1
|
Zeigler G, Harrington CA, Rosendale N, Ganos C, Roldan V, Pace A, Alick-Lindstrom S, Orozco-Poore C, Deeb W, Hansen ML, L'Erario ZP. Neurologic Care for Transgender and Gender-Diverse People: A Review of Current Evidence and Clinical Implications. Neurol Clin Pract 2024; 14:e200332. [PMID: 38919931 PMCID: PMC11195436 DOI: 10.1212/cpj.0000000000200332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 04/11/2024] [Indexed: 06/27/2024]
Abstract
Purpose of Review To summarize the literature on neurologic care for transgender and gender-diverse (TGD) people and provide implications for clinical practice. Recent Findings There are limited data on the frequency and management of neurologic conditions among TGD people. TGD people have a higher prevalence of various neurologic conditions compared with cisgender or general population cohorts, including migraine, subjective cognitive decline, sleep disturbances, functional disorders, and cerebrovascular disease. Gender-affirming hormone therapy interacts with commonly prescribed neurologic medications and increases stroke risk among transfeminine people. Sex hormones and sex chromosomes may play a role in neurodegeneration and disability progression in neuroimmunologic diseases. Clitoral reduction surgeries on intersex children can cause neurologic disability and sexual dysfunction in adulthood. Socioeconomic disparities among TGD people contribute to health care barriers. Summary Neurologists should consider the unique experiences and health care needs of TGD people in their clinical practice and research protocols.
Collapse
Affiliation(s)
- Gwen Zeigler
- Department of Neurology (GZ), Albany Medical College, NY; Department of Neurology (CAH), The Ohio State University College of Medicine, Columbus; Department of Neurology and Weill Institute for Neurosciences (NR), University of California San Francisco; Movement Disorder Clinic (CG), Edmond J. Safra Program in Parkinson's Disease, Division of Neurology, University of Toronto, Toronto Western Hospital, Ontario, Canada; Facultad de Medicina Alberto Hurtado (VR), Universidad Peruana Cayetano Heredia, Lima, Peru; Department of Neurology (AP), Icahn School of Medicine at Mount Sinai, New York, NY; Department of Neurology (SA-L), University of Texas Southwestern Medical Center, Dallas; Department of Pediatric Neurology (CO-P), University of California, Los Angeles; Department of Neurology (WD), UMass Memorial Medical Center and UMass Medical School, Worcester; The Ohio State University Wexner Medical Center (MLH), Columbus; Greenburgh Pride (ZPLE), Westchester, NY
| | - Cole A Harrington
- Department of Neurology (GZ), Albany Medical College, NY; Department of Neurology (CAH), The Ohio State University College of Medicine, Columbus; Department of Neurology and Weill Institute for Neurosciences (NR), University of California San Francisco; Movement Disorder Clinic (CG), Edmond J. Safra Program in Parkinson's Disease, Division of Neurology, University of Toronto, Toronto Western Hospital, Ontario, Canada; Facultad de Medicina Alberto Hurtado (VR), Universidad Peruana Cayetano Heredia, Lima, Peru; Department of Neurology (AP), Icahn School of Medicine at Mount Sinai, New York, NY; Department of Neurology (SA-L), University of Texas Southwestern Medical Center, Dallas; Department of Pediatric Neurology (CO-P), University of California, Los Angeles; Department of Neurology (WD), UMass Memorial Medical Center and UMass Medical School, Worcester; The Ohio State University Wexner Medical Center (MLH), Columbus; Greenburgh Pride (ZPLE), Westchester, NY
| | - Nicole Rosendale
- Department of Neurology (GZ), Albany Medical College, NY; Department of Neurology (CAH), The Ohio State University College of Medicine, Columbus; Department of Neurology and Weill Institute for Neurosciences (NR), University of California San Francisco; Movement Disorder Clinic (CG), Edmond J. Safra Program in Parkinson's Disease, Division of Neurology, University of Toronto, Toronto Western Hospital, Ontario, Canada; Facultad de Medicina Alberto Hurtado (VR), Universidad Peruana Cayetano Heredia, Lima, Peru; Department of Neurology (AP), Icahn School of Medicine at Mount Sinai, New York, NY; Department of Neurology (SA-L), University of Texas Southwestern Medical Center, Dallas; Department of Pediatric Neurology (CO-P), University of California, Los Angeles; Department of Neurology (WD), UMass Memorial Medical Center and UMass Medical School, Worcester; The Ohio State University Wexner Medical Center (MLH), Columbus; Greenburgh Pride (ZPLE), Westchester, NY
| | - Christos Ganos
- Department of Neurology (GZ), Albany Medical College, NY; Department of Neurology (CAH), The Ohio State University College of Medicine, Columbus; Department of Neurology and Weill Institute for Neurosciences (NR), University of California San Francisco; Movement Disorder Clinic (CG), Edmond J. Safra Program in Parkinson's Disease, Division of Neurology, University of Toronto, Toronto Western Hospital, Ontario, Canada; Facultad de Medicina Alberto Hurtado (VR), Universidad Peruana Cayetano Heredia, Lima, Peru; Department of Neurology (AP), Icahn School of Medicine at Mount Sinai, New York, NY; Department of Neurology (SA-L), University of Texas Southwestern Medical Center, Dallas; Department of Pediatric Neurology (CO-P), University of California, Los Angeles; Department of Neurology (WD), UMass Memorial Medical Center and UMass Medical School, Worcester; The Ohio State University Wexner Medical Center (MLH), Columbus; Greenburgh Pride (ZPLE), Westchester, NY
| | - Valeria Roldan
- Department of Neurology (GZ), Albany Medical College, NY; Department of Neurology (CAH), The Ohio State University College of Medicine, Columbus; Department of Neurology and Weill Institute for Neurosciences (NR), University of California San Francisco; Movement Disorder Clinic (CG), Edmond J. Safra Program in Parkinson's Disease, Division of Neurology, University of Toronto, Toronto Western Hospital, Ontario, Canada; Facultad de Medicina Alberto Hurtado (VR), Universidad Peruana Cayetano Heredia, Lima, Peru; Department of Neurology (AP), Icahn School of Medicine at Mount Sinai, New York, NY; Department of Neurology (SA-L), University of Texas Southwestern Medical Center, Dallas; Department of Pediatric Neurology (CO-P), University of California, Los Angeles; Department of Neurology (WD), UMass Memorial Medical Center and UMass Medical School, Worcester; The Ohio State University Wexner Medical Center (MLH), Columbus; Greenburgh Pride (ZPLE), Westchester, NY
| | - Anna Pace
- Department of Neurology (GZ), Albany Medical College, NY; Department of Neurology (CAH), The Ohio State University College of Medicine, Columbus; Department of Neurology and Weill Institute for Neurosciences (NR), University of California San Francisco; Movement Disorder Clinic (CG), Edmond J. Safra Program in Parkinson's Disease, Division of Neurology, University of Toronto, Toronto Western Hospital, Ontario, Canada; Facultad de Medicina Alberto Hurtado (VR), Universidad Peruana Cayetano Heredia, Lima, Peru; Department of Neurology (AP), Icahn School of Medicine at Mount Sinai, New York, NY; Department of Neurology (SA-L), University of Texas Southwestern Medical Center, Dallas; Department of Pediatric Neurology (CO-P), University of California, Los Angeles; Department of Neurology (WD), UMass Memorial Medical Center and UMass Medical School, Worcester; The Ohio State University Wexner Medical Center (MLH), Columbus; Greenburgh Pride (ZPLE), Westchester, NY
| | - Sasha Alick-Lindstrom
- Department of Neurology (GZ), Albany Medical College, NY; Department of Neurology (CAH), The Ohio State University College of Medicine, Columbus; Department of Neurology and Weill Institute for Neurosciences (NR), University of California San Francisco; Movement Disorder Clinic (CG), Edmond J. Safra Program in Parkinson's Disease, Division of Neurology, University of Toronto, Toronto Western Hospital, Ontario, Canada; Facultad de Medicina Alberto Hurtado (VR), Universidad Peruana Cayetano Heredia, Lima, Peru; Department of Neurology (AP), Icahn School of Medicine at Mount Sinai, New York, NY; Department of Neurology (SA-L), University of Texas Southwestern Medical Center, Dallas; Department of Pediatric Neurology (CO-P), University of California, Los Angeles; Department of Neurology (WD), UMass Memorial Medical Center and UMass Medical School, Worcester; The Ohio State University Wexner Medical Center (MLH), Columbus; Greenburgh Pride (ZPLE), Westchester, NY
| | - Casey Orozco-Poore
- Department of Neurology (GZ), Albany Medical College, NY; Department of Neurology (CAH), The Ohio State University College of Medicine, Columbus; Department of Neurology and Weill Institute for Neurosciences (NR), University of California San Francisco; Movement Disorder Clinic (CG), Edmond J. Safra Program in Parkinson's Disease, Division of Neurology, University of Toronto, Toronto Western Hospital, Ontario, Canada; Facultad de Medicina Alberto Hurtado (VR), Universidad Peruana Cayetano Heredia, Lima, Peru; Department of Neurology (AP), Icahn School of Medicine at Mount Sinai, New York, NY; Department of Neurology (SA-L), University of Texas Southwestern Medical Center, Dallas; Department of Pediatric Neurology (CO-P), University of California, Los Angeles; Department of Neurology (WD), UMass Memorial Medical Center and UMass Medical School, Worcester; The Ohio State University Wexner Medical Center (MLH), Columbus; Greenburgh Pride (ZPLE), Westchester, NY
| | - Wissam Deeb
- Department of Neurology (GZ), Albany Medical College, NY; Department of Neurology (CAH), The Ohio State University College of Medicine, Columbus; Department of Neurology and Weill Institute for Neurosciences (NR), University of California San Francisco; Movement Disorder Clinic (CG), Edmond J. Safra Program in Parkinson's Disease, Division of Neurology, University of Toronto, Toronto Western Hospital, Ontario, Canada; Facultad de Medicina Alberto Hurtado (VR), Universidad Peruana Cayetano Heredia, Lima, Peru; Department of Neurology (AP), Icahn School of Medicine at Mount Sinai, New York, NY; Department of Neurology (SA-L), University of Texas Southwestern Medical Center, Dallas; Department of Pediatric Neurology (CO-P), University of California, Los Angeles; Department of Neurology (WD), UMass Memorial Medical Center and UMass Medical School, Worcester; The Ohio State University Wexner Medical Center (MLH), Columbus; Greenburgh Pride (ZPLE), Westchester, NY
| | - Margaret L Hansen
- Department of Neurology (GZ), Albany Medical College, NY; Department of Neurology (CAH), The Ohio State University College of Medicine, Columbus; Department of Neurology and Weill Institute for Neurosciences (NR), University of California San Francisco; Movement Disorder Clinic (CG), Edmond J. Safra Program in Parkinson's Disease, Division of Neurology, University of Toronto, Toronto Western Hospital, Ontario, Canada; Facultad de Medicina Alberto Hurtado (VR), Universidad Peruana Cayetano Heredia, Lima, Peru; Department of Neurology (AP), Icahn School of Medicine at Mount Sinai, New York, NY; Department of Neurology (SA-L), University of Texas Southwestern Medical Center, Dallas; Department of Pediatric Neurology (CO-P), University of California, Los Angeles; Department of Neurology (WD), UMass Memorial Medical Center and UMass Medical School, Worcester; The Ohio State University Wexner Medical Center (MLH), Columbus; Greenburgh Pride (ZPLE), Westchester, NY
| | - Z Paige L'Erario
- Department of Neurology (GZ), Albany Medical College, NY; Department of Neurology (CAH), The Ohio State University College of Medicine, Columbus; Department of Neurology and Weill Institute for Neurosciences (NR), University of California San Francisco; Movement Disorder Clinic (CG), Edmond J. Safra Program in Parkinson's Disease, Division of Neurology, University of Toronto, Toronto Western Hospital, Ontario, Canada; Facultad de Medicina Alberto Hurtado (VR), Universidad Peruana Cayetano Heredia, Lima, Peru; Department of Neurology (AP), Icahn School of Medicine at Mount Sinai, New York, NY; Department of Neurology (SA-L), University of Texas Southwestern Medical Center, Dallas; Department of Pediatric Neurology (CO-P), University of California, Los Angeles; Department of Neurology (WD), UMass Memorial Medical Center and UMass Medical School, Worcester; The Ohio State University Wexner Medical Center (MLH), Columbus; Greenburgh Pride (ZPLE), Westchester, NY
| |
Collapse
|
2
|
Geng C, Tang Y. Association between levels of sex hormones and risk of multiple sclerosis: a mendelian randomization study. Acta Neurol Belg 2024:10.1007/s13760-024-02613-x. [PMID: 39073697 DOI: 10.1007/s13760-024-02613-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 07/13/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND This research aimed to examine the causal connections between multiple sclerosis (MS) and a range of sex hormone-related traits, such as bioavailable testosterone (BT), sex hormone-binding globulin (SHBG), testosterone, and estradiol (E2). METHODS A bidirectional two-sample Mendelian randomization (MR) analysis using summary statistics from genome-wide association studies (GWAS) was conducted to investigate the relationship between sex hormone-related traits and MS. Moreover, the Inverse-variance weighted (IVW) method was employed as the primary analysis approach. RESULTS The MR analysis, using the IVW method, found a significant correlation between genetically determined SHBG levels and MS (OR = 1.634, 95% CI: 1.029-2.599, p = 0.038). Similarly, the reverse MR analysis suggested a causal link between MS and SHBG (OR = 1.005, 95% CI: 1.001-1.009, P = 0.003). However, no association was observed between MS risk and E2, testosterone, or BT levels. CONCLUSION Our MR analysis demonstrated that genetically predicted higher SHBG may be positively correlated with the risk of MS. Moreover, the role of SHBG in MS could be further investigated.
Collapse
Affiliation(s)
- Chaofan Geng
- Department of Neurology and Innovation Center for Neurological Disorders, Xuanwu Hospital, National Center for Neurological Disorders, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Yi Tang
- Department of Neurology and Innovation Center for Neurological Disorders, Xuanwu Hospital, National Center for Neurological Disorders, Capital Medical University, 45 Changchun Street, Beijing, 100053, China.
- Neurodegenerative Laboratory of Ministry of Education of the People's Republic of China, Beijing, China.
| |
Collapse
|
3
|
Haghmorad D, Soltanmohammadi A, Jadid Tavaf M, Zargarani S, Yazdanpanah E, Shadab A, Yousefi B. The protective role of interaction between vitamin D, sex hormones and calcium in multiple sclerosis. Int J Neurosci 2024; 134:735-753. [PMID: 36369838 DOI: 10.1080/00207454.2022.2147431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 11/01/2022] [Accepted: 11/07/2022] [Indexed: 11/14/2022]
Abstract
Multiple sclerosis (MS) is a neurological disorder that causes disability and paralysis, especially among young adults. Although interactions of several factors, such as viral infections, autoimmunity, genetic and environmental factors, performance a role in the beginning and progression of the disease, the exact cause of MS is unknown to date. Different immune cells such as Th1 and Th17 play an impressive role in the immunopathogenesis of MS, while, regulatory cells such as Th2 and Treg diminish the severity of the illness. Sex hormones have a vital role in many autoimmune disorders, including multiple sclerosis. Testosterone, estrogen and progesterone have various roles in the progress of MS, which higher prevalence of disease in women and more severe in men reveals the importance of sex hormones' role in this disease. Vitamin D after chemical changes in the body, as an active hormone called calcitriol, plays an important role in regulating immune responses and improves MS by modulating the immune system. The optimum level of calcium in the body with vitamin D modulates immune responses and calcium as an essential ion in the body plays a key role in the treatment of autoimmune diseases. The interaction between vitamin D and sex hormones has protective and therapeutic effects against MS and functional synergy between estrogen and calcitriol occurs in disease recovery. Moreover, vitamin D and calcium interact with each other to regulate the immune system and shift them to anti-inflammatory responses.
Collapse
Affiliation(s)
- Dariush Haghmorad
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Azita Soltanmohammadi
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Maryam Jadid Tavaf
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Simin Zargarani
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Esmaeil Yazdanpanah
- Immunology Research Center, Department of Immunology and Allergy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Shadab
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Bahman Yousefi
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
4
|
Winschel I, Willing A, Engler JB, Walkenhorst M, Meurs N, Binkle-Ladisch L, Woo MS, Pfeffer LK, Sonner JK, Borgmeyer U, Hagen SH, Grünhagel B, Claussen JM, Altfeld M, Friese MA. Sex- and species-specific contribution of CD99 to T cell costimulation during multiple sclerosis. Biol Sex Differ 2024; 15:41. [PMID: 38750588 PMCID: PMC11097467 DOI: 10.1186/s13293-024-00618-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 05/07/2024] [Indexed: 05/18/2024] Open
Abstract
BACKGROUND Differences in immune responses between women and men are leading to a strong sex bias in the incidence of autoimmune diseases that predominantly affect women, such as multiple sclerosis (MS). MS manifests in more than twice as many women, making sex one of the most important risk factor. However, it is incompletely understood which genes contribute to sex differences in autoimmune incidence. To address that, we conducted a gene expression analysis in female and male human spleen and identified the transmembrane protein CD99 as one of the most significantly differentially expressed genes with marked increase in men. CD99 has been reported to participate in immune cell transmigration and T cell regulation, but sex-specific implications have not been comprehensively investigated. METHODS In this study, we conducted a gene expression analysis in female and male human spleen using the Genotype-Tissue Expression (GTEx) project dataset to identify differentially expressed genes between women and men. After successful validation on protein level of human immune cell subsets, we assessed hormonal regulation of CD99 as well as its implication on T cell regulation in primary human T cells and Jurkat T cells. In addition, we performed in vivo assays in wildtype mice and in Cd99-deficient mice to further analyze functional consequences of differential CD99 expression. RESULTS Here, we found higher CD99 gene expression in male human spleens compared to females and confirmed this expression difference on protein level on the surface of T cells and pDCs. Androgens are likely dispensable as the cause shown by in vitro assays and ex vivo analysis of trans men samples. In cerebrospinal fluid, CD99 was higher on T cells compared to blood. Of note, male MS patients had lower CD99 levels on CD4+ T cells in the CSF, unlike controls. By contrast, both sexes had similar CD99 expression in mice and Cd99-deficient mice showed equal susceptibility to experimental autoimmune encephalomyelitis compared to wildtypes. Functionally, CD99 increased upon human T cell activation and inhibited T cell proliferation after blockade. Accordingly, CD99-deficient Jurkat T cells showed decreased cell proliferation and cluster formation, rescued by CD99 reintroduction. CONCLUSIONS Our results demonstrate that CD99 is sex-specifically regulated in healthy individuals and MS patients and that it is involved in T cell costimulation in humans but not in mice. CD99 could potentially contribute to MS incidence and susceptibility in a sex-specific manner.
Collapse
Affiliation(s)
- Ingo Winschel
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anne Willing
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jan Broder Engler
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Mark Walkenhorst
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nina Meurs
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lars Binkle-Ladisch
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marcel S Woo
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lena Kristina Pfeffer
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jana K Sonner
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Uwe Borgmeyer
- Center of Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sven Hendrik Hagen
- Research Department Virus Immunology, Leibniz Institute of Virology, Hamburg, Germany
| | - Benjamin Grünhagel
- Research Department Virus Immunology, Leibniz Institute of Virology, Hamburg, Germany
| | - Janna M Claussen
- Research Department Virus Immunology, Leibniz Institute of Virology, Hamburg, Germany
| | - Marcus Altfeld
- Research Department Virus Immunology, Leibniz Institute of Virology, Hamburg, Germany
| | - Manuel A Friese
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
5
|
Hammad MAM, Rush A, Loeb CA, Banton J, Abou Chawareb E, Khanmammadova N, Gevorkyan RR, Barham DW, Yafi FA, Jenkins LC. Multiple sclerosis and hypogonadism: is there a relationship? Sex Med Rev 2024; 12:178-182. [PMID: 38185910 DOI: 10.1093/sxmrev/qead050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 10/21/2023] [Accepted: 10/26/2023] [Indexed: 01/09/2024]
Abstract
INTRODUCTION Multiple sclerosis (MS) is a chronic autoimmune disease that affects the central nervous system and is widely recognized as a disease primarily affecting women. The relationship between MS and hypogonadism is complex and not fully understood, with recent evidence showing that hypogonadism may have a significant impact on the quality of life and disease progression of patients with MS. OBJECTIVES This review aims to provide an overview of the current knowledge regarding the relationship between MS and hypogonadism, including the mechanisms underlying this relationship; the effects of hypogonadism on patients with MS; and the potential benefits and drawbacks of testosterone replacement therapy for patients with MS and hypogonadism. METHODS This scientific review analyzed 19 articles that investigated the potential relationship among MS, testosterone levels, and hypogonadism. The articles were published between November 2008 and March 2022 and were identified through a comprehensive search of the PubMed database. The search terms used included "multiple sclerosis," "testosterone," "hypogonadism," and "MS and testosterone levels." RESULTS Of the 19 articles reviewed, 11 described a positive correlation between low testosterone levels and dysfunction within the hypothalamic-pituitary-gonadal axis in individuals with MS. These findings suggest that low testosterone levels may contribute to dysfunction within the hypothalamus-pituitary-gonadal axis, which plays a crucial role in regulating testosterone production. The results also showed a relationship between sexual dysfunction and low testosterone levels, as well as a positive correlative relationship between these factors. CONCLUSION The reviewed articles indicate a complex relationship among MS, testosterone levels, and the hypothalamic-pituitary-gonadal axis, with low testosterone levels potentially contributing to dysfunction in this axis and to sexual dysfunction. Further research is needed to better understand the effects of testosterone therapy on MS and sexual dysfunction in patients with MS.
Collapse
Affiliation(s)
- Muhammed A M Hammad
- Department of Urology, University of California, Irvine, CA 92868, United States
| | - Adriana Rush
- School of Medicine Greenville, University of South Carolina, Greenville, SC 29605, United States
| | - Charles A Loeb
- Department of Urology, University of California, Irvine, CA 92868, United States
| | - Jasmin Banton
- College of Osteopathic Medicine, New York Institute of Technology, Jonesboro, Arkansas, NY 11545, United States
| | - Elia Abou Chawareb
- Department of Urology, University of California, Irvine, CA 92868, United States
| | | | - Rafael R Gevorkyan
- Department of Urology, University of California, Irvine, CA 92868, United States
| | - David W Barham
- Department of Urology, University of California, Irvine, CA 92868, United States
| | - Faysal A Yafi
- Department of Urology, University of California, Irvine, CA 92868, United States
| | - Lawrence C Jenkins
- Department of Urology, University of California, Irvine, CA 92868, United States
| |
Collapse
|
6
|
Asbelaoui N, Abi-Ghanem C, Schlecht-Louf G, Oukil H, Degerny C, Schumacher M, Ghoumari AM. Interplay between androgen and CXCR4 chemokine signaling in myelin repair. Acta Neuropathol Commun 2024; 12:18. [PMID: 38291527 PMCID: PMC10826258 DOI: 10.1186/s40478-024-01730-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 01/11/2024] [Indexed: 02/01/2024] Open
Abstract
In men, reduced levels of testosterone are associated with the prevalence and progression of multiple sclerosis (MS), a chronic and disabling demyelinating disorder. Testosterone has been shown to promote myelin repair. Here, we demonstrate that the cooperation between testosterone and CXCR4 signaling involving astrocytes is required for myelin regeneration after focal demyelination produced in the ventral mouse spinal cord by the infusion of lysolecithin. The testosterone-dependent remyelination of axons by oligodendrocytes was accompanied by an increase in astrocytes expressing CXCR4, its ligand CXCL12 and the androgen receptor (AR) within the demyelinated area. Depriving males of their testosterone or pharmacological inhibition of CXCR4, with the selective antagonist AMD3100, prevented the appearance of astrocytes expressing CXCR4, CXCL12 and AR within the demyelinated area and the concomitant recruitment of myelin forming oligodendrocytes. Conditional genetic ablation of either CXCR4 or AR in astrocytes also completely blocked the formation of new myelin by oligodendrocytes. Interestingly, the gain of function mutation in CXCR4 causing WHIM syndrome allows remyelination to take place, even in the absence of testosterone, but its potentiating effects remained observable. After testosterone deprivation or CXCR4 inhibition, the absence of astrocytes within the demyelinated area led to the incursion of Schwann cells, most likely derived from spinal nerves, and the formation of peripheral nerve type myelin. In patients with progressive MS, astrocytes expressing CXCR4 and AR surrounded myelin lesions, and their presence opposed the incursion of Schwann cells. These results highlight a mechanism of promyelinating testosterone signaling and the importance of normalizing its levels in combined myelin repair therapies.
Collapse
Affiliation(s)
- Narimène Asbelaoui
- UMR1195, "Diseases and Hormones of the Nervous System", Inserm and University Paris-Saclay, 80, Rue du Général Leclerc, 94276, Kremlin-Bicêtre, France
| | - Charly Abi-Ghanem
- UMR1195, "Diseases and Hormones of the Nervous System", Inserm and University Paris-Saclay, 80, Rue du Général Leclerc, 94276, Kremlin-Bicêtre, France
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, 12208, USA
| | - Géraldine Schlecht-Louf
- INSERM UMR 996, Inserm, Inflammation, Microbiome and Immunosurveillance, Faculté de Pharmacie, Université Paris-Saclay, Orsay, France
| | - Hania Oukil
- UMR1195, "Diseases and Hormones of the Nervous System", Inserm and University Paris-Saclay, 80, Rue du Général Leclerc, 94276, Kremlin-Bicêtre, France
| | - Cindy Degerny
- UMR1195, "Diseases and Hormones of the Nervous System", Inserm and University Paris-Saclay, 80, Rue du Général Leclerc, 94276, Kremlin-Bicêtre, France
| | - Michael Schumacher
- UMR1195, "Diseases and Hormones of the Nervous System", Inserm and University Paris-Saclay, 80, Rue du Général Leclerc, 94276, Kremlin-Bicêtre, France.
| | - Abdel Mouman Ghoumari
- UMR1195, "Diseases and Hormones of the Nervous System", Inserm and University Paris-Saclay, 80, Rue du Général Leclerc, 94276, Kremlin-Bicêtre, France.
| |
Collapse
|
7
|
Shayestehfar M, Salari M, Karimi S, Vosough M, Memari A, Nabavi SM. Sex hormone therapy in Multiple Sclerosis: A systematic review of randomized clinical trials. J Cent Nerv Syst Dis 2024; 16:11795735231223411. [PMID: 38188227 PMCID: PMC10768623 DOI: 10.1177/11795735231223411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 12/05/2023] [Indexed: 01/09/2024] Open
Abstract
Background In spite of the observed immunomodulatory properties of different sex hormones on Multiple Sclerosis (MS) in different investigations, to date, there has been no study to systematically review the documents to add more powerful data to the field. Objectives Therefore, in this paper we aim to systematically review clinical and randomized controlled trials (RCT) assessing the effect of sex hormone therapies on individuals with MS. Design A comprehensive search of electronic databases including PubMed, EMBASE, and Scopus was conducted. Clinical trials and RCTs that assessed the impact of sex hormones on individuals with MS were selected and included in the systematic review. Data sources and methods In the final phase of the search strategy, 9 papers reached the criteria for entering in the systematic review. Two independent reviewers extracted the relevant data from each article according to the standardized data extraction form. Two reviewers also assessed the quality of each study independently using PEDro scale. Results We categorized three different classifications of outcomes including clinical, MRI, and immune system findings and put each measured outcome in the category which matched best. Conclusion In conclusion, the existed investigations on the effect of sex hormones on inflammatory and neurodegenerative components of MS are promising particularly in relapsing-remitting MS (RRMS).
Collapse
Affiliation(s)
- Monir Shayestehfar
- Sports Medicine Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehri Salari
- Neuro Functional Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shahedeh Karimi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Tehran, Iran
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Tehran, Iran
| | - Amirhossein Memari
- Sports Medicine Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Massood Nabavi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Tehran, Iran
| |
Collapse
|
8
|
Hsu S, Bove R. Hormonal Therapies in Multiple Sclerosis: a Review of Clinical Data. Curr Neurol Neurosci Rep 2024; 24:1-15. [PMID: 38102502 PMCID: PMC10774191 DOI: 10.1007/s11910-023-01326-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/27/2023] [Indexed: 12/17/2023]
Abstract
PURPOSE OF REVIEW Given the potential for exogenous hormones to influence risk and course of MS, this narrative review aims to summarize current knowledge from observational and interventional studies of exogenous hormones in humans with MS. RECENT FINDINGS Large randomized clinical trials for combined oral contraceptives and estriol both show modest effect on inflammatory activity, with the latter showing potential neuroprotective effect. After fertility treatment, large actively treated cohorts have not confirmed any elevated risk of relapse. Preclinical data suggest that androgens, selective estrogen receptor modulators (SERMs), and selective androgen receptor modulators (SARMs) may be neuroprotective but clinical data are lacking. Gender affirming treatment, particularly estrogen in trans-women, could possibly be associated with elevated risk of inflammation. For women with MS entering menopause, hormone therapy appears safe during the appropriate menopausal window, but its long-term effects on neuroprotection are unknown. Exogenous hormones, used in varied doses and for diverse indications, have variable effects on MS risk, inflammatory activity, and neuroprotection. Large randomized trials are needed before it is possible to determine the true effect of exogenous hormones in a condition as complex as MS.
Collapse
Affiliation(s)
- Stephanie Hsu
- UCSF Weill Institute for Neuroscience, Division of Neuroimmunology and Glial Biology, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Riley Bove
- UCSF Weill Institute for Neuroscience, Division of Neuroimmunology and Glial Biology, Department of Neurology, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
9
|
Zhang S, Wang T, Feng Y, Li F, Qu A, Guan X, Wang H, Xu D. Pregnenolone 16α-carbonitrile negatively regulates hippocampal cytochrome P450 enzymes and ameliorates phenytoin-induced hippocampal neurotoxicity. J Pharm Anal 2023; 13:1510-1525. [PMID: 38223454 PMCID: PMC10785155 DOI: 10.1016/j.jpha.2023.07.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 07/18/2023] [Accepted: 07/19/2023] [Indexed: 01/16/2024] Open
Abstract
The central nervous system is susceptible to the modulation of various neurophysiological processes by the cytochrome P450 enzyme (CYP), which plays a crucial role in the metabolism of neurosteroids. The antiepileptic drug phenytoin (PHT) has been observed to induce neuronal side effects in patients, which could be attributed to its induction of CYP expression and testosterone (TES) metabolism in the hippocampus. While pregnane X receptor (PXR) is widely known for its regulatory function of CYPs in the liver, we have discovered that the treatment of mice with pregnenolone 16α-carbonitrile (PCN), a PXR agonist, has differential effects on CYP expression in the liver and hippocampus. Specifically, the PCN treatment resulted in the induction of cytochrome P450, family 3, subfamily a, polypeptide 11 (CYP3A11), and CYP2B10 expression in the liver, while suppressing their expression in the hippocampus. Functionally, the PCN treatment protected mice from PHT-induced hippocampal nerve injury, which was accompanied by the inhibition of TES metabolism in the hippocampus. Mechanistically, we found that the inhibition of hippocampal CYP expression and attenuation of PHT-induced neurotoxicity by PCN were glucocorticoid receptor dependent, rather than PXR independent, as demonstrated by genetic and pharmacological models. In conclusion, our study provides evidence that PCN can negatively regulate hippocampal CYP expression and attenuate PHT-induced hippocampal neurotoxicity independently of PXR. Our findings suggest that glucocorticoids may be a potential therapeutic strategy for managing the neuronal side effects of PHT.
Collapse
Affiliation(s)
- Shuai Zhang
- Department of Obstetric, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University, Wuhan, 430071, China
| | - Tingting Wang
- Department of Obstetric, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University, Wuhan, 430071, China
| | - Ye Feng
- Department of Endocrinology and Metabolic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Fei Li
- Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Aijuan Qu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
- Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing, 100069, China
| | - Xiuchen Guan
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, 100069, China
| | - Hui Wang
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University, Wuhan, 430071, China
| | - Dan Xu
- Department of Obstetric, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University, Wuhan, 430071, China
| |
Collapse
|
10
|
Wang H, Zhang X, Li H, Sun Z, Zhong Y. Gender differences in the burden of multiple sclerosis in China from 1990 to 2019 and its 25-year projection: An analysis of the Global Burden of Diseases Study. Health Sci Rep 2023; 6:e1738. [PMID: 38033712 PMCID: PMC10685393 DOI: 10.1002/hsr2.1738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 11/02/2023] [Accepted: 11/06/2023] [Indexed: 12/02/2023] Open
Abstract
Background and Aims Multiple sclerosis (MS) is a crippling, chronic, gender-related disease that causes burdens to individuals and society. China has a considerable and increasing population of MS. We aim to analyze the gender disparities in the burden of MS in China and predict the trends. Methods The study was conducted based on the Global Burden of Disease Study 2019. Data on incidence, prevalence, deaths, and disability-adjusted life years (DALYs) of MS in China from 1990 to 2019 was descriptively analyzed by year, gender, and age group. The Nordpred package in R (version 4.2.2) was used for age-period-cohort analysis to predict the all-ages numbers and age-standardized rates of incidence, prevalence, deaths, and DALYs in China from 2020 to 2044. Results The number of prevalent cases of MS in 2019 reached 18,143.56 (95% uncertainty intervals [UI]: 13,997.71-22,658.60) in males and 24,427.11 (95% UI: 18,906.02-30,530.21) in females in China. The peak age of prevalence was shifted from 40-44 years in 1990 to 45-49 years in 2019 in females but remained unchanged in males. In contrast to the increased age-standardized prevalence rate, the age-standardized death rate (ASDR) and age-standardized DALYs rate showed downward trends, which were more significant in females. Different from the global, Chinese males showed lower prevalence but higher deaths and DALYs than females for age-standardized rates and numbers. In the next 25 years, the patient population will remain large and peak around 44,599.78 in 2025-2029. The ASDR, age-standardized DALYs rate, and DALYs number were expected to decrease. The improvements in deaths and DALYs will be more significant in females. Conclusion Males with MS had a lower prevalence but higher deaths and DALYs than females in China. The ASDR and age-standardized DALYs rate have reduced over the past 30 years and were expected to continue decreasing, especially in females. The burden of MS will remain notable in the next 25 years.
Collapse
Affiliation(s)
- Heng Wang
- Department of Ophthalmology, Peking Union Medical College HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Xia Zhang
- Department of Ophthalmology, Peking Union Medical College HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Heyan Li
- Beijing Tongren Eye Center, Beijing Tongren HospitalCapital Medical UniversityBeijingChina
| | - Zixi Sun
- Department of Ophthalmology, Peking Union Medical College HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Yong Zhong
- Department of Ophthalmology, Peking Union Medical College HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| |
Collapse
|
11
|
Hosseinzadeh S, Afshari S, Molaei S, Rezaei N, Dadkhah M. The role of genetics and gender specific differences in neurodegenerative disorders: Insights from molecular and immune landscape. J Neuroimmunol 2023; 384:578206. [PMID: 37813041 DOI: 10.1016/j.jneuroim.2023.578206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/09/2023] [Accepted: 09/23/2023] [Indexed: 10/11/2023]
Abstract
Neurodegenerative disorders (NDDs) are the most common neurological disorders with high prevalence and have significant socioeconomic implications. Understanding the underlying cellular and molecular mechanisms associated with the immune system can be effective in disease etiology, leading to more effective therapeutic approaches for both females and males. The central nervous system (CNS) actively participates in immune responses, both within and outside the CNS. Immune system activation is a common feature in NDDs. Gender-specific factors play a significant role in the prevalence, progression, and manifestation of NDDs. Neuroinflammation, in both inflammatory neurological and neurodegenerative conditions, is defined by the triggering of microglia and astrocyte cell activation. This results in the secretion of pro-inflammatory cytokines and chemokines. Numerous studies have documented the role of neuroinflammation in neurological diseases, highlighting the involvement of immune signaling pathways in disease development. Converging evidence support immune system involvement during neurodegeneration in NDDs. In this review, we summarize emerging evidence that reveals gender-dependent differences in immune responses related to NDDs. Also, we highlight sex differences in immune responses and discuss how these sex-specific influences can increase the risk of NDDs. Understanding the role of gender-specific factors can aid in developing targeted therapeutic strategies and improving patient outcomes. Ultimately, the better understanding of these mechanisms contributed to sex-dependent immune response in NDDs, can be critically usful in targeting of immune signaling cascades in such disorders. In this regard, sex-related immune responses in NDDs may be promising and effective targets in therapeutic strategies.
Collapse
Affiliation(s)
- Shahnaz Hosseinzadeh
- Department of Microbiology & Immunology, School of Medicine, Ardabil University of Medical Sciences, Iran; Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Salva Afshari
- Students Research Committee, Pharmacy School, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Soheila Molaei
- Zoonoses Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center Hospital, Tehran University of Medical Sciences, Tehran 1419733151, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education Research Network (USERN), Tehran, Iran
| | - Masoomeh Dadkhah
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
12
|
Geng H, Ye Q, Lin X, Simpson-Yap S, Zhou Y, Tang D. Impact of multiple sclerosis on male sexual and reproductive health. Mult Scler Relat Disord 2023; 79:105059. [PMID: 37832256 DOI: 10.1016/j.msard.2023.105059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 10/03/2023] [Accepted: 10/07/2023] [Indexed: 10/15/2023]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory and autoimmune neurodegenerative disease characterized by the destruction of myelin in the central nervous system, leading to significant health and quality of life burdens for patients. MS is most prevalent in younger individuals aged 20-40, a critical period when many patients hope to establish relationships and start families. While neurological disability, such as fatigue, sensory dysfunction, spasticity, and cognitive dysfunction, have been greatly improved with the advances in managing MS, physicians are frequently confronted with sexual and reproductive problems among younger male people with MS (PwMS). These issues mainly include erectile dysfunction, ejaculatory disorders, reduced libido, decreased sperm quality, and impaired male fertility. Despite recent studies indicating that MS negatively impacts the sexuality and fertility of male PwMS, these issues have not received sufficient attention. Genetic factors, autoimmunity, chronic inflammation, psychological factors, and the use of drugs may contribute to sexual/reproductive dysfunction in PwMS. However, like the overall understanding of MS pathophysiology, the complete mechanisms of its development remain unclear. In this study, we review the existing literature to summarize the range of sexual and reproductive issues unique to males with MS, explore potential underlying mechanisms, and aim to improve these issues in male PwMS. By shedding light on this overlooked aspect of MS, we hope to enhance the care and well-being of male PwMS facing these challenges.
Collapse
Affiliation(s)
- Hao Geng
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Qinglin Ye
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Xin Lin
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Steve Simpson-Yap
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia; Neuroepidemiology Unit, Melbourne School of Population & Global Health, The University of Melbourne, Melbourne, Australia; CORe, School of Medicine, The University of Melbourne, Melbourne, Australia
| | - Yuan Zhou
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Dongdong Tang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia.
| |
Collapse
|
13
|
Fazazi MR, Ruda GF, Brennan PE, Rangachari M. The X-linked histone demethylases KDM5C and KDM6A as regulators of T cell-driven autoimmunity in the central nervous system. Brain Res Bull 2023; 202:110748. [PMID: 37657612 DOI: 10.1016/j.brainresbull.2023.110748] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 08/18/2023] [Accepted: 08/21/2023] [Indexed: 09/03/2023]
Abstract
T cell-driven autoimmune responses are subject to striking sex-dependent effects. While the contributions of sex hormones are well-understood, those of sex chromosomes are meeting with increased appreciation. Here, we outline what is known about the contribution of sex chromosome-linked factors to experimental autoimmune encephalomyelitis (EAE), a mouse model that recapitulates many of the T cell-driven mechanisms of multiple sclerosis (MS) pathology. Particular attention is paid to the KDM family of histone demethylases, several of which - KDM5C, KDM5D and KDM6A - are sex chromosome encoded. Finally, we provide evidence that functional inhibition of KDM5 molecules can suppress interferon (IFN)γ production from murine male effector T cells, and that an increased ratio of inflammatory Kdm6a to immunomodulatory Kdm5c transcript is observed in T helper 17 (Th17) cells from women with the autoimmune disorder ankylosing spondylitis (AS). Histone lysine demethlyases thus represent intriguing targets for the treatment of T cell-driven autoimmune disorders.
Collapse
Affiliation(s)
- Mohamed Reda Fazazi
- axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | - Gian Filippo Ruda
- Centre for Medicines Discovery and NIHR, Oxford Biomedical Research Centre, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, UK
| | - Paul E Brennan
- Centre for Medicines Discovery and NIHR, Oxford Biomedical Research Centre, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, UK; Alzheimer's Research UK, Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, UK
| | - Manu Rangachari
- axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Département de médecine moléculaire, Faculté de médecine, Université Laval, Québec, QC, Canada.
| |
Collapse
|
14
|
Meyer CE, Smith AW, Padilla-Requerey AA, Farkhondeh V, Itoh N, Itoh Y, Gao JL, Herbig PD, Nguyen Q, Ngo KH, Oberoi MR, Siddarth P, Voskuhl RR, MacKenzie-Graham A. Neuroprotection in Cerebral Cortex Induced by the Pregnancy Hormone Estriol. J Transl Med 2023; 103:100189. [PMID: 37245852 DOI: 10.1016/j.labinv.2023.100189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/12/2023] [Accepted: 05/19/2023] [Indexed: 05/30/2023] Open
Abstract
In multiple sclerosis (MS), demyelination occurs in the cerebral cortex, and cerebral cortex atrophy correlates with clinical disabilities. Treatments are needed in MS to induce remyelination. Pregnancy is protective in MS. Estriol is made by the fetoplacental unit, and maternal serum estriol levels temporally align with fetal myelination. Here, we determined the effect of estriol treatment on the cerebral cortex in the preclinical model of MS, experimental autoimmune encephalomyelitis (EAE). Estriol treatment initiated after disease onset decreased cerebral cortex atrophy. Neuropathology of the cerebral cortex showed increased cholesterol synthesis proteins in oligodendrocytes, more newly formed remyelinating oligodendrocytes, and increased myelin in estriol-treated EAE mice. Estriol treatment also decreased the loss of cortical layer V pyramidal neurons and their apical dendrites and preserved synapses. Together, estriol treatment after EAE onset reduced atrophy and was neuroprotective in the cerebral cortex.
Collapse
Affiliation(s)
- Cassandra E Meyer
- Department of Neurology, Ahmanson-Lovelace Brain Mapping Center, David Geffen School of Medicine at the University of California, Los Angeles, California; UCLA Multiple Sclerosis Program, Department of Neurology, David Geffen School of Medicine at the University of California, Los Angeles, California
| | - Andrew W Smith
- Department of Neurology, Ahmanson-Lovelace Brain Mapping Center, David Geffen School of Medicine at the University of California, Los Angeles, California; UCLA Multiple Sclerosis Program, Department of Neurology, David Geffen School of Medicine at the University of California, Los Angeles, California
| | - Aitana A Padilla-Requerey
- Department of Neurology, Ahmanson-Lovelace Brain Mapping Center, David Geffen School of Medicine at the University of California, Los Angeles, California; UCLA Multiple Sclerosis Program, Department of Neurology, David Geffen School of Medicine at the University of California, Los Angeles, California
| | - Vista Farkhondeh
- UCLA Multiple Sclerosis Program, Department of Neurology, David Geffen School of Medicine at the University of California, Los Angeles, California
| | - Noriko Itoh
- UCLA Multiple Sclerosis Program, Department of Neurology, David Geffen School of Medicine at the University of California, Los Angeles, California
| | - Yuichiro Itoh
- UCLA Multiple Sclerosis Program, Department of Neurology, David Geffen School of Medicine at the University of California, Los Angeles, California
| | - Josephine L Gao
- Department of Neurology, Ahmanson-Lovelace Brain Mapping Center, David Geffen School of Medicine at the University of California, Los Angeles, California; UCLA Multiple Sclerosis Program, Department of Neurology, David Geffen School of Medicine at the University of California, Los Angeles, California
| | - Patrick D Herbig
- Department of Neurology, Ahmanson-Lovelace Brain Mapping Center, David Geffen School of Medicine at the University of California, Los Angeles, California; UCLA Multiple Sclerosis Program, Department of Neurology, David Geffen School of Medicine at the University of California, Los Angeles, California
| | - Quynhanh Nguyen
- Department of Neurology, Ahmanson-Lovelace Brain Mapping Center, David Geffen School of Medicine at the University of California, Los Angeles, California; UCLA Multiple Sclerosis Program, Department of Neurology, David Geffen School of Medicine at the University of California, Los Angeles, California
| | - Katelyn H Ngo
- Department of Neurology, Ahmanson-Lovelace Brain Mapping Center, David Geffen School of Medicine at the University of California, Los Angeles, California; UCLA Multiple Sclerosis Program, Department of Neurology, David Geffen School of Medicine at the University of California, Los Angeles, California
| | - Mandavi R Oberoi
- Department of Neurology, Ahmanson-Lovelace Brain Mapping Center, David Geffen School of Medicine at the University of California, Los Angeles, California; UCLA Multiple Sclerosis Program, Department of Neurology, David Geffen School of Medicine at the University of California, Los Angeles, California
| | - Prabha Siddarth
- Jane and Terry Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, California
| | - Rhonda R Voskuhl
- UCLA Multiple Sclerosis Program, Department of Neurology, David Geffen School of Medicine at the University of California, Los Angeles, California
| | - Allan MacKenzie-Graham
- Department of Neurology, Ahmanson-Lovelace Brain Mapping Center, David Geffen School of Medicine at the University of California, Los Angeles, California; UCLA Multiple Sclerosis Program, Department of Neurology, David Geffen School of Medicine at the University of California, Los Angeles, California.
| |
Collapse
|
15
|
Bridge F, Butzkueven H, Van der Walt A, Jokubaitis VG. The impact of menopause on multiple sclerosis. Autoimmun Rev 2023; 22:103363. [PMID: 37230311 DOI: 10.1016/j.autrev.2023.103363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 05/21/2023] [Indexed: 05/27/2023]
Abstract
Menopause, defined as the permanent cessation of ovarian function, represents a period of significant fluctuation in sex hormone concentrations. Sex hormones including oestrogen, progesterone, testosterone and anti-Mullerian hormone are thought have neuroinflammatory effects and are implicated in both neuroprotection and neurodegeneration. Sex hormones are thought to have a role in modifying clinical trajectory in multiple sclerosis (MS) throughout the lifespan. Multiple sclerosis predominantly effects women and is typically diagnosed early in a woman's reproductive life. Most women with MS will undergo menopause. Despite this, the effect of menopause on MS disease course remains unclear. This review examines the relationship between sex hormones and MS disease activity and clinical course, particularly around the time of menopause. It will consider the role of interventions such as exogenous hormone replacement therapy in modulating clinical outcomes in this period. Understanding the impact of menopause on multiple sclerosis is fundamental for delivering optimal care to women with MS as they age and will inform treatment decisions with the aim of minimising relapses, disease accrual and improving quality of life.
Collapse
Affiliation(s)
- Francesca Bridge
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia; Department of Neurology, Alfred Health, Melbourne, Victoria, Australia.
| | - Helmut Butzkueven
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia; Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| | - Anneke Van der Walt
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia; Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| | - Vilija G Jokubaitis
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia; Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| |
Collapse
|
16
|
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
|
17
|
Murgia F, Giagnoni F, Lorefice L, Caria P, Dettori T, D’Alterio MN, Angioni S, Hendren AJ, Caboni P, Pibiri M, Monni G, Cocco E, Atzori L. Sex Hormones as Key Modulators of the Immune Response in Multiple Sclerosis: A Review. Biomedicines 2022; 10:biomedicines10123107. [PMID: 36551863 PMCID: PMC9775368 DOI: 10.3390/biomedicines10123107] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 11/25/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND A variety of autoimmune diseases, including MS, amplify sex-based physiological differences in immunological responsiveness. Female MS patients experience pathophysiological changes during reproductive phases (pregnancy and menopause). Sex hormones can act on immune cells, potentially enabling them to modify MS risk, activity, and progression, and to play a role in treatment. METHODS Scientific papers (published between 1998 and 2021) were selected through PubMed, Google Scholar, and Web of Science literature repositories. The search was limited to publications analyzing the hormonal profile of male and female MS patients during different life phases, in particular focusing on sex hormone treatment. RESULTS Both men and women with MS have lower testosterone levels compared to healthy controls. The levels of estrogens and progesterone increase during pregnancy and then rapidly decrease after delivery, possibly mediating an immune-stabilizing process. The literature examined herein evidences the neuroprotective effect of testosterone and estrogens in MS, supporting further examinations of their potential therapeutic uses. CONCLUSIONS A correlation has been identified between sex hormones and MS clinical activity. The combination of disease-modifying therapies with estrogen or estrogen plus a progestin receptor modulator promoting myelin repair might represent an important strategy for MS treatment in the future.
Collapse
Affiliation(s)
- Federica Murgia
- Clinical Metabolomics Unit, Department of Biomedical Sciences, University of Cagliari, 09124 Cagliari, Italy
- Correspondence:
| | - Florianna Giagnoni
- Clinical Metabolomics Unit, Department of Biomedical Sciences, University of Cagliari, 09124 Cagliari, Italy
| | - Lorena Lorefice
- Multiple Sclerosis Regional Center, ASSL Cagliari, ATS Sardinia, 09126 Cagliari, Italy
| | - Paola Caria
- Department of Biomedical Sciences, Section of Biochemistry, Biology, and Genetics, University of Cagliari, Cittadella Universitaria, 09124 Cagliari, Italy
| | - Tinuccia Dettori
- Department of Biomedical Sciences, Section of Biochemistry, Biology, and Genetics, University of Cagliari, Cittadella Universitaria, 09124 Cagliari, Italy
| | - Maurizio N. D’Alterio
- Division of Gynecology and Obstetrics, Department of Surgical Sciences, University of Cagliari, 09124 Cagliari, Italy
| | - Stefano Angioni
- Division of Gynecology and Obstetrics, Department of Surgical Sciences, University of Cagliari, 09124 Cagliari, Italy
| | - Aran J. Hendren
- Sussex Neuroscience, University of Sussex, Brighton BN1 9QG, UK
| | - Pierluigi Caboni
- Department of Life and Environmental Sciences, University of Cagliari, 09124 Cagliari, Italy
| | - Monica Pibiri
- Department of Biomedical Sciences, University of Cagliari, 09124 Cagliari, Italy
| | - Giovanni Monni
- Department of Obstetrics and Gynecology, Prenatal and Preimplantation Genetic Diagnosis, Fetal Therapy, Microcitemico Pediatric Hospital “A. Cao”, 09121 Cagliari, Italy
| | - Eleonora Cocco
- Multiple Sclerosis Regional Center, ASSL Cagliari, ATS Sardinia, Department of Medical Sciences and Public Health, University of Cagliari, 09126 Cagliari, Italy
| | - Luigi Atzori
- Clinical Metabolomics Unit, Department of Biomedical Sciences, University of Cagliari, 09124 Cagliari, Italy
| |
Collapse
|
18
|
Saleki K, Banazadeh M, Saghazadeh A, Rezaei N. Aging, testosterone, and neuroplasticity: friend or foe? Rev Neurosci 2022; 34:247-273. [PMID: 36017670 DOI: 10.1515/revneuro-2022-0033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 07/03/2022] [Indexed: 11/15/2022]
Abstract
Neuroplasticity or neural plasticity implicates the adaptive potential of the brain in response to extrinsic and intrinsic stimuli. The concept has been utilized in different contexts such as injury and neurological disease. Neuroplasticity mechanisms have been classified into neuroregenerative and function-restoring processes. In the context of injury, neuroplasticity has been defined in three post-injury epochs. Testosterone plays a key yet double-edged role in the regulation of several neuroplasticity alterations. Research has shown that testosterone levels are affected by numerous factors such as age, stress, surgical procedures on gonads, and pharmacological treatments. There is an ongoing debate for testosterone replacement therapy (TRT) in aging men; however, TRT is more useful in young individuals with testosterone deficit and more specific subgroups with cognitive dysfunction. Therefore, it is important to pay early attention to testosterone profile and precisely uncover its harms and benefits. In the present review, we discuss the influence of environmental factors, aging, and gender on testosterone-associated alterations in neuroplasticity, as well as the two-sided actions of testosterone in the nervous system. Finally, we provide practical insights for further study of pharmacological treatments for hormonal disorders focusing on restoring neuroplasticity.
Collapse
Affiliation(s)
- Kiarash Saleki
- Student Research Committee, Babol University of Medical Sciences, 47176 47745 Babol, Iran.,USERN Office, Babol University of Medical Sciences, 47176 47745 Babol, Iran.,Systematic Review and Meta-analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), 14197 33151 Tehran, Iran
| | - Mohammad Banazadeh
- Systematic Review and Meta-analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), 14197 33151 Tehran, Iran.,Pharmaceutical Sciences and Cosmetic Products Research Center, Kerman University of Medical Sciences, 76169 13555 Kerman, Iran
| | - Amene Saghazadeh
- Systematic Review and Meta-analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), 14197 33151 Tehran, Iran.,Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, 14197 33151 Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, 14197 33151 Tehran, Iran.,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, 14176 13151 Tehran, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), 14197 33151 Tehran, Iran
| |
Collapse
|
19
|
Umair M, Fazazi MR, Rangachari M. Biological Sex As a Critical Variable in CD4 + Effector T Cell Function in Preclinical Models of Multiple Sclerosis. Antioxid Redox Signal 2022; 37:135-149. [PMID: 34538129 PMCID: PMC9293683 DOI: 10.1089/ars.2021.0202] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Significance: T cells play a pivotal role in maintaining adaptive immune responses against pathogens. However, misdirected T cell responses against self-tissues may lead to autoimmune disease. Biological sex has profound effects on T cell function and is an important determinant of disease incidence and severity in autoimmune diseases such as multiple sclerosis (MS). Recent Advances: Many autoimmune diseases skew toward higher female incidence, including MS; however, it is has become increasingly more accepted that men living with MS are more prone to developing a progressive disease course and to having worsened disease outcomes. Critical Issues: In this review, we discuss what is known about the role of biological sex on T cell development and differentiation, examining evidence that male sex can augment T helper 17 (Th17) responses. Next, we outline what is known about sex differences in animal models of MS, and about the distinct roles played by sex hormones versus sex chromosomes in pathogenesis in these models. Finally, we discuss recent advances that examine the molecular basis for worsened disease outcomes in males, with a particular focus on the role played by Th17 cells in these models. Future Directions: Better understanding the role of biological sex in T cell function may pave the way to effective personalized treatment strategies in MS and other autoimmune diseases. Antioxid. Redox Signal. 37, 135-149.
Collapse
Affiliation(s)
- Muhammad Umair
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Pavillon CHUL, Quebec, Canada
| | - Mohamed Reda Fazazi
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Pavillon CHUL, Quebec, Canada
| | - Manu Rangachari
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Pavillon CHUL, Quebec, Canada.,Faculty of Medicine, Université Laval, Quebec, Canada
| |
Collapse
|
20
|
Zorrilla Veloz RI, McKenzie T, Palacios BE, Hu J. Nuclear hormone receptors in demyelinating diseases. J Neuroendocrinol 2022; 34:e13171. [PMID: 35734821 PMCID: PMC9339486 DOI: 10.1111/jne.13171] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 04/20/2022] [Accepted: 05/27/2022] [Indexed: 11/28/2022]
Abstract
Demyelination results from the pathological loss of myelin and is a hallmark of many neurodegenerative diseases. Despite the prevalence of demyelinating diseases, there are no disease modifying therapies that prevent the loss of myelin or promote remyelination. This review aims to summarize studies in the field that highlight the importance of nuclear hormone receptors in the promotion and maintenance of myelination and the relevance of nuclear hormone receptors as potential therapeutic targets for demyelinating diseases. These nuclear hormone receptors include the estrogen receptor, progesterone receptor, androgen receptor, vitamin D receptor, thyroid hormone receptor, peroxisome proliferator-activated receptor, liver X receptor, and retinoid X receptor. Pre-clinical studies in well-established animal models of demyelination have shown a prominent role of these nuclear hormone receptors in myelination through their promotion of oligodendrocyte maturation and development. The activation of the nuclear hormone receptors by their ligands also promotes the synthesis of myelin proteins and lipids in mouse models of demyelination. There are limited clinical studies that focus on how the activation of these nuclear hormone receptors could alleviate demyelination in patients with diseases such as multiple sclerosis (MS). However, the completed clinical trials have reported improved clinical outcome in MS patients treated with the ligands of some of these nuclear hormone receptors. Together, the positive results from both clinical and pre-clinical studies point to nuclear hormone receptors as promising therapeutic targets to counter demyelination.
Collapse
Affiliation(s)
- Rocío I Zorrilla Veloz
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Cancer Biology Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Takese McKenzie
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Neuroscience Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Bridgitte E Palacios
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Cancer Biology Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
- Neuroscience Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Jian Hu
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Cancer Biology Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
- Neuroscience Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| |
Collapse
|
21
|
Collongues N, Becker G, Jolivel V, Ayme-Dietrich E, de Seze J, Binamé F, Patte-Mensah C, Monassier L, Mensah-Nyagan AG. A Narrative Review on Axonal Neuroprotection in Multiple Sclerosis. Neurol Ther 2022; 11:981-1042. [PMID: 35610531 PMCID: PMC9338208 DOI: 10.1007/s40120-022-00363-7] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 05/03/2022] [Indexed: 01/08/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) resulting in demyelination and neurodegeneration. The therapeutic strategy is now largely based on reducing inflammation with immunosuppressive drugs. Unfortunately, when disease progression is observed, no drug offers neuroprotection apart from its anti-inflammatory effect. In this review, we explore current knowledge on the assessment of neurodegeneration in MS and look at putative targets that might prove useful in protecting the axon from degeneration. Among them, Bruton's tyrosine kinase inhibitors, anti-apoptotic and antioxidant agents, sex hormones, statins, channel blockers, growth factors, and molecules preventing glutamate excitotoxicity have already been studied. Some of them have reached phase III clinical trials and carry a great message of hope for our patients with MS.
Collapse
Affiliation(s)
- Nicolas Collongues
- Department of Neurology, University Hospital of Strasbourg, Strasbourg, France. .,Center for Clinical Investigation, INSERM U1434, Strasbourg, France. .,Biopathology of Myelin, Neuroprotection and Therapeutic Strategy, INSERM U1119, Strasbourg, France. .,University Department of Pharmacology, Addictology, Toxicology and Therapeutic, Strasbourg University, Strasbourg, France.
| | - Guillaume Becker
- University Department of Pharmacology, Addictology, Toxicology and Therapeutic, Strasbourg University, Strasbourg, France.,NeuroCardiovascular Pharmacology and Toxicology Laboratory, UR7296, University Hospital of Strasbourg, Strasbourg, France
| | - Valérie Jolivel
- Biopathology of Myelin, Neuroprotection and Therapeutic Strategy, INSERM U1119, Strasbourg, France
| | - Estelle Ayme-Dietrich
- University Department of Pharmacology, Addictology, Toxicology and Therapeutic, Strasbourg University, Strasbourg, France.,NeuroCardiovascular Pharmacology and Toxicology Laboratory, UR7296, University Hospital of Strasbourg, Strasbourg, France
| | - Jérôme de Seze
- Department of Neurology, University Hospital of Strasbourg, Strasbourg, France.,Center for Clinical Investigation, INSERM U1434, Strasbourg, France.,Biopathology of Myelin, Neuroprotection and Therapeutic Strategy, INSERM U1119, Strasbourg, France
| | - Fabien Binamé
- Biopathology of Myelin, Neuroprotection and Therapeutic Strategy, INSERM U1119, Strasbourg, France
| | - Christine Patte-Mensah
- Biopathology of Myelin, Neuroprotection and Therapeutic Strategy, INSERM U1119, Strasbourg, France
| | - Laurent Monassier
- University Department of Pharmacology, Addictology, Toxicology and Therapeutic, Strasbourg University, Strasbourg, France.,NeuroCardiovascular Pharmacology and Toxicology Laboratory, UR7296, University Hospital of Strasbourg, Strasbourg, France
| | - Ayikoé Guy Mensah-Nyagan
- Biopathology of Myelin, Neuroprotection and Therapeutic Strategy, INSERM U1119, Strasbourg, France
| |
Collapse
|
22
|
McCombe PA. The role of sex and pregnancy in multiple sclerosis: what do we know and what should we do? Expert Rev Neurother 2022; 22:377-392. [PMID: 35354378 DOI: 10.1080/14737175.2022.2060079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Multiple sclerosis (MS) is more prevalent in women than in men. The sex of the patient, and pregnancy, are reported to be associated with the clinical features of MS. The mechanism of this is unclear. AREAS COVERED This review summarizes data about sex differences in MS and the role of pregnancy. Possible mechanisms for the effects of sex and pregnancy are summarized, and practical suggestions for addressing these issues are provided. EXPERT OPINION There is considerable interdependence of the variables that are associated with MS. Men have a worse outcome of MS, and this could be due to the same factors that lead to greater incidence of neurodegenerative disease in men. The possible role of parity on the long-term outcome of MS is of interest. Future studies that look at the mechanisms of the effects of the sex of the patient on the outcome of MS are required. However, there are some actions that can be taken without further research. We can concentrate on public health measures that address the modifiable risk factors for MS and ensure that disease is controlled in women who intend to become pregnant and use appropriate disease modifying agents during pregnancy.
Collapse
Affiliation(s)
- Pamela A McCombe
- The University of Queensland, Centre for Clinical Research, Royal Brisbane and Women's Hospital, Herston, Australia
| |
Collapse
|
23
|
Evans SF, Hull ML, Hutchinson MR, Rolan PE. Androgens, Endometriosis and Pain. FRONTIERS IN REPRODUCTIVE HEALTH 2021; 3:792920. [PMID: 36303965 PMCID: PMC9580713 DOI: 10.3389/frph.2021.792920] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 11/11/2021] [Indexed: 01/04/2023] Open
Abstract
The intriguing relationship between androgens, endometriosis and chronic pain continues to unfold. Determining this relationship is of crucial importance to gynecologists managing people with these conditions, as common treatments dramatically alter her hormonal profiles, with both intended and unintended consequences. Although they may be present in the same individual, there is a recognized disconnect between pain or pain-related symptoms, and the presence or extent of endometriosis lesions. Reduced androgen levels provide a potential mechanism to link the development of endometriosis lesions and the presence of chronic pain. This research paper expands the presentation of our research at the World Endometriosis Congress in 2021, subsequently published in the Journal of Pain Research which demonstrated a strong inverse relationship between androgen levels and days per month of pelvic and period pain. Here we extend and further explore the evidence for a role for androgens in the etiology and management of dysmenorrhea and pelvic pain in women, both with and without endometriosis. We explore the potential for inflammation to induce low androgen levels and consider ways in which clinicians can optimize levels of androgens when treating women with these conditions. This article prompts the question: Is it estrogens that predispose people to a life of pain, or androgens that are protective?
Collapse
Affiliation(s)
- Susan F. Evans
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
- *Correspondence: Susan F. Evans
| | - M. Louise Hull
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
- Robinson Research Institute, School of Pediatrics and Reproductive Health, University of Adelaide, Adelaide, SA, Australia
| | - Mark R. Hutchinson
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
- ARC Centre of Excellence for Nanoscale Biophotonics, University of Adelaide, Adelaide, SA, Australia
| | - Paul E. Rolan
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
24
|
Plata-Bello J, Plata-Bello A, Pérez-Martín Y, López-Curtis D, Acosta-López S, Modroño C, Concepción-Massip T. Changes in resting-state measures of prostate cancer patients exposed to androgen deprivation therapy. Sci Rep 2021; 11:23350. [PMID: 34857811 PMCID: PMC8639725 DOI: 10.1038/s41598-021-02611-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 11/10/2021] [Indexed: 11/09/2022] Open
Abstract
The aim of the present work is to describe the differences in rs-fMRI measures (Amplitude of low frequency fluctuations [ALFF], Regional Homogeneity [ReHo] and Functional Connectivity [FC]) between patients exposed to Androgen deprivation therapy (ADT) and a control group. Forty-nine ADT patients and fifteen PC-non-ADT patients (Controls) were included in the study. A neuropsychological evaluation and a resting-state fMRI was performed to evaluate differences in ALFF and ReHo. Region of interest (ROI) analysis was also performed. ROIs were selected among those whose androgen receptor expression (at RNA-level) was the highest. FC analysis was performed using the same ROIs. Higher ALFF in frontal regions and temporal regions was identified in Controls than in ADT patients. In the ROI analysis, higher activity for Controls than ADT patients was shown in the left inferior frontal gyrus and in the left precentral gyrus. Lower ALFF in the right hippocampus and the lateral geniculate nucleus of the right thalamus was identified for Controls than ADT patients. Higher ReHo was observed in Controls in the left parietal-occipital area. Finally, ADT patients presented an increase of FC in more regions than Controls. These differences may reflect an impairment in brain functioning in ADT users.
Collapse
Affiliation(s)
- Julio Plata-Bello
- Department of Neurosurgery, Hospital Universitario de Canarias, CP 38320, S/C de Tenerife, Spain.
- Cognitive Neuroscience Research Group, University of La Laguna, S/C de Tenerife, Spain.
- Neuroscience Department, Hospital Universitario de Canarias, Calle Ofra s/n La Cuesta, La Laguna, CP 38320, S/C de Tenerife, Spain.
| | - Ana Plata-Bello
- Department of Urology, Hospital Universitario de Canarias, CP 38320, S/C de Tenerife, Spain
- Cognitive Neuroscience Research Group, University of La Laguna, S/C de Tenerife, Spain
| | - Yaiza Pérez-Martín
- Department of Neurology, Hospital Universitario de Canarias, CP 38320, S/C de Tenerife, Spain
| | - David López-Curtis
- Cognitive Neuroscience Research Group, University of La Laguna, S/C de Tenerife, Spain
| | - Silvia Acosta-López
- Cognitive Neuroscience Research Group, University of La Laguna, S/C de Tenerife, Spain
| | - Cristián Modroño
- Department of Physiology, Faculty of Medicine, University of La Laguna, CP 38320, S/C de Tenerife, Spain
| | | |
Collapse
|
25
|
Ysrraelit MC, Correale J. Impact of Andropause on Multiple Sclerosis. Front Neurol 2021; 12:766308. [PMID: 34803897 PMCID: PMC8602357 DOI: 10.3389/fneur.2021.766308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 10/18/2021] [Indexed: 11/18/2022] Open
Abstract
Andropause results from the natural decrease in testosterone levels that occurs with age. In contrast to menopause, which is a universal, well-characterized process associated with absolute gonadal failure, andropause ensues after gradual decline of both hypothalamic-pituitary-gonadal axis activity, as well as of testicular function, a process which usually develops over a period of many years. Increasing evidence on greater risk of Multiple sclerosis (MS) associated with lower testosterone levels is being reported. Likewise, epidemiological studies have shown a later age of onset of MS in men, relative to women, which could perhaps respond to the decline in protective testosterone levels. In this review, we will discuss the role of androgens in the development and function of the innate and adaptive immune response, as well as in neuroprotective mechanisms relevant to MS. Testosterone effects observed in different animal models and in epidemiological studies in humans will be discussed, as well as their correlation with physical disability and cognitive function levels. Finally, published and ongoing clinical trials exploring the role of androgens, particularly at key stages of sexual maturation, will be reviewed.
Collapse
Affiliation(s)
- Maria C Ysrraelit
- Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Buenos Aires, Argentina
| | - Jorge Correale
- Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Buenos Aires, Argentina
| |
Collapse
|
26
|
Almaguer-Mederos LE, Aguilera-Rodríguez R, Almaguer-Gotay D, Hechavarría-Barzaga K, Álvarez-Sosa A, Chapman-Rodríguez Y, Silva-Ricardo Y, González-Zaldivar Y, Vázquez-Mojena Y, Cuello-Almarales D, Rodríguez-Estupiñán A. Testosterone Levels Are Decreased and Associated with Disease Duration in Male Spinocerebellar Ataxia Type 2 Patients. THE CEREBELLUM 2021; 19:597-604. [PMID: 32440846 DOI: 10.1007/s12311-020-01134-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Spinocerebellar ataxia type 2 (SCA2) is a progressive neurodegenerative disorder due to an unstable expansion of a CAG repeat in the ATXN2 gene. Despite clinical and experimental evidence indicating the relevance of the gonadotropic axis to the prognosis and therapeutics for several late-onset neurodegenerative disorders, its functioning and association with disease severity have not been previously explored in SCA2. To assess serum levels of testosterone, luteinizing hormone (LH), and follicle-stimulating hormone (FSH), and their clinical relevance in SCA2 patients. A case-control study involving 94 Cuban SCA2 patients and 101 gender- and age-matched healthy controls was conducted. Testosterone, LH, and FSH serum levels were determined by radioimmunoassay or immunoradiometric assay systems. Clinical outcomes included age at onset, disease duration, Scale for the Assessment and Rating of Ataxia (SARA) score, and progression rate. Univariate general linear models were generated. Testosterone, LH, and FSH serum levels were significantly reduced in male SCA2 patients relative to control individuals. On average, there was a 35% reduction in testosterone levels in male patients versus male control individuals. Testosterone levels were associated with disease duration (r = 0.383; p = 0.025) and age at onset (r = 0.414; p = 0.011) in male SCA2 patients, but no association was observed between testosterone and CAG expansion size, SARA score, or progression rate. Testosterone levels might be a biomarker of disease progression in male SCA2 patients. Further studies are needed to explore the effects of low testosterone levels on non-motor symptoms, and to assess the potential of testosterone replacement therapy in male SCA2 patients.
Collapse
Affiliation(s)
- Luis E Almaguer-Mederos
- Center for the Investigation and Rehabilitation of Hereditary Ataxias (CIRAH), Holguin, Cuba.
| | - Raúl Aguilera-Rodríguez
- Center for the Investigation and Rehabilitation of Hereditary Ataxias (CIRAH), Holguin, Cuba
| | - Dennis Almaguer-Gotay
- Center for the Investigation and Rehabilitation of Hereditary Ataxias (CIRAH), Holguin, Cuba
| | | | | | | | | | | | - Yaimé Vázquez-Mojena
- Center for the Investigation and Rehabilitation of Hereditary Ataxias (CIRAH), Holguin, Cuba
| | - Dany Cuello-Almarales
- Center for the Investigation and Rehabilitation of Hereditary Ataxias (CIRAH), Holguin, Cuba
| | | |
Collapse
|
27
|
Mansilla MJ, Presas-Rodríguez S, Teniente-Serra A, González-Larreategui I, Quirant-Sánchez B, Fondelli F, Djedovic N, Iwaszkiewicz-Grześ D, Chwojnicki K, Miljković Đ, Trzonkowski P, Ramo-Tello C, Martínez-Cáceres EM. Paving the way towards an effective treatment for multiple sclerosis: advances in cell therapy. Cell Mol Immunol 2021; 18:1353-1374. [PMID: 33958746 PMCID: PMC8167140 DOI: 10.1038/s41423-020-00618-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 12/03/2020] [Indexed: 12/13/2022] Open
Abstract
Multiple sclerosis (MS) is a leading cause of chronic neurological disability in young to middle-aged adults, affecting ~2.5 million people worldwide. Currently, most therapeutics for MS are systemic immunosuppressive or immunomodulatory drugs, but these drugs are unable to halt or reverse the disease and have the potential to cause serious adverse events. Hence, there is an urgent need for the development of next-generation treatments that, alone or in combination, stop the undesired autoimmune response and contribute to the restoration of homeostasis. This review analyzes current MS treatments as well as different cell-based therapies that have been proposed to restore homeostasis in MS patients (tolerogenic dendritic cells, regulatory T cells, mesenchymal stem cells, and vaccination with T cells). Data collected from preclinical studies performed in the experimental autoimmune encephalomyelitis (EAE) model of MS in animals, in vitro cultures of cells from MS patients and the initial results of phase I/II clinical trials are analyzed to better understand which parameters are relevant for obtaining an efficient cell-based therapy for MS.
Collapse
Affiliation(s)
- M J Mansilla
- Division of Immunology, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Barcelona, Spain. .,Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain.
| | - S Presas-Rodríguez
- Multiple Sclerosis Unit, Department of Neurosciences, Germans Trias i Pujol University Hospital, Barcelona, Spain.,Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - A Teniente-Serra
- Division of Immunology, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Barcelona, Spain.,Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - I González-Larreategui
- Division of Immunology, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Barcelona, Spain.,Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - B Quirant-Sánchez
- Division of Immunology, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Barcelona, Spain.,Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - F Fondelli
- Division of Immunology, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Barcelona, Spain.,Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - N Djedovic
- Department of Immunology, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - D Iwaszkiewicz-Grześ
- Department of Medical Immunology, Medical University of Gdańsk, Gdańsk, Poland.,Poltreg S.A., Gdańsk, Poland
| | - K Chwojnicki
- Department of Anaesthesiology & Intensive Care, Medical University of Gdańsk, Gdańsk, Poland
| | - Đ Miljković
- Department of Immunology, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - P Trzonkowski
- Department of Medical Immunology, Medical University of Gdańsk, Gdańsk, Poland.,Poltreg S.A., Gdańsk, Poland
| | - C Ramo-Tello
- Multiple Sclerosis Unit, Department of Neurosciences, Germans Trias i Pujol University Hospital, Barcelona, Spain.,Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - E M Martínez-Cáceres
- Division of Immunology, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Barcelona, Spain. .,Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain.
| |
Collapse
|
28
|
Duncan KA, Garijo-Garde S. Sex, Genes, and Traumatic Brain Injury (TBI): A Call for a Gender Inclusive Approach to the Study of TBI in the Lab. Front Neurosci 2021; 15:681599. [PMID: 34025346 PMCID: PMC8131651 DOI: 10.3389/fnins.2021.681599] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 04/12/2021] [Indexed: 11/13/2022] Open
Affiliation(s)
- Kelli A Duncan
- Department of Biology, Vassar College, Poughkeepsie, NY, United States.,Program in Neuroscience and Behavior, Vassar College, Poughkeepsie, NY, United States
| | - Sarah Garijo-Garde
- Program in Neuroscience and Behavior, Vassar College, Poughkeepsie, NY, United States
| |
Collapse
|
29
|
Milosevic A, Bjelobaba I, Bozic ID, Lavrnja I, Savic D, Tesovic K, Jakovljevic M, Stojilkovic SS, Janjic MM. Testicular steroidogenesis is suppressed during experimental autoimmune encephalomyelitis in rats. Sci Rep 2021; 11:8996. [PMID: 33903635 PMCID: PMC8076254 DOI: 10.1038/s41598-021-88305-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 04/09/2021] [Indexed: 11/09/2022] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disease that usually occurs during the reproductive years in both sexes. Many male patients with MS show lower blood testosterone levels, which was also observed in male rats during experimental autoimmune encephalomyelitis (EAE), an animal model of MS. To better understand the causes of decreased testosterone production during EAE, we investigated the expression status of genes and proteins associated with steroidogenesis in the testes. No changes in the number of interstitial cells were observed in EAE animals, but the expression of the insulin-like 3 gene was reduced at the peak of the disease, implying that the Leydig cell functional capacity was affected. Consistent with this finding, the expression of most steroidogenic enzyme genes and proteins was reduced during EAE, including StAR, CYP11A1, CYP17A1 and HSD3B. No signs of testicular inflammation were observed. Recovery of steroidogenesis was observed after injection of hCG, the placental gonadotropin, or buserelin acetate, a gonadotropin-releasing hormone analogue, at the peak of EAE. Together, our results are consistent with the hypothesis that impaired testicular steroidogenesis originates upstream of the testes and that low serum LH is the main cause of decreased testosterone levels during EAE.
Collapse
Affiliation(s)
- Ana Milosevic
- Department for Neurobiology, Institute for Biological Research "Siniša Stanković"-National Institute of Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11000, Belgrade, Serbia
| | - Ivana Bjelobaba
- Department for Neurobiology, Institute for Biological Research "Siniša Stanković"-National Institute of Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11000, Belgrade, Serbia
| | - Iva D Bozic
- Department for Neurobiology, Institute for Biological Research "Siniša Stanković"-National Institute of Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11000, Belgrade, Serbia
| | - Irena Lavrnja
- Department for Neurobiology, Institute for Biological Research "Siniša Stanković"-National Institute of Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11000, Belgrade, Serbia
| | - Danijela Savic
- Department for Neurobiology, Institute for Biological Research "Siniša Stanković"-National Institute of Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11000, Belgrade, Serbia
| | - Katarina Tesovic
- Department for Neurobiology, Institute for Biological Research "Siniša Stanković"-National Institute of Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11000, Belgrade, Serbia
| | - Marija Jakovljevic
- Department for Neurobiology, Institute for Biological Research "Siniša Stanković"-National Institute of Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11000, Belgrade, Serbia
| | - Stanko S Stojilkovic
- Section on Cellular Signaling, Eunice Kennedy Shriver National Institute for Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Marija M Janjic
- Department for Neurobiology, Institute for Biological Research "Siniša Stanković"-National Institute of Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11000, Belgrade, Serbia.
| |
Collapse
|
30
|
Lin CH, Lai JN, Lee IC, Chou IC, Lin WD, Lin MC, Hong SY. Association Between Kawasaki Disease and Childhood Epilepsy: A Nationwide Cohort Study in Taiwan. Front Neurol 2021; 12:627712. [PMID: 33889123 PMCID: PMC8055829 DOI: 10.3389/fneur.2021.627712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 03/10/2021] [Indexed: 11/23/2022] Open
Abstract
Background: Kawasaki disease is a common vasculitis of childhood in East Asia. The complications following Kawasaki disease mostly included cardiovascular sequelae; non-cardiac complications have been reported but less studied. This study investigated potential epilepsy following Kawasaki disease in Taiwanese children. Objectives: Through National Health Insurance Research Database, we retrospectively analyzed the data of children aged <18 years with clinically diagnosed Kawasaki disease from January 1, 2000 to December 31, 2012 in Taiwan. These patients were followed up to estimate the incidence of epilepsy in the Kawasaki cohort in comparison with that in the non-Kawasaki cohort in Taiwan. Results: A total of 8,463 and 33,872 patients in the Kawasaki and non-Kawasaki cohorts were included in the study, respectively. Of the total eligible study subjects, 61.1% were boys and 38.9% were girls; most patients with newly diagnosed Kawasaki disease were aged <5 years [88.1%]. Patients with Kawasaki disease showed a higher incidence rate [47.98 vs. 27.45 every 100,000 person years] and significantly higher risk [adjusted hazard ratio = 1.66, 95% confidence interval = 1.13–2.44] of epilepsy than those without the disease. Additionally, female sex [adjusted hazard ratio = 2.30, 95% confidence interval = 1.31–4.04] and age <5 years [adjusted hazard ratio = 1.82, 95% confidence interval = 1.22–2.72] showed a significantly higher risk of epilepsy in the Kawasaki cohort. Conclusion: Results revealed a higher incidence rate and significant risk of epilepsy in Taiwanese children with Kawasaki disease than in those without the disease. Therefore, children diagnosed with Kawasaki disease are recommended follow-up as they have a high risk of epilepsy and seizure disorders.
Collapse
Affiliation(s)
- Chien-Heng Lin
- Division of Pediatrics Pulmonology, China Medical University, Children's Hospital, Taichung, Taiwan.,Department of Biomedical Imaging and Radiological Science, College of Medicine, China Medical University, Taichung, Taiwan
| | - Jung-Nien Lai
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan.,Department of Chinese Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Inn-Chi Lee
- Department of Pediatrics, School of Medicine, Chung Shan Medical University Hospital and Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - I-Ching Chou
- Division of Pediatrics Neurology, China Medical University, Children's Hospital, Taichung, Taiwan.,College of Chinese Medicine, Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan
| | - Wei-De Lin
- Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Mei-Chen Lin
- Management Office for Health Data, China Medical University Hospital, Taichung, Taiwan.,College of Medicine, China Medical University, Taichung, Taiwan
| | - Syuan-Yu Hong
- Division of Pediatrics Neurology, China Medical University, Children's Hospital, Taichung, Taiwan
| |
Collapse
|
31
|
Breton JM, Long KLP, Barraza MK, Perloff OS, Kaufer D. Hormonal Regulation of Oligodendrogenesis II: Implications for Myelin Repair. Biomolecules 2021; 11:290. [PMID: 33669242 PMCID: PMC7919830 DOI: 10.3390/biom11020290] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/10/2021] [Accepted: 02/13/2021] [Indexed: 02/07/2023] Open
Abstract
Alterations in myelin, the protective and insulating sheath surrounding axons, affect brain function, as is evident in demyelinating diseases where the loss of myelin leads to cognitive and motor dysfunction. Recent evidence suggests that changes in myelination, including both hyper- and hypo-myelination, may also play a role in numerous neurological and psychiatric diseases. Protecting myelin and promoting remyelination is thus crucial for a wide range of disorders. Oligodendrocytes (OLs) are the cells that generate myelin, and oligodendrogenesis, the creation of new OLs, continues throughout life and is necessary for myelin plasticity and remyelination. Understanding the regulation of oligodendrogenesis and myelin plasticity within disease contexts is, therefore, critical for the development of novel therapeutic targets. In our companion manuscript, we review literature demonstrating that multiple hormone classes are involved in the regulation of oligodendrogenesis under physiological conditions. The majority of hormones enhance oligodendrogenesis, increasing oligodendrocyte precursor cell differentiation and inducing maturation and myelin production in OLs. Thus, hormonal treatments present a promising route to promote remyelination. Here, we review the literature on hormonal regulation of oligodendrogenesis within the context of disorders. We focus on steroid hormones, including glucocorticoids and sex hormones, peptide hormones such as insulin-like growth factor 1, and thyroid hormones. For each hormone, we describe whether they aid in OL survival, differentiation, or remyelination, and we discuss their mechanisms of action, if known. Several of these hormones have yielded promising results in both animal models and in human conditions; however, a better understanding of hormonal effects, interactions, and their mechanisms will ultimately lead to more targeted therapeutics for myelin repair.
Collapse
Affiliation(s)
- Jocelyn M Breton
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA
| | - Kimberly L P Long
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA
| | - Matthew K Barraza
- Molecular and Cellular Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - Olga S Perloff
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Daniela Kaufer
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA
- Integrative Biology, University of California Berkeley, Berkeley, CA 94720, USA
- Canadian Institute for Advanced Research, Toronto, ON M5G1M1, Canada
| |
Collapse
|
32
|
Sparaco M, Bonavita S. The role of sex hormones in women with multiple sclerosis: From puberty to assisted reproductive techniques. Front Neuroendocrinol 2021; 60:100889. [PMID: 33189769 DOI: 10.1016/j.yfrne.2020.100889] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 11/02/2020] [Accepted: 11/08/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Multiple Sclerosis is a multifactorial chronic autoimmune disease, affecting predominantly females in the fertile age. Sex hormones changes during a woman's life, from puberty to menopause, including pregnancy and puerperium, may influence the onset and course of Multiple Sclerosis. The effect of estrogen levels on immune, clinical and radiological aspects of Multiple Sclerosis, also stimulated investigation on the effect of sexual hormones therapies, such as oral contraceptives and assisted reproductive technique, on the Multiple Sclerosis course. SEARCH STRATEGY AND SELECTION CRITERIA A literature search for original articles and reviews was conducted in the databases, including PubMed, Scopus, and ClinicalTrials.gov of the U.S. National Library of Medicine site from 1988 to 2020. RESULTS AND CONCLUSION This review reports the effects of the physiological and iatrogenic hormonal changes either on immune or clinical or paraclinical features in the different life stages of women affected by Multiple Sclerosis.
Collapse
Affiliation(s)
- Maddalena Sparaco
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Miraglia, 2, 80138 Naples, Italy
| | - Simona Bonavita
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Miraglia, 2, 80138 Naples, Italy.
| |
Collapse
|
33
|
Wang H, Zhou WX, Huang JF, Zheng XQ, Tian HJ, Wang B, Fu WL, Wu AM. Endocrine Therapy for the Functional Recovery of Spinal Cord Injury. Front Neurosci 2020; 14:590570. [PMID: 33390881 PMCID: PMC7773784 DOI: 10.3389/fnins.2020.590570] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 11/23/2020] [Indexed: 02/05/2023] Open
Abstract
Spinal cord injury (SCI) is a major cause of physical disability and leads to patient dissatisfaction with their quality of life. Patients with SCI usually exhibit severe clinical symptoms, including sensory and motor dysfunction below the injured levels, paraplegia, quadriplegia and urinary retention, which can exacerbate the substantial medical and social burdens. The major pathological change observed in SCI is inflammatory reaction, which induces demyelination, axonal degeneration, and the apoptosis and necrosis of neurons. Traditional medical treatments are mainly focused on the recovery of motor function and prevention of complications. To date, numerous studies have been conducted to explore the cellular and molecular mechanism of SCI and have proposed lots of effective treatments, but the clinical applications are still limited due to the complex pathogenesis and poor prognosis after SCI. Endocrine hormones are kinds of molecules that are synthesized by specialized endocrine organs and can participate in the regulation of multiple physiological activities, and their protective effects on several disorders have been widely discussed. In addition, many studies have identified that endocrine hormones can promote nerve regeneration and functional recovery in individuals with central nervous system diseases. Therefore, studies investigating the clinical applications of endocrine hormones as treatments for SCI are necessary. In this review, we described the neuroprotective roles of several endocrine hormones in SCI; endocrine hormone administration reduces cell death and promotes functional repair after SCI. We also proposed novel therapies for SCI.
Collapse
Affiliation(s)
- Hui Wang
- Zhejiang Provincial Key Laboratory of Orthopaedics, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Wen-Xian Zhou
- Zhejiang Provincial Key Laboratory of Orthopaedics, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Jin-Feng Huang
- Zhejiang Provincial Key Laboratory of Orthopaedics, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Xuan-Qi Zheng
- Zhejiang Provincial Key Laboratory of Orthopaedics, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Hai-Jun Tian
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bin Wang
- Department of Sports Medicine and Adult Reconstruction Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Wei-Li Fu
- Department of Orthopaedics, West China Hospital, Sichuan University, Chengdu, China
| | - Ai-Min Wu
- Zhejiang Provincial Key Laboratory of Orthopaedics, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
34
|
Lasrado N, Jia T, Massilamany C, Franco R, Illes Z, Reddy J. Mechanisms of sex hormones in autoimmunity: focus on EAE. Biol Sex Differ 2020; 11:50. [PMID: 32894183 PMCID: PMC7475723 DOI: 10.1186/s13293-020-00325-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 08/11/2020] [Indexed: 12/17/2022] Open
Abstract
Sex-related differences in the occurrence of autoimmune diseases is well documented, with females showing a greater propensity to develop these diseases than their male counterparts. Sex hormones, namely dihydrotestosterone and estrogens, have been shown to ameliorate the severity of inflammatory diseases. Immunologically, the beneficial effects of sex hormones have been ascribed to the suppression of effector lymphocyte responses accompanied by immune deviation from pro-inflammatory to anti-inflammatory cytokine production. In this review, we present our view of the mechanisms of sex hormones that contribute to their ability to suppress autoimmune responses with an emphasis on the pathogenesis of experimental autoimmune encephalomyelitis.
Collapse
Affiliation(s)
- Ninaad Lasrado
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE, 68583, USA
| | - Ting Jia
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE, 68583, USA
| | | | - Rodrigo Franco
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE, 68583, USA
| | - Zsolt Illes
- Department of Neurology, Odense University Hospital, University of Southern Denmark, Odense, Denmark
| | - Jay Reddy
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE, 68583, USA.
| |
Collapse
|
35
|
Bianchi VE, Rizzi L, Bresciani E, Omeljaniuk RJ, Torsello A. Androgen Therapy in Neurodegenerative Diseases. J Endocr Soc 2020; 4:bvaa120. [PMID: 33094209 PMCID: PMC7568521 DOI: 10.1210/jendso/bvaa120] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 08/18/2020] [Indexed: 12/14/2022] Open
Abstract
Neurodegenerative diseases, including Alzheimer disease (AD), Parkinson disease (PD), multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), and Huntington disease, are characterized by the loss of neurons as well as neuronal function in multiple regions of the central and peripheral nervous systems. Several studies in animal models have shown that androgens have neuroprotective effects in the brain and stimulate axonal regeneration. The presence of neuronal androgen receptors in the peripheral and central nervous system suggests that androgen therapy might be useful in the treatment of neurodegenerative diseases. To illustrate, androgen therapy reduced inflammation, amyloid-β deposition, and cognitive impairment in patients with AD. As well, improvements in remyelination in MS have been reported; by comparison, only variable results are observed in androgen treatment of PD. In ALS, androgen administration stimulated motoneuron recovery from progressive damage and regenerated both axons and dendrites. Only a few clinical studies are available in human individuals despite the safety and low cost of androgen therapy. Clinical evaluations of the effects of androgen therapy on these devastating diseases using large populations of patients are strongly needed.
Collapse
Affiliation(s)
- Vittorio Emanuele Bianchi
- Department of Endocrinology and Metabolism, Clinical Center Stella Maris, Strada Rovereta, Falciano, San Marino
| | - Laura Rizzi
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Elena Bresciani
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | | | - Antonio Torsello
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| |
Collapse
|
36
|
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
|
37
|
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
|
38
|
Gingele S, Stangel M. Emerging myelin repair agents in preclinical and early clinical development for the treatment of multiple sclerosis. Expert Opin Investig Drugs 2020; 29:583-594. [PMID: 32348161 DOI: 10.1080/13543784.2020.1762567] [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] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Remyelination is a highly effective regenerative process that can restore axon function, prevent axonal loss, and reverse clinical deficits after demyelination. Hence, the promotion of remyelination is a logical goal in patients with multiple sclerosis (MS) in which remyelination is often insufficient. However, despite great progress regarding the development of immunomodulatory therapies for MS and an abundance of promising evidence from preclinical experiments so far, no therapy has convincingly demonstrated clinically significant remyelination properties. Therefore, enhancing myelin repair is an urgent and unmet need in MS. AREAS COVERED We searched clinicaltrials.gov and pubmed.ncbi.nlm.nih.gov and focused on therapeutic agents in development from the preclinical stage to clinical phase II. We selected agents for which data are available from in vitro experiments and at least one toxic demyelination animal model that reached at least phase I in clinical development in MS patients. EXPERT OPINION The evidence to promote remyelination is very promising for several agents, some of which possess anti-muscarinergic properties. Since remyelination is a complex process that involves various coordinated steps, a combination of different therapeutic approaches addressing different aspects of this regenerative mechanism may be reasonable. Furthermore, suitable surrogate markers of remyelination are necessary for proof-of-concept clinical trials.
Collapse
Affiliation(s)
- Stefan Gingele
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School , Hannover, Germany
| | - Martin Stangel
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School , Hannover, Germany
| |
Collapse
|
39
|
Roles of Progesterone, Testosterone and Their Nuclear Receptors in Central Nervous System Myelination and Remyelination. Int J Mol Sci 2020; 21:ijms21093163. [PMID: 32365806 PMCID: PMC7246940 DOI: 10.3390/ijms21093163] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 04/27/2020] [Accepted: 04/28/2020] [Indexed: 12/14/2022] Open
Abstract
Progesterone and testosterone, beyond their roles as sex hormones, are neuroactive steroids, playing crucial regulatory functions within the nervous system. Among these, neuroprotection and myelin regeneration are important ones. The present review aims to discuss the stimulatory effects of progesterone and testosterone on the process of myelination and remyelination. These effects have been demonstrated in vitro (i.e., organotypic cultures) and in vivo (cuprizone- or lysolecithin-induced demyelination and experimental autoimmune encephalomyelitis (EAE)). Both steroids stimulate myelin formation and regeneration by acting through their respective intracellular receptors: progesterone receptors (PR) and androgen receptors (AR). Activation of these receptors results in multiple events involving direct transcription and translation, regulating general homeostasis, cell proliferation, differentiation, growth and myelination. It also ameliorates immune response as seen in the EAE model, resulting in a significant decrease in inflammation leading to a fast recovery. Although natural progesterone and testosterone have a therapeutic potential, their synthetic derivatives—the 19-norprogesterone (nestorone) and 7α-methyl-nortestosterone (MENT), already used as hormonal contraception or in postmenopausal hormone replacement therapies, may offer enhanced benefits for myelin repair. We summarize here a recent advancement in the field of myelin biology, to treat demyelinating disorders using the natural as well as synthetic analogs of progesterone and testosterone.
Collapse
|
40
|
From baby brain to mommy brain: Widespread gray matter gain after giving birth. Cortex 2020; 126:334-342. [PMID: 32105976 DOI: 10.1016/j.cortex.2019.12.029] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 08/26/2019] [Accepted: 12/16/2019] [Indexed: 01/26/2023]
Abstract
Pregnancy results in obvious physiological changes to the female body, but data as to what happens to the maternal brain after giving birth are sparse as well as inconsistent. The overall goal of this study is to determine the nature of cerebral change in the postpartum period. For this purpose, we analyzed T1-weighted brain images of 14 healthy women (age range: 25-38 years) at two time points, specifically within 1-2 days of childbirth (immediate postpartum) and at 4-6 weeks after childbirth (late postpartum). When comparing voxel-wise gray matter between these two time points, there was no evidence of any significant decrease. Instead, we detected a pronounced gray matter increase involving both cortical and subcortical regions, such as the pre- and postcentral gyrus, the frontal and central operculum, the inferior frontal gyrus, the precuneus, and the middle occipital gyrus, as well as the thalamus and caudate. These structural changes occurring within only 4-6 weeks after delivery are reflective of a high degree of neuroplasticity and massive adaptations in the maternal brain. They may suggest a restoration of brain tissue following pregnancy and/or a substantial brain reorganization, possibly to accommodate a multi-faceted repertoire of complex behaviors associated with being a mother.
Collapse
|
41
|
Abstract
Sex differences in the incidence or severity of disease characterize many autoimmune and neurodegenerative diseases. Multiple sclerosis is a complex disease with both autoimmune and neurodegenerative aspects and is characterized by sex differences in susceptibility and progression. Research in the study sex differences is a way to capitalize on a known clinical observation, mechanistically disentangle it at the laboratory bench, then translate basic research findings back to the clinic as a novel treatment trial tailored to optimally benefit each sex. This "Bedside to Bench to Bedside" approach based on sex differences in MS will be reviewed here, first for disease susceptibility then for disability progression.
Collapse
Affiliation(s)
- Rhonda R Voskuhl
- Department of Neurology, University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
42
|
Giatti S, Diviccaro S, Serafini MM, Caruso D, Garcia-Segura LM, Viviani B, Melcangi RC. Sex differences in steroid levels and steroidogenesis in the nervous system: Physiopathological role. Front Neuroendocrinol 2020; 56:100804. [PMID: 31689419 DOI: 10.1016/j.yfrne.2019.100804] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 10/10/2019] [Accepted: 10/30/2019] [Indexed: 12/13/2022]
Abstract
The nervous system, in addition to be a target for steroid hormones, is the source of a variety of neuroactive steroids, which are synthesized and metabolized by neurons and glial cells. Recent evidence indicates that the expression of neurosteroidogenic proteins and enzymes and the levels of neuroactive steroids are different in the nervous system of males and females. We here summarized the state of the art of neuroactive steroids, particularly taking in consideration sex differences occurring in the synthesis and levels of these molecules. In addition, we discuss the consequences of sex differences in neurosteroidogenesis for the function of the nervous system under healthy and pathological conditions and the implications of neuroactive steroids and neurosteroidogenesis for the development of sex-specific therapeutic interventions.
Collapse
Affiliation(s)
- Silvia Giatti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Silvia Diviccaro
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Melania Maria Serafini
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Donatella Caruso
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Luis Miguel Garcia-Segura
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Barbara Viviani
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Roberto C Melcangi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy.
| |
Collapse
|
43
|
Abstract
Neuroinflammation is implicated in contributing to a variety of neurologic and somatic illnesses including Alzheimer's disease (AD), Parkinson's disease (PD), and depression. In this chapter, we focus on the role of neuroinflammation in mediating these three illnesses and portray interactions between the immune response and the central nervous system in the context of sex differences in disease progression. The majority of this chapter is supported by clinical findings; however, we occasionally utilize preclinical models where human studies are currently lacking. We begin by detailing the pathology of neuroinflammation, distinguishing between acute and chronic inflammation, and examining contributions from the innate and adaptive immune systems. Next, we summarize potential mechanisms of immune cell mediators including interleukin-1 beta (IL-1β), tumor necrosis factor α, and IL-6 in AD, PD, and depression development. Given the strong sex bias seen in these illnesses, we additionally examine the role of sex hormones, e.g., estrogen and testosterone in mediating neuroinflammation at the cellular level. Systematically, we detail how sex hormones may contribute to distinct behavioral and clinical symptoms and prognosis between males and females with AD, PD, or depression. Finally, we highlight the possible role of exercise in alleviating neuroinflammation, as well as evidence that antiinflammatory drug therapies improve cognitive symptoms observed in brain-related diseases.
Collapse
Affiliation(s)
- Deepika Mukhara
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, United States
| | - Unsong Oh
- Department of Neurology, Virginia Commonwealth University, Richmond, VA, United States
| | - Gretchen N Neigh
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, United States.
| |
Collapse
|
44
|
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
|
45
|
Tobore TO. Towards a comprehensive etiopathogenetic and pathophysiological theory of multiple sclerosis. Int J Neurosci 2019; 130:279-300. [PMID: 31588832 DOI: 10.1080/00207454.2019.1677648] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Background: Multiple sclerosis (MS) is a neurodegenerative disease caused by dysfunction of the immune system that affects the central nervous system (CNS). It is characterized by demyelination, chronic inflammation, neuronal and oligodendrocyte loss and reactive astrogliosis. It can result in physical disability and acute neurological and cognitive problems. Despite the gains in knowledge of immunology, cell biology, and genetics in the last five decades, the ultimate etiology or specific elements that trigger MS remain unknown. The objective of this review is to propose a theoretical basis for MS etiopathogenesis.Methods: Search was done by accessing PubMed/Medline, EBSCO, and PsycINFO databases. The search string used was "(multiple sclerosis* OR EAE) AND (pathophysiology* OR etiopathogenesis)". The electronic databases were searched for titles or abstracts containing these terms in all published articles between January 1, 1960, and June 30, 2019. The search was filtered down to 362 articles which were included in this review.Results: A framework to better understand the etiopathogenesis and pathophysiology of MS can be derived from four essential factors; mitochondria dysfunction (MtD) & oxidative stress (OS), vitamin D (VD), sex hormones and thyroid hormones. These factors play a direct role in MS etiopathogenesis and have a modulatory effect on many other factors involved in the disease.Conclusions: For better MS prevention and treatment outcomes, efforts should be geared towards treating thyroid problems, sex hormone alterations, VD deficiency, sleep problems and melatonin alterations. MS patients should be encouraged to engage in activities that boost total antioxidant capacity (TAC) including diet and regular exercise and discouraged from activities that promote OS including smoking and alcohol consumption.
Collapse
|
46
|
Tobore TO. On elucidation of the role of mitochondria dysfunction and oxidative stress in multiple sclerosis. ACTA ACUST UNITED AC 2019. [DOI: 10.1111/ncn3.12335] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
47
|
Plata-Bello J, Plata-Bello A, Pérez-Martín Y, Fajardo V, Concepción-Massip T. Androgen deprivation therapy increases brain ageing. Aging (Albany NY) 2019; 11:5613-5627. [PMID: 31377745 PMCID: PMC6710035 DOI: 10.18632/aging.102142] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 07/30/2019] [Indexed: 11/25/2022]
Abstract
BACKGROUND Prostate cancer (PC) is the most frequent neoplasia in the male population and androgen deprivation therapy (ADT) is frequently used in the management of the disease. AIM To evaluate the effect of ADT exposure on cognitive status, grey matter volume (GMV) and white matter lesion (WML) load. METHODS Fifty ADT patients and fifteen PC-non-ADT (control) patients were included in the study. A neuropsychological evaluation was performed and a magnetic resonance imaging (MRI), with anatomical T1 and FLAIR sequences, was performed to evaluate the GMV and the WML burden. RESULTS Most of the patients included in the study presented a significant cognitive impairment (CI). No significant differences were identified in the cognitive assessment between the studied groups, but when considering the educational background intragroup differences were found.No significant difference of GMV and WML volume were identified between groups, but a negative relationship between the ADT period and the GMV was identified. Furthermore, a significant positive association between the age and the lesion volume was found in the ADT group (β=.406; p=.004). CONCLUSION PC patients exposed to ADT present an acceleration of age-related brain changes, such as WML development and GMV loss.
Collapse
Affiliation(s)
- Julio Plata-Bello
- Department of Neuroscience, Hospital Universitario de Canarias, S/C de Tenerife, CP 38320, Spain
| | - Ana Plata-Bello
- Department of Urology, Hospital Universitario de Canarias, S/C de Tenerife, CP 38320, Spain
| | - Yaiza Pérez-Martín
- Department of Neuroscience, Hospital Universitario de Canarias, S/C de Tenerife, CP 38320, Spain
| | - Victor Fajardo
- Department of Neuroscience, Hospital Universitario de Canarias, S/C de Tenerife, CP 38320, Spain
| | | |
Collapse
|
48
|
Mendell AL, MacLusky NJ. The testosterone metabolite 3α-androstanediol inhibits oxidative stress-induced ERK phosphorylation and neurotoxicity in SH-SY5Y cells through an MKP3/DUSP6-dependent mechanism. Neurosci Lett 2018; 696:60-66. [PMID: 30552945 DOI: 10.1016/j.neulet.2018.12.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 12/04/2018] [Accepted: 12/07/2018] [Indexed: 01/08/2023]
Abstract
Testosterone exerts neuroprotective effects on the brain, but the mechanisms by which these effects are exerted appear to be different in males and females. While in females they involve local conversion to estradiol, in males they may be androgen receptor-dependent, or mediated through metabolism to neurosteroids such as 5α-androstane-3α,17β-diol (3α-diol), which acts through different mechanisms than testosterone itself. Recently, we demonstrated that 3α-diol can protect neurons and neuronal-like cells against oxidative stress-induced neurotoxicity associated with prolonged phosphorylation of the extracellular signal-regulated kinase (ERK). The mechanism(s) responsible for these effects remain unknown. In the present study, we sought to determine whether the ERK-specific phosphatase, mitogen-activated protein kinase phosphatase 3/dual specificity phosphatase 6 (MKP3/DUSP6), is involved in the cytoprotective effects of 3α-diol in SH-SY5Y human female neuroblastoma cells. 3α-diol inhibited ERK phosphorylation and ameliorated cell death induced by the oxidative stressor hydrogen peroxide (H2O2). These protective effects were significantly reduced by pre-treatment with the MKP3/DUSP6 inhibitor BCI. In addition, H2O2 decreased expression of MKP3/DUSP6, and this was prevented by co-treatment with 3α-diol. These findings suggest that the protective effects of 3α-diol are mediated through regulation of ERK phosphorylation in neurotoxic conditions and indicate that these effects may be exerted through modulation of MKP3/DUSP6. Targeting the regulation of MKP3/DUSP6 may be beneficial in reducing toxicity under conditions of oxidative stress.
Collapse
Affiliation(s)
- Ari Loren Mendell
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, 50 Stone Road East, Guelph, ON, N1G 2W1, Canada.
| | - Neil James MacLusky
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, 50 Stone Road East, Guelph, ON, N1G 2W1, Canada.
| |
Collapse
|
49
|
Gold SM, Willing A, Leypoldt F, Paul F, Friese MA. Sex differences in autoimmune disorders of the central nervous system. Semin Immunopathol 2018; 41:177-188. [PMID: 30361800 DOI: 10.1007/s00281-018-0723-8] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 10/04/2018] [Indexed: 12/25/2022]
Abstract
Stronger adaptive immune responses in females can be observed in different mammals, resulting in better control of infections compared to males. However, this presumably evolutionary difference likely also drives higher incidence of autoimmune diseases observed in humans. Here, we summarize sex differences in the most common autoimmune diseases of the central nervous system (CNS) and discuss recent advances in the understanding of possible underlying immunological and CNS intrinsic mechanisms. In multiple sclerosis (MS), the most common inflammatory disease of the CNS, but also in rarer conditions, such as neuromyelitis optica spectrum disorders (NMOSD) or neuronal autoantibody-mediated autoimmune encephalitis (AE), sex is one of the top risk factors, with women being more often affected than men. Immunological mechanisms driving the sex bias in autoimmune CNS diseases are complex and include hormonal as well as genetic and epigenetic effects, which could also be exerted indirectly via modulation of the microbiome. Furthermore, CNS intrinsic differences could underlie the sex bias in autoimmunity by differential responses to injury. The strong effects of sex on incidence and possibly also activity and progression of autoimmune CNS disorders suggest that treatments need to be tailored to each sex to optimize efficacy. To date, however, due to a lack of systematic studies on treatment responses in males versus females, evidence in this area is still sparse. We argue that studies taking sex differences into account could pave the way for sex-specific and therefore personalized treatment.
Collapse
Affiliation(s)
- Stefan M Gold
- Institute of Neuroimmunology and Multiple Sclerosis (INIMS), Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg Eppendorf, Hamburg, Germany.,Department of Psychiatry, Charité - Universitätsmedizin Berlin, Cooperate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Anne Willing
- Institute of Neuroimmunology and Multiple Sclerosis (INIMS), Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Frank Leypoldt
- Department of Neurology, Christian-Albrechts-University Kiel, Kiel, Germany.,Neuroimmunology Section, Institute of Clinical Chemistry, University Hospital Schleswig-Holstein Kiel/Lübeck, Kiel/Lübeck, Germany
| | - Friedemann Paul
- NeuroCure Clinical Research Center, Charité - Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany.,Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Manuel A Friese
- Institute of Neuroimmunology and Multiple Sclerosis (INIMS), Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg Eppendorf, Hamburg, Germany.
| |
Collapse
|
50
|
Abstract
Low plasma testosterone (T) levels correlated with metabolic syndrome, cardiovascular diseases, and increased mortality risk. T exerts a significant effect on the regulation of adipose tissue accumulation, and in the glucose and lipids metabolism. Adipocytes are the primary source of the most important adipokines responsible for inflammation and chronic diseases. This review aims to analyze the possible effect of T on the regulation of the proinflammatory cytokines secretion. A systematic literature search on MEDLINE, Google Scholar, and Cochrane using the combination of the following keywords: “testosterone” with “inflammation,” “cytokines,” “adiponectin, CRP, IL-1B, IL-6, TNFα, leptin” was conducted. Sixteen articles related to the effect of low T level and 18 to the effect of T therapy on proinflammatory cytokine were found. T exerts a significant inhibitory effect on adipose tissue formation and the expression of various adipocytokines, such as leptin, TNF-α, IL-6, IL-1, and is positively correlated with adiponectin level, whereas a low T level is correlated with increased expression of markers of inflammation. Further studies are necessary to investigate the role of T, integrated with weight loss and physical activity, on its action on the mechanisms of production and regulation of proinflammatory cytokines.
Collapse
|