulatory cells (Tregs) and thymic capacity to instruct/predetermine IL-17-producing T-cell differentiation, and thymopietic efficacy-associated accumulation of "inflammescent" cytotoxic CD28- T cells in the periphery, and (ii) the key underlying mechanisms in young and old non-immunised AO rats and their counterparts immunised for EAE (on the 16th day post-immunisation when the disease in old rats reached the plateau) using flow cytometry analysis and/or RT-qPCR. It was found that thymic involution impairs: (i) the efficacy of negative selection (by affecting thymocyte expression of CD90, negative regulator of selection threshold and the expression of thymic stromal cell integrity factors) and (ii) Treg generation (by diminishing expression of cytokines supporting their differentiation/maturation). Additionally, the results suggest that thymic involution facilitates CD8+ T-cell differentiation into IL-17-producing cells (previously linked to the development of clinical EAE in old AO rats). Furthermore, they confirmed that ageing-related decrease in thymic T-cell output (as indicated by diminished frequency of recent thymic emigrants in peripheral blood) resulted in the accumulation of CD28- T cells in peripheral blood and, upon immunisation, in the target organ. On the other hand, the development of EAE (most likely by increasing circulatory levels of proinflammatory cytokines) contributed to the decline in thymic output of T cells, including Tregs, and thereby to the progression/maintenance of clinical EAE. Thus, in AO rats thymic involution via multi-layered mechanisms may favour the development of clinically manifested autoimmunity, which, in turn, precipitates the thymus atrophy.
Collapse
Affiliation(s)
- Marija Petrušić
- Department of Pathobiology, University of Belgrade, Faculty of Pharmacy, 450 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Zorica Stojić-Vukanić
- Department of Microbiology and Immunology, University of Belgrade, Faculty of Pharmacy, 450 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Ivan Pilipović
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", 458 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Duško Kosec
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", 458 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Ivana Prijić
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", 458 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Gordana Leposavić
- Department of Pathobiology, University of Belgrade, Faculty of Pharmacy, 450 Vojvode Stepe, 11221 Belgrade, Serbia.
| |
Collapse
|
5
|
Ketchem JM, Bowman EJ, Isales CM. Male sex hormones, aging, and inflammation. Biogerontology 2023; 24:1-25. [PMID: 36596999 PMCID: PMC9810526 DOI: 10.1007/s10522-022-10002-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 11/15/2022] [Indexed: 01/05/2023]
Abstract
Adequate levels of androgens (eugonadism), and specifically testosterone, are vital compounds for male quality of life, longevity, and positive health outcomes. Testosterone exerts its effects by binding to the androgen receptor, which is expressed in numerous tissues throughout the body. Significant research has been conducted on the impact of this steroid hormone on skeletal, muscle and adipose tissues and on the cardiovascular, immune, and nervous systems. Testosterone levels have also been studied in relation to the impact of diseases, aging, nutrition and the environment on its circulating levels. Conversely, the impact of testosterone on health has also been evaluated with respect to its cardiac and vascular protective effects, body composition, autoimmunity and all-cause mortality. The male aging process results in decreasing testosterone levels over time. The exact mechanisms and impact of these changes in testosterone levels with age on health- and life-span are still not completely clear. Further research is needed to determine the optimal testosterone and androgen levels to protect from chronic age-related conditions such as frailty and osteoporosis.
Collapse
Affiliation(s)
- Justin M. Ketchem
- grid.410427.40000 0001 2284 9329Medical College of Georgia at Augusta University, Augusta, GA 30912 USA
| | | | - Carlos M. Isales
- grid.410427.40000 0001 2284 9329Departments of Medicine, Neuroscience and Regenerative Medicine, Augusta University, Augusta, GA 30912 USA
| |
Collapse
|
6
|
McCombe PA, Greer JM. Effects of biological sex and pregnancy in experimental autoimmune encephalomyelitis: It's complicated. Front Immunol 2022; 13:1059833. [PMID: 36518769 PMCID: PMC9742606 DOI: 10.3389/fimmu.2022.1059833] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 11/03/2022] [Indexed: 11/29/2022] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE) can be induced in many animal strains by inoculation with central nervous system antigens and adjuvant or by the passive transfer of lymphocytes reactive with these antigens and is widely used as an animal model for multiple sclerosis (MS). There are reports that female sex and pregnancy affect EAE. Here we review the effects of biological sex and the effects of pregnancy on the clinical features (including disease susceptibility) and pathophysiology of EAE. We also review reports of the possible mechanisms underlying these differences. These include sex-related differences in the immune system and in the central nervous system, the effects of hormones and the sex chromosomes and molecules unique to pregnancy. We also review sex differences in the response to factors that can modify the course of EAE. Our conclusion is that the effects of biological sex in EAE vary amongst animal models and should not be widely extrapolated. In EAE, it is therefore essential that studies looking at the effects of biological sex or pregnancy give full information about the model that is used (i.e. animal strain, sex, the inducing antigen, timing of EAE induction in relation to pregnancy, etc.). In addition, it would be preferable if more than one EAE model were used, to show if any observed effects are generalizable. This is clearly a field that requires further work. However, understanding of the mechanisms of sex differences could lead to greater understanding of EAE, and suggest possible therapies for MS.
Collapse
Affiliation(s)
| | - Judith M. Greer
- UQ Centre for Clinical Research, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
7
|
Li K, Li M, Wen L, Wang Q, Ding X, Wang J. Clinical Presentation and Outcomes of Acute Disseminated Encephalomyelitis in Adults Worldwide: Systematic Review and Meta-Analysis. Front Immunol 2022; 13:870867. [PMID: 35757742 PMCID: PMC9218070 DOI: 10.3389/fimmu.2022.870867] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 05/03/2022] [Indexed: 02/05/2023] Open
Abstract
Background Acute disseminated encephalomyelitis (ADEM) is a rare demyelinating disorder that is often misdiagnosed. To improve early diagnosis, we performed a systematic review and meta-analysis of clinical features, outcomes for ADEM in adults. Methods The PubMed, Embase, Web of Science and Cochrane Library databases were searched for studies reporting the clinical features of adults with ADEM between January 1990 and May 2021. A random-effects meta-analysis model was used to pool data on clinical features and functional outcomes. Results Twelve studies examining 437 adults with ADEM met the inclusion criteria. Overall, the clinical features and diagnostic findings observed in more than two-thirds of the patients were white matter lesions [87.1%, 95% confidence interval (CI)=75-95.6], polyfocal onset (80.5%, 95% CI=50.5-98.9) and pyramidal signs (68.7%, 95% CI =40.0-91.9). The mortality rate was 7.8% (95% CI = 3.3–13.5), and the risk of residual deficits was 47.5% (95% CI = 31.8–63.4). Conclusions Adults with ADEM had worse outcomes than children. Clinicians should maintain high clinical suspicion for patients presenting with certain clinical features and diagnostic findings.
Collapse
Affiliation(s)
- Kunyi Li
- Department of Neurology, the Second People's Hospital of Chengdu, Chengdu, China
| | - Maolin Li
- Department of Neurology, People's Hospital of Deyang City, Deyang, China
| | - Lan Wen
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Qiancheng Wang
- Department of Neurology, Chongqing North-Kuanren General Hospital, ChongQing, China
| | - Xin Ding
- Department of Neurology, the Second People's Hospital of Chengdu, Chengdu, China
| | - Jian Wang
- Department of Neurology, the Second People's Hospital of Chengdu, Chengdu, China
| |
Collapse
|
8
|
Atkinson JR, Jerome AD, Sas AR, Munie A, Wang C, Ma A, Arnold WD, Segal BM. Biological aging of CNS-resident cells alters the clinical course and immunopathology of autoimmune demyelinating disease. JCI Insight 2022; 7:158153. [PMID: 35511417 PMCID: PMC9309055 DOI: 10.1172/jci.insight.158153] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 05/04/2022] [Indexed: 11/29/2022] Open
Abstract
Biological aging is the strongest factor associated with the clinical phenotype of multiple sclerosis (MS). Relapsing-remitting MS typically presents in the third or fourth decade, whereas the mean age of presentation of progressive MS (PMS) is 45 years old. Here, we show that experimental autoimmune encephalomyelitis (EAE), induced by the adoptive transfer of encephalitogenic CD4+ Th17 cells, was more severe, and less likely to remit, in middle-aged compared with young adult mice. Donor T cells and neutrophils were more abundant, while B cells were relatively sparse, in CNS infiltrates of the older mice. Experiments with reciprocal bone marrow chimeras demonstrated that radio-resistant, nonhematopoietic cells played a dominant role in shaping age-dependent features of the neuroinflammatory response, as well as the clinical course, during EAE. Reminiscent of PMS, EAE in middle-aged adoptive transfer recipients was characterized by widespread microglial activation. Microglia from older mice expressed a distinctive transcriptomic profile suggestive of enhanced chemokine synthesis and antigen presentation. Collectively, our findings suggest that drugs that suppress microglial activation, and acquisition or expression of aging-associated properties, may be beneficial in the treatment of progressive forms of inflammatory demyelinating disease.
Collapse
Affiliation(s)
- Jeffrey R Atkinson
- Department of Neurology, The Ohio State University, Columbus, United States of America
| | - Andrew D Jerome
- Department of Neurology, The Ohio State University, Columbus, United States of America
| | - Andrew R Sas
- Department of Neurology, The Ohio State University, Columbus, United States of America
| | - Ashley Munie
- Department of Neurology, The Ohio State University, Columbus, United States of America
| | - Cankun Wang
- Department of Biomedical Informatics, The Ohio Sate University, Columbus, United States of America
| | - Anjun Ma
- Department of Biomedical Informatics, The Ohio State University, Columbus, United States of America
| | - William D Arnold
- Department of Neurology, The Ohio State University, Columbus, United States of America
| | - Benjamin M Segal
- Department of Neurology, The Ohio State University, Columbus, United States of America
| |
Collapse
|
9
|
Huntemann N, Vogelsang A, Groeneweg L, Willison A, Herrmann AM, Meuth SG, Eichler S. An optimized and validated protocol for inducing chronic experimental autoimmune encephalomyelitis in C57BL/6J mice. J Neurosci Methods 2022; 367:109443. [PMID: 34920025 DOI: 10.1016/j.jneumeth.2021.109443] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 12/10/2021] [Accepted: 12/12/2021] [Indexed: 02/04/2023]
Abstract
BACKGROUND Myelin oligodendrocyte glycoprotein induced experimental autoimmune encephalomyelitis (EAE) is the most commonly used animal model of multiple sclerosis. However, variations in the induction protocol can affect EAE progression, and may reduce the comparability of data. OPTIMIZED METHOD In the present study, we investigated the influence of the different components used for EAE induction in C57BL/6J mice on disease progression. In the present study, MOG35-55-induced chronic EAE in C57BL/6J mice has been applied as a model to challenge optimal pertussis toxin (PTx) dosing, while considering variations in batch potency. RESULTS We demonstrate that the dosage of PTx, adjusted to its potency, influences EAE development in a dose-dependent manner. Our data show that with our protocol, which considers PTx potency, C57BL/6J mice consistently develop symptoms of EAE. The mice show a typical chronic course with symptom onset after 10.5 ± 1.08 days and maximum severity around day 16 postimmunization followed by a mild remission of symptoms. COMPARISON WITH EXISTING METHODS Previously studies reveal that alterations in PTx dosing directly modify EAE progression. Our present study highlights that PTx batches differ in potency, resulting in inconsistent EAE induction. We also provide a clear protocol that allows a reduction in the number of mice used in EAE experiments, while maintaining consistent results. CONCLUSION Higher standards for comparability and reproducibility are needed to ensure and maximize the generation of reliable EAE data. Specifically, consideration of PTx potency. With our method of establishing consistent EAE pathogenesis, improved animal welfare standards and a reduction of mice used in experimentation can be achieved.
Collapse
Affiliation(s)
- Niklas Huntemann
- Department of Neurology, University Hospital Duesseldorf, Heinrich Heine University Duesseldorf, Duesseldorf, Germany; Department of Neurology with Institute of Translational Neurology, University Hospital Muenster, Muenster, Germany
| | - Anna Vogelsang
- Department of Neurology with Institute of Translational Neurology, University Hospital Muenster, Muenster, Germany.
| | - Linda Groeneweg
- Institute of Immunology, University of Muenster, Muenster, Germany
| | - Alice Willison
- The Northern Foundation School, Newcastle-upon-Tyne University Hospitals, Newcastle-upon-Tyne, United Kingdom
| | - Alexander M Herrmann
- Department of Neurology, University Hospital Duesseldorf, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| | - Sven G Meuth
- Department of Neurology, University Hospital Duesseldorf, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| | - Susann Eichler
- Department of Neurology with Institute of Translational Neurology, University Hospital Muenster, Muenster, Germany
| |
Collapse
|
10
|
Cystatin C Plays a Sex-Dependent Detrimental Role in Experimental Autoimmune Encephalomyelitis. Cell Rep 2021; 33:108236. [PMID: 33027652 PMCID: PMC8603395 DOI: 10.1016/j.celrep.2020.108236] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 07/17/2020] [Accepted: 09/15/2020] [Indexed: 12/31/2022] Open
Abstract
The cysteine protease inhibitor Cystatin C (CST3) is highly expressed in the brains of multiple sclerosis (MS) patients and C57BL/6J mice with experimental autoimmune encephalomyelitis (EAE; a model of MS), but its roles in the diseases are unknown. Here, we show that CST3 plays a detrimental function in myelin oligodendrocyte glycoprotein 35–55 (MOG35–55)-induced EAE but only in female animals. Female Cst3 null mice display significantly lower clinical signs of disease compared to wild-type (WT) littermates. This difference is associated with reduced interleukin-6 production and lower expression of key proteins (CD80, CD86, major histocompatibility complex [MHC] II, LC3A/B) involved in antigen processing, presentation, and co-stimulation in antigen-presenting cells (APCs). In contrast, male WT and Cst3−/− mice and cells show no differences in EAE signs or APC function. Further, the sex-dependent effect of CST3 in EAE is sensitive to gonadal hormones. Altogether, we have shown that CST3 has a sex-dependent role in MOG35–55-induced EAE. Cystatin C (CST3) is increased in the brains of multiple sclerosis patients, but its role is unknown. In a mouse model of the disease, Hoghooghi et al. find that CST3 has a detrimental function but only in female animals. The effect is related to activation of antigen-presenting cells of the immune system.
Collapse
|
11
|
Andreas N, Müller S, Templin N, Jordan PM, Schuhwerk H, Müller M, Gerstmeier J, Miek L, Andreas S, Werz O, Kamradt T. Incidence and severity of G6PI-induced arthritis are not increased in genetically distinct mouse strains upon aging. Arthritis Res Ther 2021; 23:222. [PMID: 34429153 PMCID: PMC8383389 DOI: 10.1186/s13075-021-02596-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 08/02/2021] [Indexed: 12/02/2022] Open
Abstract
Background The incidence of rheumatoid arthritis is correlated with age. In this study, we analyzed the association of the incidence and severity of glucose-6-phosphate isomerase (G6PI)-induced arthritis with age in two different mouse strains. Methods Young and very old mice from two different arthritis-susceptible wild-type mouse strains were analyzed after a single subcutaneous injection of G6PI s.c. The metabolism and the function of synoviocytes were analyzed in vitro, the production of bioactive lipid mediators by myeloid cells and synoviocytes was assessed in vitro and ex vivo by UPLC-MS-MS, and flow cytometry was used to verify age-related changes of immune cell composition and function. Results While the severity of arthritis was independent from age, the onset was delayed in old mice. Old mice showed common signs of immune aging like thymic atrophy associated with decreased CD4+ effector T cell numbers. Despite its decrease, the effector T helper (Th) cell compartment in old mice was reactive and functionally intact, and their Tregs exhibited unaltered suppressive capacities. In homeostasis, macrophages and synoviocytes from old mice produced higher amounts of pro-inflammatory cyclooxygenase (COX)-derived products. However, this functional difference did not remain upon challenge in vitro nor upon arthritis reactions ex vivo. Conclusion While old mice show a higher baseline of inflammatory functions, this does not result in increased reaction towards self-antigens in arthritis-susceptible mouse strains. Together, our data from two different mouse strains show that the susceptibility for G6PI-induced arthritis is not age-dependent. Supplementary Information The online version contains supplementary material available at 10.1186/s13075-021-02596-7.
Collapse
Affiliation(s)
- Nico Andreas
- Institute of Immunology, University Hospital Jena, Leutragraben 3, 07743, Jena, Germany.
| | - Sylvia Müller
- Institute of Immunology, University Hospital Jena, Leutragraben 3, 07743, Jena, Germany
| | - Nicole Templin
- Institute of Immunology, University Hospital Jena, Leutragraben 3, 07743, Jena, Germany
| | - Paul M Jordan
- Department of Pharmaceutical/Medicinal Chemistry, Friedrich Schiller University, Philosophenweg 14, 07743, Jena, Germany
| | - Harald Schuhwerk
- Institute of Immunology, University Hospital Jena, Leutragraben 3, 07743, Jena, Germany
| | - Michael Müller
- Institute of Immunology, University Hospital Jena, Leutragraben 3, 07743, Jena, Germany
| | - Jana Gerstmeier
- Department of Pharmaceutical/Medicinal Chemistry, Friedrich Schiller University, Philosophenweg 14, 07743, Jena, Germany
| | - Laura Miek
- Department of Pharmaceutical/Medicinal Chemistry, Friedrich Schiller University, Philosophenweg 14, 07743, Jena, Germany
| | - Saskia Andreas
- Department of Pharmaceutical/Medicinal Chemistry, Friedrich Schiller University, Philosophenweg 14, 07743, Jena, Germany
| | - Oliver Werz
- Department of Pharmaceutical/Medicinal Chemistry, Friedrich Schiller University, Philosophenweg 14, 07743, Jena, Germany
| | - Thomas Kamradt
- Institute of Immunology, University Hospital Jena, Leutragraben 3, 07743, Jena, Germany.
| |
Collapse
|
12
|
Rojo JM, Montes-Casado M, Aragoneses-Fenoll L, Ojeda G, Dianzani U, Portolés P. PI3-Kinase p110α Deficiency Modulates T Cell Homeostasis and Function and Attenuates Experimental Allergic Encephalitis in Mature Mice. Int J Mol Sci 2021; 22:ijms22168698. [PMID: 34445401 PMCID: PMC8395417 DOI: 10.3390/ijms22168698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/09/2021] [Accepted: 08/11/2021] [Indexed: 11/16/2022] Open
Abstract
Class I phosphoinositide 3-kinases (PI3K) are involved in the development of normal and autoimmune responses, including Experimental Autoimmune Encephalomyelitis (EAE), a mouse model for human multiple sclerosis (MS). Here, the role of the ubiquitously expressed class IA PI3K p110α catalytic subunits in EAE has been analyzed using a model of Cre/flox mediated T cell specific deletion of p110α catalytic chain (p110αΔT). Comparison of two month-old (young) and six month-old (mature) p110αΔT mice and their wild type (WT) counterparts indicated loss of spleen CD4+ T cells that increased with age, indicating a role of p110α in their homeostasis. In contrast, CD4+ T regulatory (Treg) cells were enhanced in mature p110αΔT mice when compared to WT mice. Since Myelin Oligodendrocyte Glycoprotein (MOG) peptide-induced EAE is dependent on, or mediated by CD4+ T cells and CD4+ T cell-derived cytokines and controlled by Treg cells, development of EAE in young and mature WT or p110αΔT mice was analyzed. EAE clinical symptoms and disease scores in six month p110αΔT mice were significantly lower than those of mature WT, or young WT and p110αΔT mice. Furthermore, ex vivo antigen activation of lymph node cells from MOG immunized mature p110αΔT mice induced significantly lower levels of IFN-γ and IL-17A than young p110αΔT or young and mature WT mice. Other cytokines including IL-2, IL-10 or TNF-α showed no significant differences between p110αΔT and WT mature mice. Our data show a lower incidence of MOG-induced EAE in mature p110αΔT mice linked to altered T cell homeostasis and lower secretion of inflammatory cytokines.
Collapse
Affiliation(s)
- José M. Rojo
- Departamento de Biomedicina Molecular, Centro de Investigaciones Biológicas Margarita Salas, CSIC, 28040 Madrid, Spain
- Correspondence: (J.M.R.); (P.P.)
| | - María Montes-Casado
- Unidad de Inmunología Celular, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, 28220 Madrid, Spain; (M.M.-C.); (L.A.-F.); (G.O.)
| | - Laura Aragoneses-Fenoll
- Unidad de Inmunología Celular, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, 28220 Madrid, Spain; (M.M.-C.); (L.A.-F.); (G.O.)
| | - Gloria Ojeda
- Unidad de Inmunología Celular, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, 28220 Madrid, Spain; (M.M.-C.); (L.A.-F.); (G.O.)
| | - Umberto Dianzani
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), Department of Health Sciences, University of Piemonte Orientale (UPO), 28100 Novara, Italy;
| | - Pilar Portolés
- Unidad de Inmunología Celular, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, 28220 Madrid, Spain; (M.M.-C.); (L.A.-F.); (G.O.)
- Presidencia, Consejo Superior de Investigaciones Científicas (CSIC), 28006 Madrid, Spain
- Correspondence: (J.M.R.); (P.P.)
| |
Collapse
|
13
|
Abstract
Animal models with high translational validity are essential tools in understanding disease pathogenesis and in the development of therapeutic strategies. Multiple sclerosis (MS) is an autoimmune demyelinating disease of the central nervous system characterized by progressive neurological deficits and socioeconomic burden. Experimental autoimmune encephalomyelitis (EAE) is the most extensively utilized animal model of MS, with well-characterized rodent and non-human primate variants. The EAE model is typically induced by either active immunization with myelin-derived proteins or peptides in adjuvant or by passive transfer of activated myelin-specific CD4+ T lymphocytes. To date, the EAE model has been an essential tool in the development of at least seven U.S. Food and Drug Administration (FDA)-approved immunomodulatory drugs for the treatment of MS, including glatiramer acetate, fingolimod, and natalizumab. However, the translational validity of the EAE model is frequently compromised due to poor study design, inconsistent clinical scoring endpoints, and inappropriate statistical calculations. No single animal model accurately reflects the complexity of human MS pathogenesis. Beyond EAE, multiple additional animal models are described, including Theiler's murine encephalomyelitis virus and cuprizone-induced demyelination, which facilitate the study of pathogen-induced CNS autoimmunity and remyelination, respectively. This overview summarizes several of the most frequently used animal models of MS and highlights key factors that significantly influence the experimental outcome and affect translational validity. © 2021 Wiley Periodicals LLC.
Collapse
Affiliation(s)
- Paul Smith
- Incyte Research Institute, Wilmington, Delaware
| |
Collapse
|
14
|
Taherian N, Vaezi G, Neamati A, Etemad L, Hojjati V, Gorji-Valokola M. Vitamin B12 and estradiol benzoate improve memory retrieval through activation of the hippocampal AKT, BDNF, and CREB proteins in a rat model of multiple sclerosis. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2021; 24:256-263. [PMID: 33953866 PMCID: PMC8061324 DOI: 10.22038/ijbms.2021.51469.11681] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 12/14/2020] [Indexed: 11/29/2022]
Abstract
OBJECTIVES Multiple sclerosis (MS) causes extensive damage in the hippocampus. Vitamin B12 (vit B12) and estradiol benzoate (EB) have anti-inflammatory and re-myelination properties that make them proper in improvement of cognitive impairment. This study aimed to evaluate the effects of these compounds on learning and memory disturbances. MATERIALS AND METHODS 77 adult male rats were implanted with stainless steel guide cannula bilaterally into the hippocampal area. The animals received 3 μl intrahippocampal EtB 0.01% and were randomly divided into eleven groups (7 rats/group). The groups included control, peanut oil (sham1), distilled water (sham 2), vit B12 (0.25, 0.5, 1 mg/kg), EB (25 and 50 mg/kg), vit B12 (0.25 mg/kg) plus EB (25 mg/kg), vit B12 (0.5 mg/kg) plus EB (25 mg/kg), and vit B12 (1 mg/kg) plus EB (50 mg/kg). The control group received intrahippocampal saline (as solvent). The locomotor activity and learning and memory functions were evaluated by open-field and shuttle-box tests, respectively. AKT, CREB, and BDNF levels were analyzed by Western blotting. RESULTS This study has found significant deficit in passive avoidance learning, locomotor activity, as well as decrease in the levels of phosphorylated AKT, BDNF, and CREB in groups that received EtB. Vit B12 (1 mg/kg), EB (50 mg/kg), and their combination markedly improved these side effects. CONCLUSION This study demonstrated that vit B12 and estradiol benzoate, especially in combination therapy, can be helpful in treatment of memory problems and MS-induced dysfunction through activation of the hippocampal AKT, BDNF, and CREB proteins.
Collapse
Affiliation(s)
- Narjes Taherian
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Gholamhassan Vaezi
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Ali Neamati
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Leila Etemad
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vida Hojjati
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Mahmoud Gorji-Valokola
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
15
|
Naffaa V, Laprévote O, Schang AL. Effects of endocrine disrupting chemicals on myelin development and diseases. Neurotoxicology 2020; 83:51-68. [PMID: 33352275 DOI: 10.1016/j.neuro.2020.12.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/10/2020] [Accepted: 12/16/2020] [Indexed: 12/14/2022]
Abstract
In the central and peripheral nervous systems, myelin is essential for efficient conduction of action potentials. During development, oligodendrocytes and Schwann cells differentiate and ensure axon myelination, and disruption of these processes can contribute to neurodevelopmental disorders. In adults, demyelination can lead to important disabilities, and recovery capacities by remyelination often decrease with disease progression. Among environmental chemical pollutants, endocrine disrupting chemicals (EDCs) are of major concern for human health and are notably suspected to participate in neurodevelopmental and neurodegenerative diseases. In this review, we have combined the current knowledge on EDCs impacts on myelin including several persistent organic pollutants, bisphenol A, triclosan, heavy metals, pesticides, and nicotine. Besides, we presented several other endocrine modulators, including pharmaceuticals and the phytoestrogen genistein, some of which are candidates for treating demyelinating conditions but could also be deleterious as contaminants. The direct impacts of EDCs on myelinating cells were considered as well as their indirect consequences on myelin, particularly on immune mechanisms associated with demyelinating conditions. More studies are needed to describe the effects of these compounds and to further understand the underlying mechanisms in relation to the potential for endocrine disruption.
Collapse
Affiliation(s)
- Vanessa Naffaa
- Université de Paris, UMR 8038 (CiTCoM), CNRS, Faculté de Pharmacie de Paris, 4 avenue de l'Observatoire, 75006 Paris, France.
| | - Olivier Laprévote
- Université de Paris, UMR 8038 (CiTCoM), CNRS, Faculté de Pharmacie de Paris, 4 avenue de l'Observatoire, 75006 Paris, France; Hôpital Européen Georges Pompidou, AP-HP, Service de Biochimie, 20 rue Leblanc, 75015 Paris, France.
| | - Anne-Laure Schang
- Université de Paris, UMR 1153 (CRESS), Faculté de Pharmacie de Paris, 4 avenue de l'Observatoire, 75006 Paris, France.
| |
Collapse
|
16
|
Henze L, Schwinge D, Schramm C. The Effects of Androgens on T Cells: Clues to Female Predominance in Autoimmune Liver Diseases? Front Immunol 2020; 11:1567. [PMID: 32849531 PMCID: PMC7403493 DOI: 10.3389/fimmu.2020.01567] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 06/15/2020] [Indexed: 12/16/2022] Open
Abstract
The immune system responds differently in women and in men. Generally speaking, adult females show stronger innate and adaptive immune responses than males. This results in lower risk of developing most of the infectious diseases and a better ability to clear viral infection in women (1–5). On the other hand, women are at increased risk of developing autoimmune diseases (AID) such as rheumatoid arthritis, multiple sclerosis (MS), systemic lupus erythematosus (SLE), Sjögren's syndrome, and the autoimmune liver diseases autoimmune hepatitis (AIH) and primary biliary cholangitis (PBC) (6). Factors contributing to the female sex bias in autoimmune diseases include environmental exposure, e.g., microbiome, behavior, and genetics including X chromosomal inactivation of genes. Several lines of evidence and clinical observations clearly indicate that sex hormones contribute significantly to disease pathogenesis, and the role of estrogen in autoimmune diseases has been extensively studied. In many of these diseases, including the autoimmune liver diseases, T cells are thought to play an important pathogenetic role. We will use this mini-review to focus on the effects of androgens on T cells and how the two major androgens, testosterone and dihydrotestosterone, potentially contribute to the pathogenesis of autoimmune liver diseases (AILD).
Collapse
Affiliation(s)
- Lara Henze
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Dorothee Schwinge
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christoph Schramm
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Martin Zeitz Centre for Rare Diseases, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
17
|
Gilli F, DiSano KD, Pachner AR. SeXX Matters in Multiple Sclerosis. Front Neurol 2020; 11:616. [PMID: 32719651 PMCID: PMC7347971 DOI: 10.3389/fneur.2020.00616] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 05/27/2020] [Indexed: 12/12/2022] Open
Abstract
Multiple sclerosis (MS) is the most common chronic inflammatory and neurodegenerative disease of the central nervous system (CNS). An interesting feature that this debilitating disease shares with many other inflammatory disorders is that susceptibility is higher in females than in males, with the risk of MS being three times higher in women compared to men. Nonetheless, while men have a decreased risk of developing MS, many studies suggest that males have a worse clinical outcome. MS exhibits an apparent sexual dimorphism in both the immune response and the pathophysiology of the CNS damage, ultimately affecting disease susceptibility and progression differently. Overall, women are predisposed to higher rates of inflammatory relapses than men, but men are more likely to manifest signs of disease progression and worse CNS damage. The observed sexual dimorphism in MS may be due to sex hormones and sex chromosomes, acting in parallel or combination. In this review, we outline current knowledge on the sexual dimorphism in MS and discuss the interplay of sex chromosomes, sex hormones, and the immune system in driving MS disease susceptibility and progression.
Collapse
Affiliation(s)
- Francesca Gilli
- Department of Neurology, Dartmouth Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | | | | |
Collapse
|
18
|
Natural killer cells as participants in pathogenesis of rat experimental autoimmune encephalomyelitis (EAE): lessons from research on rats with distinct age and strain. Cent Eur J Immunol 2020; 44:337-356. [PMID: 32140045 PMCID: PMC7050050 DOI: 10.5114/ceji.2019.92777] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 11/12/2018] [Indexed: 01/08/2023] Open
Abstract
Natural killer (NK) cells, influencing dendritic cell (DC)-mediated CD4+ lymphocyte priming in draining lymph nodes (dLNs) and controlling spinal cord (SC) infiltration with encephalitogenic CD4+T lymphocytes, modulate EAE (multiple sclerosis model). This study examined their putative contribution to age-related differences in EAE development in Dark Agouti (DA) (exhibiting age-related decrease in EAE susceptibility) and Albino Oxford (AO) (becoming susceptible to EAE with aging) rats. Aging increased NK cell number in dLNs from rats of both strains. In AO rats, but not in DA ones, it also increased the numbers of IFN-γ-producing NK cells (important for DC activation) and activated/matured DCs, thereby increasing activated/matured DC/conventional Foxp3-CD4+ cell ratio and activated CD25+Foxp3-CD4+ cell number. Aging in DA rats diminished activated/matured DC/conventional Foxp3-CD4+ cell ratio and activated Foxp3-CD4+ cell number. However, MBP-stimulated CD4+ cell proliferation did not differ in dLN cell cultures from young and aged AO rats (as more favorable activated/matured DC/Foxp3-CD4+ cell ratio was abrogated by lower intrinsic CD4+ cell proliferative capacity and a greater regulatory CD25+Foxp3+CD4+ lymphocyte frequency), but was lower in those from aged compared with young DA rats. At SC level, aging shifted Foxp3-CD4+/cytotoxic CX3CR1+ NK cell ratio towards the former in AO rats, so it was less favorable in aged AO rats exhibiting prolonged neurological deficit compared with their DA counterparts. The study showed strain and age differences in number of IFN-γ-producing NK cells in EAE rat dLNs, and suggested that their pathogenetic relevance depends on frequency and/or activity of other cells involved in CD4+ T cell (auto)immune response.
Collapse
|
19
|
Brown MA, Su MA. An Inconvenient Variable: Sex Hormones and Their Impact on T Cell Responses. THE JOURNAL OF IMMUNOLOGY 2020; 202:1927-1933. [PMID: 30885988 DOI: 10.4049/jimmunol.1801403] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 12/05/2018] [Indexed: 12/15/2022]
Abstract
Epidemiologic data demonstrate sex differences in autoimmune diseases, immune responses against infection, and antitumor immunity, and accumulating evidence suggests a major role for sex hormones in mediating these differences. In this study, we review recent advances in understanding how sex hormones regulate T cell responses to alter susceptibility to autoimmunity. Although sex hormones can directly alter gene transcriptional programs of T cells, we focus in this study on how sex hormones alter T cell development and function through their effects on thymic stromal cells and innate cell types. In addition to contributing to our understanding of sex differences, these findings also have implications for the therapeutic use of sex hormones and sex hormone modulators, which are now being prescribed to increasing numbers of patients for a wide variety of indications.
Collapse
Affiliation(s)
- Melissa A Brown
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Maureen A Su
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, College of Life Sciences, University of California at Los Angeles, Los Angeles, CA 90095; and .,Department of Pediatrics, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095
| |
Collapse
|
20
|
Mayne K, White JA, McMurran CE, Rivera FJ, de la Fuente AG. Aging and Neurodegenerative Disease: Is the Adaptive Immune System a Friend or Foe? Front Aging Neurosci 2020; 12:572090. [PMID: 33173502 PMCID: PMC7538701 DOI: 10.3389/fnagi.2020.572090] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 08/31/2020] [Indexed: 12/11/2022] Open
Abstract
Neurodegenerative diseases of the central nervous system (CNS) are characterized by progressive neuronal death and neurological dysfunction, leading to increased disability and a loss of cognitive or motor functions. Alzheimer's disease, Parkinson's disease and amyotrophic lateral sclerosis have neurodegeneration as a primary feature. However, in other CNS diseases such as multiple sclerosis, stroke, traumatic brain injury, and spinal cord injury, neurodegeneration follows another insult, such as demyelination or ischaemia. Although there are different primary causes to these diseases, they all share a hallmark of neuroinflammation. Neuroinflammation can occur through the activation of resident immune cells such as microglia, cells of the innate and adaptive peripheral immune system, meningeal inflammation and autoantibodies directed toward components of the CNS. Despite chronic inflammation being pathogenic in these diseases, local inflammation after insult can also promote endogenous regenerative processes in the CNS, which are key to slowing disease progression. The normal aging process in the healthy brain is associated with a decline in physiological function, a steady increase in levels of neuroinflammation, brain shrinkage, and memory deficits. Likewise, aging is also a key contributor to the progression and exacerbation of neurodegenerative diseases. As there are associated co-morbidities within an aging population, pinpointing the precise relationship between aging and neurodegenerative disease progression can be a challenge. The CNS has historically been considered an isolated, "immune privileged" site, however, there is mounting evidence that adaptive immune cells are present in the CNS of both healthy individuals and diseased patients. Adaptive immune cells have also been implicated in both the degeneration and regeneration of the CNS. In this review, we will discuss the key role of the adaptive immune system in CNS degeneration and regeneration, with a focus on how aging influences this crosstalk.
Collapse
Affiliation(s)
- Katie Mayne
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen’s University Belfast, Belfast, United Kingdom
| | - Jessica A. White
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen’s University Belfast, Belfast, United Kingdom
| | | | - Francisco J. Rivera
- Laboratory of Stem Cells and Neuroregeneration, Institute of Anatomy, Histology and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
| | - Alerie G. de la Fuente
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen’s University Belfast, Belfast, United Kingdom
- *Correspondence: Alerie G. de la Fuente,
| |
Collapse
|
21
|
Sun M, McDonald SJ, Brady RD, Collins-Praino L, Yamakawa GR, Monif M, O'Brien TJ, Cloud GC, Sobey CG, Mychasiuk R, Loane DJ, Shultz SR. The need to incorporate aged animals into the preclinical modeling of neurological conditions. Neurosci Biobehav Rev 2019; 109:114-128. [PMID: 31877345 DOI: 10.1016/j.neubiorev.2019.12.027] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 12/04/2019] [Accepted: 12/19/2019] [Indexed: 12/14/2022]
Abstract
Neurological conditions such as traumatic brain injury, stroke, Parkinson's disease, epilepsy, multiple sclerosis, and Alzheimer's disease are serious clinical problems that affect millions of people worldwide. The majority of clinical trials for these common conditions have failed, and there is a critical need to understand why treatments in preclinical animal models do not translate to patients. Many patients with these conditions are middle-aged or older, however, the majority of preclinical studies have used only young-adult animals. Considering that aging involves biological changes that are relevant to the pathobiology of neurological diseases, the lack of aged subjects in preclinical research could contribute to translational failures. This paper details how aging affects biological processes involved in neurological conditions, and reviews aging research in the context of traumatic brain injury, stroke, Parkinson's disease, epilepsy, multiple sclerosis, and Alzheimer's disease. We conclude that aging is an important, but often overlooked, factor that influences biology and outcomes in neurological conditions, and provide suggestions to improve our understanding and treatment of these diseases in aged patients.
Collapse
Affiliation(s)
- Mujun Sun
- Department of Neuroscience, Monash University, Melbourne, VIC 3004, Australia
| | - Stuart J McDonald
- Department of Neuroscience, Monash University, Melbourne, VIC 3004, Australia; Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, VIC 3086, Australia
| | - Rhys D Brady
- Department of Neuroscience, Monash University, Melbourne, VIC 3004, Australia; Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC 3052, Australia
| | - Lyndsey Collins-Praino
- Department of Medical Sciences, Adelaide Medical School, The University of Adelaide, Adelaide, SA 5005, Australia
| | - Glenn R Yamakawa
- Department of Neuroscience, Monash University, Melbourne, VIC 3004, Australia
| | - Mastura Monif
- Department of Neuroscience, Monash University, Melbourne, VIC 3004, Australia
| | - Terence J O'Brien
- Department of Neuroscience, Monash University, Melbourne, VIC 3004, Australia; Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC 3052, Australia; Department of Neurology, Alfred Health, Melbourne, VIC 3004, Australia
| | - Geoffrey C Cloud
- Department of Neuroscience, Monash University, Melbourne, VIC 3004, Australia; Department of Stroke Services, Alfred Hospital, Melbourne, VIC 3004, Australia
| | - Christopher G Sobey
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, VIC 3086, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Monash University, Melbourne, VIC 3004, Australia
| | - David J Loane
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA; School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College, Dublin 2, Ireland
| | - Sandy R Shultz
- Department of Neuroscience, Monash University, Melbourne, VIC 3004, Australia; Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC 3052, Australia; Department of Neurology, Alfred Health, Melbourne, VIC 3004, Australia.
| |
Collapse
|
22
|
TGFB1-Mediated Gliosis in Multiple Sclerosis Spinal Cords Is Favored by the Regionalized Expression of HOXA5 and the Age-Dependent Decline in Androgen Receptor Ligands. Int J Mol Sci 2019; 20:ijms20235934. [PMID: 31779094 PMCID: PMC6928867 DOI: 10.3390/ijms20235934] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 11/18/2019] [Accepted: 11/22/2019] [Indexed: 02/07/2023] Open
Abstract
In multiple sclerosis (MS) patients with a progressive form of the disease, spinal cord (SC) functions slowly deteriorate beyond age 40. We previously showed that in the SC of these patients, large areas of incomplete demyelination extend distance away from plaque borders and are characterized by a unique progliotic TGFB1 (Transforming Growth Factor Beta 1) genomic signature. Here, we attempted to determine whether region- and age-specific physiological parameters could promote the progression of SC periplaques in MS patients beyond age 40. An analysis of transcriptomics databases showed that, under physiological conditions, a set of 10 homeobox (HOX) genes are highly significantly overexpressed in the human SC as compared to distinct brain regions. Among these HOX genes, a survey of the human proteome showed that only HOXA5 encodes a protein which interacts with a member of the TGF-beta signaling pathway, namely SMAD1 (SMAD family member 1). Moreover, HOXA5 was previously found to promote the TGF-beta pathway. Interestingly, SMAD1 is also a protein partner of the androgen receptor (AR) and an unsupervised analysis of gene ontology terms indicates that the AR pathway antagonizes the TGF-beta/SMAD pathway. Retrieval of promoter analysis data further confirmed that AR negatively regulates the transcription of several members of the TGF-beta/SMAD pathway. On this basis, we propose that in progressive MS patients, the physiological SC overexpression of HOXA5 combined with the age-dependent decline in AR ligands may favor the slow progression of TGFB1-mediated gliosis. Potential therapeutic implications are discussed.
Collapse
|
23
|
Vom Steeg LG, Attreed SE, Zirkin B, Klein SL. Testosterone treatment of aged male mice improves some but not all aspects of age-associated increases in influenza severity. Cell Immunol 2019; 345:103988. [PMID: 31540670 PMCID: PMC6876866 DOI: 10.1016/j.cellimm.2019.103988] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 09/12/2019] [Accepted: 09/13/2019] [Indexed: 12/13/2022]
Abstract
The severity of influenza increases with age, with worse disease in aged males than females. Testosterone concentrations decline with age in males, which may impact influenza pathogenesis. Aged male mice were treated with testosterone or placebo and outcomes during influenza A virus (IAV) infection were compared with adult male mice. Aged males experienced greater morbidity and mortality than adult males, which was partially improved by testosterone treatment of aged males. Aged males cleared IAV from lungs slower than adult males, regardless of testosterone treatment. As compared with adult males, aged males experienced pulmonary, but not systemic, cytokine dysregulation, and delayed influx and contraction of IAV-specific CD8+ T cells in the lungs. Testosterone treatment in aged males partially restored pulmonary cytokine responses to levels consistent with adult males but did not alter the age-associated changes in IAV-specific CD8+ T cells. Testosterone only modestly improves outcomes of influenza in aged males.
Collapse
Affiliation(s)
- Landon G Vom Steeg
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Sarah E Attreed
- Department of Environmental Health and Engineering, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Barry Zirkin
- Department of Biochemistry and Molecular Biology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Sabra L Klein
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA; Department of Biochemistry and Molecular Biology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| |
Collapse
|
24
|
Mendibe Bilbao M, Boyero Durán S, Bárcena Llona J, Rodriguez-Antigüedad A. Multiple sclerosis: pregnancy and women's health issues. NEUROLOGÍA (ENGLISH EDITION) 2019. [DOI: 10.1016/j.nrleng.2016.06.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
25
|
de Souza RG, de Paiva CS, Alves MR. Age-related Autoimmune Changes in Lacrimal Glands. Immune Netw 2019; 19:e3. [PMID: 30838158 PMCID: PMC6399097 DOI: 10.4110/in.2019.19.e3] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 02/10/2019] [Accepted: 02/11/2019] [Indexed: 02/08/2023] Open
Abstract
Aging is a complex process associated with dysregulation of the immune system and low levels of inflammation, often associated with the onset of many pathologies. The lacrimal gland (LG) plays a vital role in the maintenance of ocular physiology and changes related to aging directly affect eye diseases. The dysregulation of the immune system in aging leads to quantitative and qualitative changes in antibodies and cytokines. While there is a gradual decline of the immune system, there is an increase in autoimmunity, with a reciprocal pathway between low levels of inflammation and aging mechanisms. Elderly C57BL/6J mice spontaneously show LGs infiltration that is characterized by Th1 but not Th17 cells. The aging of the LG is related to functional alterations, reduced innervation and decreased secretory activities. Lymphocytic infiltration, destruction, and atrophy of glandular parenchyma, ductal dilatation, and secretion of inflammatory mediators modify the volume and composition of tears. Oxidative stress, the capacity to metabolize and eliminate toxic substances decreased in aging, is also associated with the reduction of LG functionality and the pathogenesis of autoimmune diseases. Although further studies are required for a better understanding of autoimmunity and aging of the LG, we described anatomic and immunology aspects that have been described so far.
Collapse
Affiliation(s)
- Rodrigo G de Souza
- University of Sao Paulo, Sao Paulo, Brazil.,Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA
| | - Cintia S de Paiva
- University of Sao Paulo, Sao Paulo, Brazil.,Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA
| | | |
Collapse
|
26
|
Burrows DJ, McGown A, Jain SA, De Felice M, Ramesh TM, Sharrack B, Majid A. Animal models of multiple sclerosis: From rodents to zebrafish. Mult Scler 2018; 25:306-324. [PMID: 30319015 DOI: 10.1177/1352458518805246] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Multiple sclerosis (MS) is a chronic, immune-mediated demyelinating disease of the central nervous system. Animal models of MS have been critical for elucidating MS pathological mechanisms and how they may be targeted for therapeutic intervention. Here we review the most commonly used animal models of MS. Although these animal models cannot fully replicate the MS disease course, a number of models have been developed to recapitulate certain stages. Experimental autoimmune encephalomyelitis (EAE) has been used to explore neuroinflammatory mechanisms and toxin-induced demyelinating models to further our understanding of oligodendrocyte biology, demyelination and remyelination. Zebrafish models of MS are emerging as a useful research tool to validate potential therapeutic candidates due to their rapid development and amenability to genetic manipulation.
Collapse
Affiliation(s)
- David John Burrows
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Alexander McGown
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Saurabh A Jain
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Milena De Felice
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Tennore M Ramesh
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Basil Sharrack
- Academic Department of Neuroscience, The Sheffield NIHR Translational Neuroscience Biomedical Research Centre, University of Sheffield, Sheffield, UK
| | - Arshad Majid
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK/Academic Department of Neuroscience, The Sheffield NIHR Translational Neuroscience Biomedical Research Centre, University of Sheffield, Sheffield, UK
| |
Collapse
|
27
|
Brown MA, Weinberg RB. Mast Cells and Innate Lymphoid Cells: Underappreciated Players in CNS Autoimmune Demyelinating Disease. Front Immunol 2018; 9:514. [PMID: 29619025 PMCID: PMC5871669 DOI: 10.3389/fimmu.2018.00514] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 02/27/2018] [Indexed: 12/29/2022] Open
Abstract
Multiple sclerosis (MS) and its mouse model, experimental autoimmune encephalomyelitis, are autoimmune CNS inflammatory diseases. As a result of a breakdown in the relatively impermeable blood–brain barrier (BBB) in affected individuals, myelin-specific CD4+ and CD8+ T cells gain entry into the immune privileged CNS and initiate myelin, oligodendrocyte, and nerve axon destruction. However, despite the absolute requirement for T cells, there is increasing evidence that innate immune cells also play critical amplifying roles in disease pathogenesis. By modulating the character and magnitude of the myelin-reactive T cell response and regulating BBB integrity, innate cells affect both disease initiation and progression. Two classes of innate cells, mast cells and innate lymphoid cells (ILCs), have been best studied in models of allergic and gastrointestinal inflammatory diseases. Yet, there is emerging evidence that these cell types also exert a profound influence in CNS inflammatory disease. Both cell types are residents within the meninges and can be activated early in disease to express a wide variety of disease-modifying cytokines and chemokines. In this review, we discuss how mast cells and ILCs can have either disease-promoting or -protecting effects on MS and other CNS inflammatory diseases and how sex hormones may influence this outcome. These observations suggest that targeting these cells and their unique mediators can be exploited therapeutically.
Collapse
Affiliation(s)
- Melissa A Brown
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Rebecca B Weinberg
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| |
Collapse
|
28
|
Bjelobaba I, Begovic-Kupresanin V, Pekovic S, Lavrnja I. Animal models of multiple sclerosis: Focus on experimental autoimmune encephalomyelitis. J Neurosci Res 2018; 96:1021-1042. [PMID: 29446144 DOI: 10.1002/jnr.24224] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 01/15/2018] [Accepted: 01/25/2018] [Indexed: 12/15/2022]
Abstract
Multiple sclerosis (MS) is a chronic, progressive disorder of the central nervous system (CNS) that affects more than two million people worldwide. Several animal models resemble MS pathology; the most employed are experimental autoimmune encephalomyelitis (EAE) and toxin- and/or virus-induced demyelination. In this review we will summarize our knowledge on the utility of different animal models in MS research. Although animal models cannot replicate the complexity and heterogeneity of the MS pathology, they have proved to be useful for the development of several drugs approved for treatment of MS patients. This review focuses on EAE because it represents both clinical and pathological features of MS. During the past decades, EAE has been effective in illuminating various pathological processes that occur during MS, including inflammation, CNS penetration, demyelination, axonopathy, and neuron loss mediated by immune cells.
Collapse
Affiliation(s)
- Ivana Bjelobaba
- Institute for Biological Research "Sinisa Stankovic," Department of Neurobiology, University of Belgrade, Belgrade, Serbia
| | | | - Sanja Pekovic
- Institute for Biological Research "Sinisa Stankovic," Department of Neurobiology, University of Belgrade, Belgrade, Serbia
| | - Irena Lavrnja
- Institute for Biological Research "Sinisa Stankovic," Department of Neurobiology, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
29
|
Male-specific IL-33 expression regulates sex-dimorphic EAE susceptibility. Proc Natl Acad Sci U S A 2018; 115:E1520-E1529. [PMID: 29378942 DOI: 10.1073/pnas.1710401115] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The cellular and molecular basis of sex-dimorphic autoimmune diseases, such as the CNS demyelinating disease multiple sclerosis (MS), remains unclear. Our studies in the SJL mouse model of MS, experimental autoimmune encephalomyelitis (EAE), reveal that sex-determined differences in Il33 expression by innate immune cells in response to myelin peptide immunization regulate EAE susceptibility. IL-33 is selectively induced in PLP139-151-immunized males and activates type 2 innate lymphoid cells (ILC2s), cells that promote and sustain a nonpathogenic Th2 myelin-specific response. Without this attenuating IL-33 response, females generate an encephalitogenic Th17-dominant response, which can be reversed by IL-33 treatment. Mast cells are one source of IL-33 and we provide evidence that testosterone directly induces Il33 gene expression and also exerts effects on the potential for Il33 gene expression during mast cell development. Thus, in contrast to their pathogenic role in allergy, we propose a sex-specific role for both mast cells and ILC2s as attenuators of the pathogenic Th response in CNS inflammatory disease.
Collapse
|
30
|
The influence and impact of ageing and immunosenescence (ISC) on adaptive immunity during multiple sclerosis (MS) and the animal counterpart experimental autoimmune encephalomyelitis (EAE). Ageing Res Rev 2018; 41:64-81. [PMID: 29101043 DOI: 10.1016/j.arr.2017.10.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 10/23/2017] [Accepted: 10/25/2017] [Indexed: 12/21/2022]
Abstract
The human ageing process encompasses mechanisms that effect a decline in homeostasis with increased susceptibility to disease and the development of chronic life-threatening illness. Increasing age affects the immune system which undergoes a progressive loss of efficiency, termed immunosenescence (ISC), to impact on quantitative and functional aspects of innate and adaptive immunity. The human demyelinating disease multiple sclerosis (MS) and the corresponding animal model experimental autoimmune encephalomyelitis (EAE) are strongly governed by immunological events that primarily involve the adaptive arm of the immune response. MS and EAE are frequently characterised by a chronic pathology and a protracted disease course which thereby creates the potential for exposure to the inherent, on-going effects and consequences of ISC. Collective evidence is presented to confirm the occurrence of established and unendorsed biological markers of ISC during the development of both diseases. Moreover, results are discussed from studies during the course of MS and EAE that reveal a premature upregulation of ISC-related biomarkers which indicates untimely alterations to the adaptive immune system. The effects of ISC and a prematurely aged immune system on autoimmune-associated neurodegenerative conditions such as MS and EAE are largely unknown but current evaluation of data justifies and encourages further investigation.
Collapse
|
31
|
Seifert HA, Benedek G, Nguyen H, Kent G, Vandenbark AA, Offner H. Estrogen protects both sexes against EAE by promoting common regulatory cell subtypes independent of endogenous estrogen. Metab Brain Dis 2017; 32:1747-1754. [PMID: 28689297 PMCID: PMC5650507 DOI: 10.1007/s11011-017-0063-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 06/22/2017] [Indexed: 10/19/2022]
Abstract
Autoimmune diseases including multiple sclerosis predominantly affect females. Although high levels of sex hormones, particularly estrogen (E2), can reduce proinflammatory immune responses, it remains unclear if a lack of endogenous sex hormones might affect treatment with exogenous sex hormones. Pretreatment with E2 almost completely prevents intact female and male mice from developing clinical and histological signs of experimental autoimmune encephalomyelitis (EAE) by promoting various regulatory immune cell phenotypes. To evaluate the effects of exogenous estrogen in the absence of endogenous sex hormones, the current study compared EAE severity and the emergence of different immunoregulatory cell populations after E2 pretreatment of ovariectomized (OVX) female versus male mice. We found that E2 equally protected both OVX females and males from EAE over a 21 day observation period concomitant with reduced total cell numbers in spleen and spinal cord (males only), but enhanced percentages of CD19+CD5+CD1dhi, CD19+CD138+CD44hi and CD19+Tim-1+ Breg cells, CD8+CD122+ Treg cells and CD11b+CD 206+ARG-1+ anti-inflammatory M2-like monocytes/macrophages in both groups. In contrast, E2 decreased the percentage of CD4+CD25+FoxP3+ Treg cells in OVX females but increased these Treg cells in males and intact female mice. These data suggest that with the exception of CD4+CD25+FoxP3+ Treg cells, E2 protection against EAE promotes highly overlapping immunoregulatory subsets in OVX females and males.
Collapse
Affiliation(s)
- Hilary A Seifert
- Department of Neurology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, USA
- Neuroimmunology Research, R&D-31, VA Portland Health Care System, 3710 SW US Veterans Hospital Rd, Portland, OR, 97239, USA
| | - Gil Benedek
- Department of Neurology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, USA
- Neuroimmunology Research, R&D-31, VA Portland Health Care System, 3710 SW US Veterans Hospital Rd, Portland, OR, 97239, USA
| | - Ha Nguyen
- Department of Neurology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, USA
- Neuroimmunology Research, R&D-31, VA Portland Health Care System, 3710 SW US Veterans Hospital Rd, Portland, OR, 97239, USA
| | - Gail Kent
- Department of Neurology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, USA
- Neuroimmunology Research, R&D-31, VA Portland Health Care System, 3710 SW US Veterans Hospital Rd, Portland, OR, 97239, USA
| | - Arthur A Vandenbark
- Department of Neurology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, USA
- Neuroimmunology Research, R&D-31, VA Portland Health Care System, 3710 SW US Veterans Hospital Rd, Portland, OR, 97239, USA
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, USA
| | - Halina Offner
- Department of Neurology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, USA.
- Neuroimmunology Research, R&D-31, VA Portland Health Care System, 3710 SW US Veterans Hospital Rd, Portland, OR, 97239, USA.
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, USA.
| |
Collapse
|
32
|
Mendibe Bilbao M, Boyero Durán S, Bárcena Llona J, Rodriguez-Antigüedad A. Multiple sclerosis: Pregnancy and women's health issues. Neurologia 2016; 34:259-269. [PMID: 27546613 DOI: 10.1016/j.nrl.2016.06.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 06/06/2016] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND The course of multiple sclerosis (MS) is influenced by sex, pregnancy and hormonal factors. AIMS To analyse the influence of the above factors in order to clarify the aetiopathogenic mechanisms involved in the disease. METHODS We conducted a comprehensive review of scientific publications in the PubMed database using a keyword search for 'multiple sclerosis', 'MS', 'EAE', 'pregnancy', 'hormonal factors', 'treatment', and related terms. We reviewed the advances presented at the meeting held by the European Committee for Treatment and Research in Multiple Sclerosis (ECTRIMS) in March 2013 in London, as well as recommendations by international experts. RESULTS AND CONCLUSIONS We provide recommendations for counselling and treating women with MS prior to and during pregnancy and after delivery. Current findings on the effects of treatment on the mother, fetus, and newborn are also presented. We issue recommendations for future research in order to address knowledge gaps and clarify any inconsistencies in currently available data.
Collapse
Affiliation(s)
- M Mendibe Bilbao
- Departamento de Neurología, Hospital Universitario Cruces, Baracaldo, España.
| | - S Boyero Durán
- Departamento de Neurología, Hospital Universitario Cruces, Baracaldo, España
| | - J Bárcena Llona
- Departamento de Neurología, Hospital Universitario Cruces, Baracaldo, España
| | | |
Collapse
|
33
|
Sex effects on inflammatory and neurodegenerative processes in multiple sclerosis. Neurosci Biobehav Rev 2016; 67:137-46. [DOI: 10.1016/j.neubiorev.2015.12.015] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 12/09/2015] [Accepted: 12/21/2015] [Indexed: 01/14/2023]
|
34
|
Kloehn I, Pillai SB, Officer L, Klement C, Gasser PJ, Evans JA. Sexual Differentiation of Circadian Clock Function in the Adrenal Gland. Endocrinology 2016; 157:1895-904. [PMID: 27007073 DOI: 10.1210/en.2015-1968] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Sex differences in glucocorticoid production are associated with increased responsiveness of the adrenal gland in females. However, the adrenal-intrinsic mechanisms that establish sexual dimorphic function remain ill defined. Glucocorticoid production is gated at the molecular level by the circadian clock, which may contribute to sexual dimorphic adrenal function. Here we examine sex differences in the adrenal gland using an optical reporter of circadian clock function. Adrenal glands were cultured from male and female Period2::Luciferase (PER2::LUC) mice to assess clock function in vitro in real time. We confirm that there is a pronounced sex difference in the intrinsic capacity to sustain PER2::LUC rhythms in vitro, with higher amplitude rhythms in adrenal glands collected from males than from females. Changes in adrenal PER2::LUC rhythms over the reproductive life span implicate T as an important factor in driving sex differences in adrenal clock function. By directly manipulating hormone levels in adult mice in vivo, we demonstrate that T increases the amplitude of PER2::LUC rhythms in adrenal glands of both male and female mice. In contrast, we find little evidence that ovarian hormones modify adrenal clock function. Lastly, we find that T in vitro can increase the amplitude of PER2::LUC rhythms in male adrenals but not female adrenals, which suggests the existence of sex differences in the mechanisms of T action in vivo. Collectively these results reveal that activational effects of T alter circadian timekeeping in the adrenal gland, which may have implications for sex differences in stress reactivity and stress-related disorders.
Collapse
Affiliation(s)
- Ian Kloehn
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin 53233
| | - Savin B Pillai
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin 53233
| | - Laurel Officer
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin 53233
| | - Claire Klement
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin 53233
| | - Paul J Gasser
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin 53233
| | - Jennifer A Evans
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin 53233
| |
Collapse
|
35
|
Stojić-Vukanić Z, Nacka-Aleksić M, Pilipović I, Vujnović I, Blagojević V, Kosec D, Dimitrijević M, Leposavić G. Aging diminishes the resistance of AO rats to EAE: putative role of enhanced generation of GM-CSF Expressing CD4+ T cells in aged rats. IMMUNITY & AGEING 2015; 12:16. [PMID: 26448779 PMCID: PMC4596406 DOI: 10.1186/s12979-015-0044-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 09/25/2015] [Indexed: 12/22/2022]
Abstract
BACKGROUND Aging influences immune response and susceptibility to EAE in a strain specific manner. The study was designed to examine influence of aging on EAE induction in Albino Oxford (AO) rats. RESULTS Differently from 3-month-old (young) rats, which were resistant to EAE induction, the majority of aged (24-26-month-old) rats developed mild chronic form of EAE. On 16(th) day post-immunization, when in aged rats the neurological deficit reached plateau, more mononuclear cells, including CD4+ T lymphocytes was retrieved from spinal cord of aged than young rats. The frequencies of IL-17+ and GM-CSF+ cells within spinal cord infiltrating CD4+ lymphocytes were greater in aged rats. To their increased frequency contributed the expansion of GM-CSF + IL-17 + IFN-γ+ cells, which are highly pathogenic in mice. The expression of the cytokines (IL-1β and IL-23/p19) driving GM-CSF + IL-17 + IFN-γ + cell differentiation in mice was also augmented in aged rat spinal cord mononuclear cells. Additionally, in aged rat spinal cord the expansion of GM-CSF + IL-17-IFN-γ- CD4+ T lymphocytes was found. Consistently, the expression of mRNAs for IL-3, the cytokine exhibiting the same expression pattern as GM-CSF, and IL-7, the cytokine driving differentiation of GM-CSF + IL-17-IFN-γ- CD4 + lymphocytes in mice, was upregulated in aged rat spinal cord mononuclear cells, and the tissue, respectively. This was in accordance with the enhanced generation of the brain antigen-specific GM-CSF+ CD4+ lymphocytes in aged rat draining lymph nodes, as suggested by (i) the higher frequency of GM-CSF+ cells (reflecting the expansion of IL-17-IFN-γ- cells) within their CD4+ lymphocytes and (ii) the upregulated GM-CSF and IL-3 mRNA expression in fresh CD4+ lymphocytes and MBP-stimulated draining lymph node cells and IL-7 mRNA in lymph node tissue from aged rats. In agreement with the upregulated GM-CSF expression in aged rats, strikingly more CD11b + CD45(int) (activated microglia) and CD45(hi) (mainly proinflammatory dendritic cells and macrophages) cells was retrieved from aged than young rat spinal cord. Besides, expression of mRNA for SOCS1, a negative regulator of proinflammatory cytokine expression in innate immunity cells, was downregulated in aged rat spinal cord mononuclear cells. CONCLUSIONS The study revealed that aging may overcome genetic resistance to EAE, and indicated the cellular and molecular mechanisms contributing to this phenomenon in AO rats.
Collapse
Affiliation(s)
- Zorica Stojić-Vukanić
- Department of Microbiology and Immunology, Faculty of Pharmacy, University of Belgrade, 450 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Mirjana Nacka-Aleksić
- Department of Physiology, Faculty of Pharmacy, University of Belgrade, 450 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Ivan Pilipović
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", 458 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Ivana Vujnović
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", 458 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Veljko Blagojević
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", 458 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Duško Kosec
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", 458 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Mirjana Dimitrijević
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", 458 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Gordana Leposavić
- Department of Physiology, Faculty of Pharmacy, University of Belgrade, 450 Vojvode Stepe, 11221 Belgrade, Serbia
| |
Collapse
|
36
|
Seo JE, Hasan M, Han JS, Kang MJ, Jung BH, Kwok SK, Kim HY, Kwon OS. Experimental autoimmune encephalomyelitis and age-related correlations of NADPH oxidase, MMP-9, and cell adhesion molecules: The increased disease severity and blood–brain barrier permeability in middle-aged mice. J Neuroimmunol 2015; 287:43-53. [DOI: 10.1016/j.jneuroim.2015.08.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 08/04/2015] [Indexed: 12/18/2022]
|
37
|
Djikić J, Nacka-Aleksić M, Pilipović I, Kosec D, Arsenović-Ranin N, Stojić-Vukanić Z, Dimitrijević M, Leposavić G. Age-related changes in spleen of Dark Agouti rats immunized for experimental autoimmune encephalomyelitis. J Neuroimmunol 2014; 278:123-35. [PMID: 25595261 DOI: 10.1016/j.jneuroim.2014.12.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 12/16/2014] [Accepted: 12/17/2014] [Indexed: 12/11/2022]
Abstract
The study was undertaken considering age-related changes in susceptibility to experimental autoimmune encephalomyelitis (EAE) and a putative role of spleen in pathogenesis of this disease. The phenotypic and functional characteristics of T splenocytes were examined in young (3-month-old), middle-aged (8-month-old) and aged (26-month-old) Dark Agouti rats immunized for EAE with rat spinal cord in complete Freund's adjuvant. The rat susceptibility to EAE induction, as well as the number of activated CD4+CD134+ lymphocytes retrieved from their spinal cords progressively decreased with aging. To the contrary, in rats immunized for EAE the number of activated CD4+ splenocytes, i.e., CD4+CD134+, CD4+CD25+FoxP3- and CD4+CD40L+ cells, progressively increased with aging. This was associated with age-related increase in (i) CD4+ splenocyte surface expression of CD44, the molecule suggested to be involved in limiting emigration of encephalitogenic CD4+ cells from spleen into blood and (ii) frequency of regulatory T cells, including CD4+CD25+FoxP3+ cells, which are also shown to control encephalitogenic cell migration from spleen into the central nervous system. In favor of expansion of T-regulatory cell pool in aged rats was the greater concentration of IL-10 in unstimulated, Concanavalin A (ConA)- and myelin basic protein (MBP)-stimulated splenocyte cultures from aged rats compared with the corresponding cultures from young ones. Consistent with the age-related increase in the expression of CD44, which is shown to favor Th1 effector cell survival by interfering with CD95-mediated signaling, the frequency of apoptotic cells among CD4+ splenocytes, despite the greater frequency of CD95+ cells, was diminished in splenocyte cultures from aged compared with young rats. In addition, in control, as well as in ConA- and MBP-stimulated splenocyte cultures from aged rats, despite of impaired CD4+ cell proliferation, IFN-γ concentrations were greater than in corresponding cultures from young rats. This most likely reflected increased abundance of IFN-γ-producing cells in splenocyte cultures from aged compared with young rats. The diminished CD4+ cell proliferation in response to ConA and MBP in splenocyte cultures from aged compared with young rats could be, at least partly, associated with an enhanced splenic expression of iNOS mRNA in aged rats. Thus, the study suggests that age-associated changes leading to entrapping of activated CD4+ cells in the spleen could contribute to the restriction in development of EAE in aged rats.
Collapse
Affiliation(s)
- Jasmina Djikić
- Department of Physiology, Faculty of Pharmacy, University of Belgrade, 450 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Mirjana Nacka-Aleksić
- Department of Physiology, Faculty of Pharmacy, University of Belgrade, 450 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Ivan Pilipović
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", 458 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Duško Kosec
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", 458 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Nevena Arsenović-Ranin
- Department of Microbiology and Immunology, Faculty of Pharmacy, University of Belgrade, 450 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Zorica Stojić-Vukanić
- Department of Microbiology and Immunology, Faculty of Pharmacy, University of Belgrade, 450 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Mirjana Dimitrijević
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", 458 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Gordana Leposavić
- Department of Physiology, Faculty of Pharmacy, University of Belgrade, 450 Vojvode Stepe, 11221 Belgrade, Serbia.
| |
Collapse
|
38
|
Age-associated changes in rat immune system: Lessons learned from experimental autoimmune encephalomyelitis. Exp Gerontol 2014; 58:179-97. [DOI: 10.1016/j.exger.2014.08.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 07/24/2014] [Accepted: 08/12/2014] [Indexed: 01/15/2023]
|
39
|
Miller DH, Fazekas F, Montalban X, Reingold SC, Trojano M. Pregnancy, sex and hormonal factors in multiple sclerosis. Mult Scler 2014; 20:527-36. [PMID: 24446387 PMCID: PMC5458871 DOI: 10.1177/1352458513519840] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 12/04/2013] [Indexed: 01/17/2023]
Abstract
BACKGROUND Multiple sclerosis (MS) is influenced by pregnancy, sex and hormonal factors. OBJECTIVES A comprehensive understanding of the role of pregnancy, sex and hormonal factors can provide insights into disease mechanisms, and new therapeutic developments and can provide improved patient care and treatment. METHODS Based on an international conference of experts and a comprehensive PubMed search for publications on these areas in MS, we provide a review of what is known about the impact of these factors on disease demographics, etiology, pathophysiology and clinical course and outcomes. RESULTS AND CONCLUSIONS Recommendations are provided for counseling and management of people with MS before conception, during pregnancy and after delivery. The use of disease-modifying and symptomatic therapies in pregnancy is problematic and such treatments are normally discontinued. Available knowledge about the impact of treatment on the mother, fetus and newborn is discussed. Recommendations for future research to fill knowledge gaps and clarify inconsistencies in available data are made.
Collapse
Affiliation(s)
- David H Miller
- Queen Square Multiple Sclerosis Centre, Department of Neuroinflammation, Institute of Neurology, University College London, UK
| | - Franz Fazekas
- Department of Neurology, Medical University of Graz, Austria
| | - Xavier Montalban
- Department of Neurology/Neuroimmunology, MS Center of Catalonia (Cemcat), Vall d’Hebron University Hospital and Research Institute, Spain
| | | | - Maria Trojano
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari, Italy
| |
Collapse
|
40
|
Abstract
Abstract: Targeted immunotherapies represent a valid strategy for the treatment of metastatic castrate-resistant prostate cancer. A randomized, double-blind, Phase II clinical trial of PROSTVAC® demonstrated a statistically significant improvement in overall survival and a large, global, Phase III trial with overall survival as the primary end point is ongoing. PROSTVAC immunotherapy contains the transgenes for prostate-specific antigen and three costimulatory molecules (designated TRICOM). Research suggests that PROSTVAC not only targets prostate-specific antigen, but also other tumor antigens via antigen cascade. PROSTVAC is well tolerated and has been safely combined with other cancer therapies, including hormonal therapy, radiotherapy, another immunotherapy and chemotherapy. Even greater benefits of PROSTVAC may be recognized in earlier-stage disease and low-disease burden settings where immunotherapy can trigger a long-lasting immune response.
Collapse
Affiliation(s)
- Neal D Shore
- Carolina Urologic Research Center, 823 82nd Parkway, Myrtle Beach, SC 29572, USA
| |
Collapse
|
41
|
Ghandour S, Voskuhl R, Schumacher M. The androgen receptor as a therapeutic target for myelin repair in demyelinating diseases. Expert Rev Endocrinol Metab 2014; 9:5-7. [PMID: 30743734 DOI: 10.1586/17446651.2014.861740] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Steroid hormones exert major influences on the development and functioning of the nervous system, extending well beyond their reproductive effects. There is now also strong experimental evidence for an important role of these hormones in myelin formation. The recent finding that testosterone, via the intracellular androgen receptor, promotes myelin repair, may inspire neurobiologists to take a closer look at this hormone. It also opens new therapeutic opportunities for androgen receptor ligands in the treatment of myelin disorders.
Collapse
Affiliation(s)
- Said Ghandour
- c Biopathology of Myelin, Neuroprotection and Therapeutic Strategies, INSERM U 1199, University of Strasbourg, France
| | - Rhonda Voskuhl
- a University of California, Jack Skirball Chair in MS Research, 635 Charles E. Young Drive South, Los Angeles, CA 90095, USA
| | - Michael Schumacher
- b Neuroprotection and Neuroregeneration: Neuroactive Small Molecules, UMR 788 Inserm and University Paris-Sud, France, 94276 Kremlin-Bicêtre, France
| |
Collapse
|
42
|
Traveled distance is a sensitive and accurate marker of motor dysfunction in a mouse model of multiple sclerosis. ISRN NEUROSCIENCE 2013; 2013:170316. [PMID: 24967302 PMCID: PMC4045566 DOI: 10.1155/2013/170316] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 11/14/2013] [Indexed: 12/20/2022]
Abstract
Multiple sclerosis (MS) is a common central nervous system disease associated with progressive physical impairment. To study the mechanisms of the disease, we used experimental autoimmune encephalomyelitis (EAE), an animal model of MS. EAE is induced by myelin oligodendrocyte glycoprotein35–55 peptide, and the severity of paralysis in the disease is generally measured using the EAE score. Here, we compared EAE scores and traveled distance using the open-field test for an assessment of EAE progression. EAE scores were obtained with a 6-step observational scoring system for paralysis, and the traveled distance was obtained by automatic trajectory analysis of natural exploratory behaviors detected by a computer. The traveled distance of the EAE mice started to decrease significantly at day 7 of the EAE process, when the EAE score still did not reflect a change. Moreover, in the relationship between the traveled distance and paralysis as measured by the EAE score after day 14, there was a high coefficient of determination between the distance and the score. The results suggest that traveled distance is a sensitive marker of motor dysfunction in the early phases of EAE progression and that it reflects the degree of motor dysfunction after the onset of paralysis in EAE.
Collapse
|
43
|
Therapeutic testosterone administration preserves excitatory synaptic transmission in the hippocampus during autoimmune demyelinating disease. J Neurosci 2012; 32:12312-24. [PMID: 22956822 DOI: 10.1523/jneurosci.2796-12.2012] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Over 50% of multiple sclerosis (MS) patients experience cognitive deficits, and hippocampal-dependent memory impairment has been reported in >30% of these patients. While postmortem pathology studies and in vivo magnetic resonance imaging demonstrate that the hippocampus is targeted in MS, the neuropathology underlying hippocampal dysfunction remains unknown. Furthermore, there are no treatments available to date to effectively prevent neurodegeneration and associated cognitive dysfunction in MS. We have recently demonstrated that the hippocampus is also targeted in experimental autoimmune encephalomyelitis (EAE), the most widely used animal model of MS. The objective of this study was to assess whether a candidate treatment (testosterone) could prevent hippocampal synaptic dysfunction and underlying pathology when administered in either a preventative or a therapeutic (postdisease induction) manner. Electrophysiological studies revealed impairments in basal excitatory synaptic transmission that involved both AMPA receptor-mediated changes in synaptic currents, and faster decay rates of NMDA receptor-mediated currents in mice with EAE. Neuropathology revealed atrophy of the pyramidal and dendritic layers of hippocampal CA1, decreased presynaptic (Synapsin-1) and postsynaptic (postsynaptic density 95; PSD-95) staining, diffuse demyelination, and microglial activation. Testosterone treatment administered either before or after disease induction restores excitatory synaptic transmission as well as presynaptic and postsynaptic protein levels within the hippocampus. Furthermore, cross-modality correlations demonstrate that fluctuations in EPSPs are significantly correlated to changes in postsynaptic protein levels and suggest that PSD-95 is a neuropathological substrate to impaired synaptic transmission in the hippocampus during EAE. This is the first report demonstrating that testosterone is a viable therapeutic treatment option that can restore both hippocampal function and disease-associated pathology that occur during autoimmune disease.
Collapse
|
44
|
Tatar C, Bessert D, Tse H, Skoff RP. Determinants of central nervous system adult neurogenesis are sex, hormones, mouse strain, age, and brain region. Glia 2012; 61:192-209. [PMID: 23027402 DOI: 10.1002/glia.22426] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Accepted: 08/30/2012] [Indexed: 11/11/2022]
Abstract
Multiple sclerosis is a sexually dimorphic (SD) disease that causes oligodendrocyte death, but SD of glial cells is poorly studied. Here, we analyze SD of neural progenitors in 6-8 weeks and 6-8 months normal C57BL/6, SJL/J, and BALB/c mice in the subventricular zone (SVZ), dorsolateral horn (DLC), corpus callosum (CC), and parenchyma. With a short 2-h bromodeoxyuridine (BrdU) pulse, no gender and strain differences are present at 6-8 weeks. At 6-8 months, the number of BrdU(+) cells decreases twofold in each sex, strain, and region, indicating that a common aging mechanism regulates BrdU incorporation. Strikingly, 2× more BrdU(+) cells are found in all brain regions in 6-8 months C57BL/6 females versus males, no gender differences in 6-8 months SJL/J, and fewer BrdU(+) cells in females versus males in BALB/cs. The number of BrdU(+) cells modestly fluctuates throughout the estrous cycle in C57BL/6 and SJLs. Castration causes a dramatic increase in BrdU(+) cells in SVZ and DLC. These findings indicate that testosterone is a major regulator of adult neural proliferation. At 6-8 months, the ratio of PDGFRα(+) cells in the CC to BrdU(+) cells in the DLC of both strains, sexes, estrous cycle, and castrated mice was essentially the same, suggesting that BrdU(+) cells in the DLC differentiate into CC oligodendrocytes. The ratio of TUNEL(+) to BrdU(+) cells does not match proliferation, indicating that these events are differentially regulated. Differential regulation of these two processes leads to the variation in glial numbers between gender and strain. Explanations of neural proliferation based upon data from one sex or strain may be very misleading.
Collapse
Affiliation(s)
- Carrie Tatar
- Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, Michigan 48201, USA
| | | | | | | |
Collapse
|
45
|
Giatti S, Caruso D, Boraso M, Abbiati F, Ballarini E, Calabrese D, Pesaresi M, Rigolio R, Santos-Galindo M, Viviani B, Cavaletti G, Garcia-Segura LM, Melcangi RC. Neuroprotective effects of progesterone in chronic experimental autoimmune encephalomyelitis. J Neuroendocrinol 2012; 24:851-61. [PMID: 22283602 DOI: 10.1111/j.1365-2826.2012.02284.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Observations so far obtained in experimental autoimmune encephalomyelitis (EAE) have revealed the promising neuroprotective effects exerted by progesterone (PROG). The findings suggest that this neuroactive steroid may potentially represent a therapeutic tool for multiple sclerosis (MS). However, up to now, the efficacy of PROG has been only tested in the acute phase of the disease, whereas it is well known that MS expresses different features depending on the phase of the disease. Accordingly, we have evaluated the effect of PROG treatment in EAE induced in Dark Agouti rats (i.e. an experimental model showing a protracted relapsing EAE). Data obtained 45 days after EAE induction show that PROG treatment exerts a beneficial effect on clinical score, confirming surrogate parameters of spinal cord damage in chronic EAE (i.e. reactive microglia, cytokine levels, activity of the Na(+) ,K(+) -ATPase pump and myelin basic protein expression). An increase of the levels of dihydroprogesterone and isopregnanolone (i.e. two PROG metabolites) was also observed in the spinal cord after PROG treatment. Taken together, these results indicate that PROG is effective in reducing the severity of chronic EAE and, consequently, may have potential with respect to MS treatment.
Collapse
Affiliation(s)
- S Giatti
- Department of Endocrinology, Pathophysiology and Applied Biology, Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Milano, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Spence RD, Voskuhl RR. Neuroprotective effects of estrogens and androgens in CNS inflammation and neurodegeneration. Front Neuroendocrinol 2012; 33:105-15. [PMID: 22209870 PMCID: PMC3616506 DOI: 10.1016/j.yfrne.2011.12.001] [Citation(s) in RCA: 170] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Revised: 12/12/2011] [Accepted: 12/15/2011] [Indexed: 12/16/2022]
Abstract
Multiple sclerosis (MS) is a disease characterized by inflammation and demyelination. Currently, the cause of MS is unknown. Experimental autoimmune encephalomyelitis (EAE) is the most common mouse model of MS. Treatments with the sex hormones, estrogens and androgens, are capable of offering disease protection during EAE and are currently being used in clinical trials of MS. Beyond endogenous estrogens and androgens, treatments with selective estrogen receptor modulators (SERMs) for estrogen receptor alpha (ERα) and estrogen receptor beta (ERβ) are also capable of providing disease protection. This protection includes, but is not limited to, prevention of clinical disease, reduction of CNS inflammation, protection against demyelination, and protection against axonal loss. In EAE, current efforts are focused on using conditional cell specific knockouts of sex hormone receptors to identify the in vivo targets of these estrogens and androgens as well as downstream molecules responsible for disease protection.
Collapse
Affiliation(s)
- Rory D Spence
- University of California, Los Angeles, Department of Neurology, UCLA Multiple Sclerosis Program, 635 Charles E Young Drive South, Neuroscience Research Building 1, Room 479, Los Angeles, CA 90095, United States.
| | | |
Collapse
|
47
|
Kipp M, van der Star B, Vogel DYS, Puentes F, van der Valk P, Baker D, Amor S. Experimental in vivo and in vitro models of multiple sclerosis: EAE and beyond. Mult Scler Relat Disord 2011; 1:15-28. [PMID: 25876447 DOI: 10.1016/j.msard.2011.09.002] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Accepted: 09/05/2011] [Indexed: 12/16/2022]
Abstract
Although the primary cause of multiple sclerosis (MS) is unknown, the widely accepted view is that aberrant (auto)immune responses possibly arising following infection(s) are responsible for the destructive inflammatory demyelination and neurodegeneration in the central nervous system (CNS). This notion, and the limited access of human brain tissue early in the course of MS, has led to the development of autoimmune, viral and toxin-induced demyelination animal models as well as the development of human CNS cell and organotypic brain slice cultures in an attempt to understand events in MS. The autoimmune models, collectively known as experimental autoimmune encephalomyelitis (EAE), and viral models have shaped ideas of how environmental factors may trigger inflammation, demyelination and neurodegeneration in the CNS. Understandably, these models have also heavily influenced the development of therapies targeting the inflammatory aspect of MS. Demyelination and remyelination in the absence of overt inflammation are better studied in toxin-induced demyelination models using cuprizone and lysolecithin. The paradigm shift of MS as an autoimmune disease of myelin to a neurodegenerative disease has required more appropriate models reflecting the axonal and neuronal damage. Thus, secondary progressive EAE and spastic models have been crucial to develop neuroprotective approaches. In this review the current in vivo and in vitro experimental models to examine pathological mechanisms involved in inflammation, demyelination and neuronal degeneration, as well as remyelination and repair in MS are discussed. Since this knowledge is the basis for the development of new therapeutic approaches for MS, we particularly address whether the currently available models truly reflect the human disease, and discuss perspectives to further optimise and develop more suitable experimental models to study MS.
Collapse
Affiliation(s)
- Markus Kipp
- Department of Pathology, VU University Medical Centre, PO Box 7057, 1007 MB Amsterdam, The Netherlands; Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Baukje van der Star
- Department of Pathology, VU University Medical Centre, PO Box 7057, 1007 MB Amsterdam, The Netherlands
| | - Daphne Y S Vogel
- Department of Pathology, VU University Medical Centre, PO Box 7057, 1007 MB Amsterdam, The Netherlands; Department of Molecular Cell Biology and Immunology, VU University Medical Centre, Amsterdam, The Netherlands
| | - Fabìola Puentes
- Neuroimmunology Unit, Blizard Institute, Queen Mary University of London, Barts and The London School of Medicine and Dentistry, London, UK
| | - Paul van der Valk
- Department of Pathology, VU University Medical Centre, PO Box 7057, 1007 MB Amsterdam, The Netherlands
| | - David Baker
- Neuroimmunology Unit, Blizard Institute, Queen Mary University of London, Barts and The London School of Medicine and Dentistry, London, UK
| | - Sandra Amor
- Department of Pathology, VU University Medical Centre, PO Box 7057, 1007 MB Amsterdam, The Netherlands; Neuroimmunology Unit, Blizard Institute, Queen Mary University of London, Barts and The London School of Medicine and Dentistry, London, UK.
| |
Collapse
|
48
|
|
49
|
Lin AA, Wojciechowski SE, Hildeman DA. Androgens suppress antigen-specific T cell responses and IFN-γ production during intracranial LCMV infection. J Neuroimmunol 2010; 226:8-19. [PMID: 20619904 DOI: 10.1016/j.jneuroim.2010.05.026] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2008] [Revised: 05/06/2010] [Accepted: 05/06/2010] [Indexed: 10/19/2022]
Abstract
Intracranial (i.c.) lymphocytic choriomeningitis virus (LCMV) infection of mice results in T cell-driven anorexia and weight loss, which is diminished in males compared to females. We investigated sex-specific effects on antigen-presenting cells (APCs) and T cells after i.c. LCMV infection. Numbers of LCMV-specific T cells, APC activation, and levels of inflammatory cytokines and chemokines in CSF were decreased in males compared to females. Orchidectomy enhanced these immune parameters in males, while dihydrotestosterone treatment of orchidectomized males and intact females decreased some of these parameters. These data suggest that qualitative and quantitative effects of androgens on APCs and T cells may contribute to the well-known, but poorly understood sex differences in immunity and autoimmunity.
Collapse
Affiliation(s)
- Adora A Lin
- Division of Immunobiology, Cincinnati Children's Hospital, 3333 Burnet Ave., MLC 7038, Cincinnati, OH 45229, USA
| | | | | |
Collapse
|
50
|
Cosimo Melcangi R, Garcia-Segura LM. Sex-specific therapeutic strategies based on neuroactive steroids: In search for innovative tools for neuroprotection. Horm Behav 2010; 57:2-11. [PMID: 19524584 DOI: 10.1016/j.yhbeh.2009.06.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2009] [Revised: 06/02/2009] [Accepted: 06/04/2009] [Indexed: 02/07/2023]
Abstract
Different pathologies of the central and peripheral nervous system show sex differences in their incidence, symptomatology and/or neurodegenerative outcome. These include Parkinson's disease, Alzheimer's disease, Huntington's disease, multiple sclerosis, traumatic brain injury, stroke, autism, schizophrenia, depression, anxiety disorders, eating disorders and peripheral neuropathy. These sex differences reveal the need for sex-specific neuroprotective strategies. This review article and other manuscripts published in this issue of Hormones and Behavior analyze possible sex-specific therapeutic strategies based on neuroactive steroids. In particular in our introductory article, the possibility that sex differences in the levels or in the action of neuroactive steroids may represent causative factors for sex differences in the incidence or manifestation of pathologies of the nervous system is considered.
Collapse
Affiliation(s)
- Roberto Cosimo Melcangi
- Department of Endocrinology, Pathophysiology and Applied Biology, Center of Excellence on Neurodegenerative Diseases, University of Milan, Milano, Italy.
| | | |
Collapse
|