1
|
Perillan C, Coto A, Arguelles J, Nuñez P. Study of the impact of multiple sclerosis on the reproductive life of Spanish women: An online survey. Mult Scler Relat Disord 2024; 90:105789. [PMID: 39096667 DOI: 10.1016/j.msard.2024.105789] [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: 04/12/2024] [Revised: 06/29/2024] [Accepted: 07/25/2024] [Indexed: 08/05/2024]
Abstract
BACKGROUND Women have a higher risk of developing multiple sclerosis (MS) than men. The natural reproductive period from menarche to menopause corresponds to the period of active inflammatory disease in MS. Mothers and pregnant women with MS need information about how their disease may affect pregnancy and breastfeeding. AIM The aim was to explore the reproductive factors in an MS-diagnosed population and to identify ways to support patients and their decision-making process. METHODS We conducted a cross-sectional, Web-based survey of women living with MS in Asturias (Spain) using a community-based participatory approach. FINDINGS Early menarche may predict the onset of MS. Pregnancy improves the general health of patients and reduces the number of relapses. Breastfeeding is often not practised and may cause concern in women. MS does not affect the age of menopause, but it can worsen symptoms. However, menopause does not increase the number of MS relapses. CONCLUSIONS MS is increasingly diagnosed at an earlier age, which increases the number of women who become pregnant after being diagnosed with MS. The decrease in MS relapses during pregnancy and the increase during the postpartum period are consistent with previous reports. Women who choose to breastfeed are in the minority due to treatment incompatibility, although some currently used treatments are compatible with breastfeeding. However, there is a lack of information on this which should be investigated.
Collapse
Affiliation(s)
- Carmen Perillan
- Department of Functional Biology, Physiology, Faculty of Medicine and Health Sciences, University of Oviedo, Oviedo, Spain
| | - Aroa Coto
- Department of Functional Biology, Physiology, Faculty of Medicine and Health Sciences, University of Oviedo, Oviedo, Spain
| | - Juan Arguelles
- Department of Functional Biology, Physiology, Faculty of Medicine and Health Sciences, University of Oviedo, Oviedo, Spain
| | - Paula Nuñez
- Department of Functional Biology, Physiology, Faculty of Medicine and Health Sciences, University of Oviedo, Oviedo, Spain.
| |
Collapse
|
2
|
Loo RTJ, Soudy M, Nasta F, Macchi M, Glaab E. Bioinformatics approaches for studying molecular sex differences in complex diseases. Brief Bioinform 2024; 25:bbae499. [PMID: 39397573 PMCID: PMC11471957 DOI: 10.1093/bib/bbae499] [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: 04/22/2024] [Revised: 09/09/2024] [Accepted: 09/30/2024] [Indexed: 10/15/2024] Open
Abstract
Many complex diseases exhibit pronounced sex differences that can affect both the initial risk of developing the disease, as well as clinical disease symptoms, molecular manifestations, disease progression, and the risk of developing comorbidities. Despite this, computational studies of molecular data for complex diseases often treat sex as a confounding variable, aiming to filter out sex-specific effects rather than attempting to interpret them. A more systematic, in-depth exploration of sex-specific disease mechanisms could significantly improve our understanding of pathological and protective processes with sex-dependent profiles. This survey discusses dedicated bioinformatics approaches for the study of molecular sex differences in complex diseases. It highlights that, beyond classical statistical methods, approaches are needed that integrate prior knowledge of relevant hormone signaling interactions, gene regulatory networks, and sex linkage of genes to provide a mechanistic interpretation of sex-dependent alterations in disease. The review examines and compares the advantages, pitfalls and limitations of various conventional statistical and systems-level mechanistic analyses for this purpose, including tailored pathway and network analysis techniques. Overall, this survey highlights the potential of specialized bioinformatics techniques to systematically investigate molecular sex differences in complex diseases, to inform biomarker signature modeling, and to guide more personalized treatment approaches.
Collapse
Affiliation(s)
- Rebecca Ting Jiin Loo
- Biomedical Data Science Group, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 6 avenue du Swing, L-4367 Belvaux, Luxembourg
| | - Mohamed Soudy
- Biomedical Data Science Group, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 6 avenue du Swing, L-4367 Belvaux, Luxembourg
| | - Francesco Nasta
- Biomedical Data Science Group, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 6 avenue du Swing, L-4367 Belvaux, Luxembourg
| | - Mirco Macchi
- Biomedical Data Science Group, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 6 avenue du Swing, L-4367 Belvaux, Luxembourg
| | - Enrico Glaab
- Biomedical Data Science Group, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 6 avenue du Swing, L-4367 Belvaux, Luxembourg
| |
Collapse
|
3
|
Nova A, Di Caprio G, Bernardinelli L, Fazia T. Genetic and early life factors influence on time-to-multiple sclerosis diagnosis: A UK Biobank study. Mult Scler 2024; 30:994-1003. [PMID: 38847449 DOI: 10.1177/13524585241257205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/31/2024]
Abstract
BACKGROUND Previous investigations into multiple sclerosis (MS) risk factors predominantly relied on retrospective studies, which do not consider different follow-up times and assume a constant risk effect throughout lifetime. OBJECTIVE We aimed to evaluate the impact of genetic and early life factors on MS diagnosis by employing a time-to-event analysis in a prospective cohort. METHODS We used the UK Biobank data, considering the observation period from birth up to 31 December 2022. We considered genetic risk, using a multiple sclerosis polygenic risk score (MS-PRS), and various early life factors. Tobacco smoking and infectious mononucleosis diagnosis were also considered as time-varying variables along the follow-up. Using a Cox proportional hazards model, we examined the associations between these factors and MS diagnosis instantaneous risk. RESULTS We analyzed 345,027 participants, of which 1669 had an MS diagnosis. Our analysis revealed age-dependent effects for sex (females vs males) and higher MS-PRS, with greater hazard ratios observed in young adults. CONCLUSION The age-dependent effects suggest that retrospective studies could have underestimated sex and genetic variants' risk roles during younger ages. Therefore, we emphasize the importance of a time-to-event approach using longitudinal data to better characterize age-dependent risk effects.
Collapse
Affiliation(s)
- Andrea Nova
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Giovanni Di Caprio
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Luisa Bernardinelli
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Teresa Fazia
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| |
Collapse
|
4
|
Pereira QC, Fortunato IM, Oliveira FDS, Alvarez MC, dos Santos TW, Ribeiro ML. Polyphenolic Compounds: Orchestrating Intestinal Microbiota Harmony during Aging. Nutrients 2024; 16:1066. [PMID: 38613099 PMCID: PMC11013902 DOI: 10.3390/nu16071066] [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: 01/25/2024] [Revised: 03/20/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024] Open
Abstract
In the aging process, physiological decline occurs, posing a substantial threat to the physical and mental well-being of the elderly and contributing to the onset of age-related diseases. While traditional perspectives considered the maintenance of life as influenced by a myriad of factors, including environmental, genetic, epigenetic, and lifestyle elements such as exercise and diet, the pivotal role of symbiotic microorganisms had been understated. Presently, it is acknowledged that the intestinal microbiota plays a profound role in overall health by signaling to both the central and peripheral nervous systems, as well as other distant organs. Disruption in this bidirectional communication between bacteria and the host results in dysbiosis, fostering the development of various diseases, including neurological disorders, cardiovascular diseases, and cancer. This review aims to delve into the intricate biological mechanisms underpinning dysbiosis associated with aging and the clinical ramifications of such dysregulation. Furthermore, we aspire to explore bioactive compounds endowed with functional properties capable of modulating and restoring balance in this aging-related dysbiotic process through epigenetics alterations.
Collapse
Affiliation(s)
- Quélita Cristina Pereira
- Laboratory of Immunopharmacology and Molecular Biology, Sao Francisco University, Av. Sao Francisco de Assis, 218, Braganca Paulista 12916-900, SP, Brazil; (Q.C.P.); (I.M.F.); (F.d.S.O.); (M.C.A.); (T.W.d.S.)
| | - Isabela Monique Fortunato
- Laboratory of Immunopharmacology and Molecular Biology, Sao Francisco University, Av. Sao Francisco de Assis, 218, Braganca Paulista 12916-900, SP, Brazil; (Q.C.P.); (I.M.F.); (F.d.S.O.); (M.C.A.); (T.W.d.S.)
| | - Fabricio de Sousa Oliveira
- Laboratory of Immunopharmacology and Molecular Biology, Sao Francisco University, Av. Sao Francisco de Assis, 218, Braganca Paulista 12916-900, SP, Brazil; (Q.C.P.); (I.M.F.); (F.d.S.O.); (M.C.A.); (T.W.d.S.)
| | - Marisa Claudia Alvarez
- Laboratory of Immunopharmacology and Molecular Biology, Sao Francisco University, Av. Sao Francisco de Assis, 218, Braganca Paulista 12916-900, SP, Brazil; (Q.C.P.); (I.M.F.); (F.d.S.O.); (M.C.A.); (T.W.d.S.)
- Hematology and Transfusion Medicine Center, University of Campinas/Hemocentro, UNICAMP, Rua Carlos Chagas 480, Campinas 13083-878, SP, Brazil
| | - Tanila Wood dos Santos
- Laboratory of Immunopharmacology and Molecular Biology, Sao Francisco University, Av. Sao Francisco de Assis, 218, Braganca Paulista 12916-900, SP, Brazil; (Q.C.P.); (I.M.F.); (F.d.S.O.); (M.C.A.); (T.W.d.S.)
| | - Marcelo Lima Ribeiro
- Laboratory of Immunopharmacology and Molecular Biology, Sao Francisco University, Av. Sao Francisco de Assis, 218, Braganca Paulista 12916-900, SP, Brazil; (Q.C.P.); (I.M.F.); (F.d.S.O.); (M.C.A.); (T.W.d.S.)
| |
Collapse
|
5
|
Hosseinpour Z, Oladosu O, Liu WQ, Pike GB, Yong VW, Metz LM, Zhang Y. Distinct characteristics and severity of brain magnetic resonance imaging lesions in women and men with multiple sclerosis assessed using verified texture analysis measures. Front Neurol 2023; 14:1213377. [PMID: 37638198 PMCID: PMC10449451 DOI: 10.3389/fneur.2023.1213377] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 07/25/2023] [Indexed: 08/29/2023] Open
Abstract
Background and goal In vivo characterization of brain lesion types in multiple sclerosis (MS) has been an ongoing challenge. Based on verified texture analysis measures from clinical magnetic resonance imaging (MRI), this study aimed to develop a method to identify two extremes of brain MS lesions that were approximately severely demyelinated (sDEM) and highly remyelinated (hREM), and compare them in terms of common clinical variables. Method Texture analysis used an optimized gray-level co-occurrence matrix (GLCM) method based on FLAIR MRI from 200 relapsing-remitting MS participants. Two top-performing metrics were calculated: texture contrast and dissimilarity. Lesion identification applied a percentile approach according to texture values calculated: ≤ 25 percentile for hREM and ≥75 percentile for sDEM. Results The sDEM had a greater total normalized volume yet smaller average size, and worse MRI texture than hREM. In lesion distribution mapping, the two lesion types appeared to overlap largely in location and were present the most in the corpus callosum and periventricular regions. Further, in sDEM, the normalized volume was greater and in hREM, the average size was smaller in men than women. There were no other significant results in clinical variable-associated analyses. Conclusion Percentile statistics of competitive MRI texture measures may be a promising method for probing select types of brain MS lesion pathology. Associated findings can provide another useful dimension for improved measurement and monitoring of disease activity in MS. The different characteristics of sDEM and hREM between men and women likely adds new information to the literature, deserving further confirmation.
Collapse
Affiliation(s)
- Zahra Hosseinpour
- Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
| | - Olayinka Oladosu
- Department of Neuroscience, Faculty of Graduate Studies, University of Calgary, Calgary, AB, Canada
| | - Wei-qiao Liu
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
| | - G. Bruce Pike
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
- Department of Radiology, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - V. Wee Yong
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Luanne M. Metz
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Yunyan Zhang
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
- Department of Radiology, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
6
|
Ismail MA, Elsayed NM. Diffusion-Weighted Images and Contrast-Enhanced MRI in the Diagnosis of Different Stages of Multiple Sclerosis of the Central Nervous System. Cureus 2023; 15:e41650. [PMID: 37575819 PMCID: PMC10420334 DOI: 10.7759/cureus.41650] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/09/2023] [Indexed: 08/15/2023] Open
Abstract
Introduction Multiple sclerosis (MS) is one of the most prevalent disorders of the central nervous system (CNS), and it can be observed in the field of radiological cross-sectional magnetic resonance imaging (MRI). The prevalence of MS in Saudi Arabia has increased as compared to the past few years. MRI is the gold standard non-invasive modality of choice in MS diagnosis according to the National Multiple Sclerosis Society (NMSS), New York City. This study aimed to highlight the significance of using diffusion-weighted images (DWIs) and the use of contrast media in the MS protocol, as well as the importance of identifying the suitable time of imaging after contrast enhancement to detect active lesions. Methods A retrospective cross-sectional study was conducted of 100 MS patients with an age range of 17 to 56 years. The data set included 41 active cases and 59 inactive cases. All patients had an MRI standard protocol of both the brain and spine in addition to DWI sequence and contrast agent (CA) injection, with images taken in early and delayed time. Results Of the patients, 71% were female and 29% were male. Active MS disease was more significant at younger ages than at older ages. Active lesions were significantly enhanced in delayed contrast images and showed high signal intensity in both the DWI and apparent diffusion coefficient (ADC) map, while inactive lesions showed no enhancement after contrast injection and showed an iso-signal intensity in both the DWI and ADC map. Conclusion The use of CA has developed over the years in the diagnosis of MS patients. In this study, the relationship between active lesions, DWI, and delayed contrast enhancement is very strong. In future research, we recommend adding a DWI sequence for the suspected active MS spine lesions in addition to delayed enhancement time in active MS after contrast injection to increase MRI sensitivity toward active MS lesions of the brain and spinal cord as well.
Collapse
Affiliation(s)
- Mashael A Ismail
- Radiologic Sciences, Faculty of Applied Medical Sciences, King Abdullah Medical Complex, Ministry of Health, Jeddah, SAU
| | - Naglaa M Elsayed
- Radiologic Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, SAU
- Diagnostic Radiology, Faculty of Medicine, Cairo University, Cairo, EGY
| |
Collapse
|
7
|
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: 12] [Impact Index Per Article: 6.0] [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
|
8
|
Gudkov SV, Burmistrov DE, Kondakova EV, Sarimov RM, Yarkov RS, Franceschi C, Vedunova MV. An emerging role of astrocytes in aging/neuroinflammation and gut-brain axis with consequences on sleep and sleep disorders. Ageing Res Rev 2023; 83:101775. [PMID: 36334910 DOI: 10.1016/j.arr.2022.101775] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 10/05/2022] [Accepted: 10/30/2022] [Indexed: 11/18/2022]
Abstract
Understanding the role of astrocytes in the central nervous system has changed dramatically over the last decade. The accumulating findings indicate that glial cells are involved not only in the maintenance of metabolic and ionic homeostasis and in the implementation of trophic functions but also in cognitive functions and information processing in the brain. Currently, there are some controversies regarding the role of astrocytes in complex processes such as aging of the nervous system and the pathogenesis of age-related neurodegenerative diseases. Many findings confirm the important functional role of astrocytes in age-related brain changes, including sleep disturbance and the development of neurodegenerative diseases and particularly Alzheimer's disease. Until recent years, neurobiological research has focused mainly on neuron-glial interactions, in which individual astrocytes locally modulate neuronal activity and communication between neurons. The review considers the role of astrocytes in the physiology of sleep and as an important "player" in the development of neurodegenerative diseases. In addition, the features of the astrocytic network reorganization during aging are discussed.
Collapse
Affiliation(s)
- Sergey V Gudkov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilov str., 119991 Moscow, Russia; Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin ave., 603022 Nizhny Novgorod, Russia.
| | - Dmitriy E Burmistrov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilov str., 119991 Moscow, Russia.
| | - Elena V Kondakova
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin ave., 603022 Nizhny Novgorod, Russia.
| | - Ruslan M Sarimov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilov str., 119991 Moscow, Russia.
| | - Roman S Yarkov
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin ave., 603022 Nizhny Novgorod, Russia.
| | - Claudio Franceschi
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin ave., 603022 Nizhny Novgorod, Russia.
| | - Maria V Vedunova
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin ave., 603022 Nizhny Novgorod, Russia.
| |
Collapse
|
9
|
Anti-Inflammatory Effects of GLP-1 Receptor Activation in the Brain in Neurodegenerative Diseases. Int J Mol Sci 2022; 23:ijms23179583. [PMID: 36076972 PMCID: PMC9455625 DOI: 10.3390/ijms23179583] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/17/2022] [Accepted: 08/21/2022] [Indexed: 11/16/2022] Open
Abstract
The glucagon-like peptide-1 (GLP-1) is a pleiotropic hormone well known for its incretin effect in the glucose-dependent stimulation of insulin secretion. However, GLP-1 is also produced in the brain and displays a critical role in neuroprotection and inflammation by activating the GLP-1 receptor signaling pathways. Several studies in vivo and in vitro using preclinical models of neurodegenerative diseases show that GLP-1R activation has anti-inflammatory properties. This review explores the molecular mechanistic action of GLP-1 RAS in relation to inflammation in the brain. These findings update our knowledge of the potential benefits of GLP-1RAS actions in reducing the inflammatory response. These molecules emerge as a potential therapeutic tool in treating neurodegenerative diseases and neuroinflammatory pathologies.
Collapse
|
10
|
Dubreuil-Vall L, Laabs TL, Eyre HA, Smith E, Catuara-Solarz S. Sex differences in invasive and noninvasive neurotechnologies. SEX AND GENDER BIAS IN TECHNOLOGY AND ARTIFICIAL INTELLIGENCE 2022:133-160. [DOI: 10.1016/b978-0-12-821392-6.00008-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
11
|
Graham BE, Plotkin B, Muglia L, Moore JH, Williams SM. Estimating prevalence of human traits among populations from polygenic risk scores. Hum Genomics 2021; 15:70. [PMID: 34903281 PMCID: PMC8670062 DOI: 10.1186/s40246-021-00370-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 11/27/2021] [Indexed: 11/21/2022] Open
Abstract
The genetic basis of phenotypic variation across populations has not been well explained for most traits. Several factors may cause disparities, from variation in environments to divergent population genetic structure. We hypothesized that a population-level polygenic risk score (PRS) can explain phenotypic variation among geographic populations based solely on risk allele frequencies. We applied a population-specific PRS (psPRS) to 26 populations from the 1000 Genomes to four phenotypes: lactase persistence (LP), melanoma, multiple sclerosis (MS) and height. Our models assumed additive genetic architecture among the polymorphisms in the psPRSs, as is convention. Linear psPRSs explained a significant proportion of trait variance ranging from 0.32 for height in men to 0.88 for melanoma. The best models for LP and height were linear, while those for melanoma and MS were nonlinear. As not all variants in a PRS may confer similar, or even any, risk among diverse populations, we also filtered out SNPs to assess whether variance explained was improved using psPRSs with fewer SNPs. Variance explained usually improved with fewer SNPs in the psPRS and was as high as 0.99 for height in men using only 548 of the initial 4208 SNPs. That reducing SNPs improves psPRSs performance may indicate that missing heritability is partially due to complex architecture that does not mandate additivity, undiscovered variants or spurious associations in the databases. We demonstrated that PRS-based analyses can be used across diverse populations and phenotypes for population prediction and that these comparisons can identify the universal risk variants.
Collapse
Affiliation(s)
- Britney E Graham
- Departments of Population and Quantitative Health Sciences and Genetics and Genome Scenes, Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, 44106, USA
- Systems Biology and Bioinformatics, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Brian Plotkin
- Departments of Population and Quantitative Health Sciences and Genetics and Genome Scenes, Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Louis Muglia
- Burroughs Wellcome Fund, Research Triangle Park, NC, 27614, USA
- Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
| | - Jason H Moore
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Scott M Williams
- Departments of Population and Quantitative Health Sciences and Genetics and Genome Scenes, Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, 44106, USA.
| |
Collapse
|
12
|
Roeder HJ, Leira EC. Effects of the Menstrual Cycle on Neurological Disorders. Curr Neurol Neurosci Rep 2021; 21:34. [PMID: 33970361 DOI: 10.1007/s11910-021-01115-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2021] [Indexed: 12/26/2022]
Abstract
PURPOSE OF REVIEW The menstrual cycle involves recurrent fluctuations in hormone levels and temperature via neuroendocrine feedback loops. This paper reviews the impact of the menstrual cycle on several common neurological conditions, including migraine, seizures, multiple sclerosis, stroke, and Parkinson's disease. RECENT FINDINGS The ovarian steroid hormones, estrogen and progesterone, have protean effects on central nervous system functioning that can impact the likelihood, severity, and presentation of many neurological diseases. Hormonal therapies have been explored as a potential treatment for many neurological diseases with varying degrees of evidence and success. Neurological conditions also impact women's reproductive health, and the cessation of ovarian function with menopause may also alter the course of neurological diseases. Medication selection must consider hormonal effects on metabolism and the potential for adverse drug reactions related to menstruation, fertility, and pregnancy outcomes. Novel medications with selective affinity for hormonal receptors are desirable. Neurologists and gynecologists must collaborate to provide optimal care for women with neurological disorders.
Collapse
Affiliation(s)
- Hannah J Roeder
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Enrique C Leira
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA. .,Department of Neurosurgery, Carver College of Medicine, University of Iowa, Iowa City, IA, USA. .,Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
13
|
Chowen JA, Garcia-Segura LM. Role of glial cells in the generation of sex differences in neurodegenerative diseases and brain aging. Mech Ageing Dev 2021; 196:111473. [PMID: 33766745 DOI: 10.1016/j.mad.2021.111473] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/14/2021] [Accepted: 03/16/2021] [Indexed: 12/11/2022]
Abstract
Diseases and aging-associated alterations of the nervous system often show sex-specific characteristics. Glial cells play a major role in the endogenous homeostatic response of neural tissue, and sex differences in the glial transcriptome and function have been described. Therefore, the possible role of these cells in the generation of sex differences in pathological alterations of the nervous system is reviewed here. Studies have shown that glia react to pathological insults with sex-specific neuroprotective and regenerative effects. At least three factors determine this sex-specific response of glia: sex chromosome genes, gonadal hormones and neuroactive steroid hormone metabolites. The sex chromosome complement determines differences in the transcriptional responses in glia after brain injury, while gonadal hormones and their metabolites activate sex-specific neuroprotective mechanisms in these cells. Since the sex-specific neuroprotective and regenerative activity of glial cells causes sex differences in the pathological alterations of the nervous system, glia may represent a relevant target for sex-specific therapeutic interventions.
Collapse
Affiliation(s)
- Julie A Chowen
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación la Princesa, Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutriciόn (CIBEROBN), Instituto de Salud Carlos III, and IMDEA Food Institute, CEIUAM+CSIC, Madrid, Spain.
| | - Luis M Garcia-Segura
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC) and Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
14
|
Midavaine É, Côté J, Marchand S, Sarret P. Glial and neuroimmune cell choreography in sexually dimorphic pain signaling. Neurosci Biobehav Rev 2021; 125:168-192. [PMID: 33582232 DOI: 10.1016/j.neubiorev.2021.01.023] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 11/03/2020] [Accepted: 01/25/2021] [Indexed: 12/17/2022]
Abstract
Chronic pain is a major global health issue that affects all populations regardless of sex, age, ethnicity/race, or country of origin, leading to persistent physical and emotional distress and to the loss of patients' autonomy and quality of life. Despite tremendous efforts in the elucidation of the mechanisms contributing to the pathogenesis of chronic pain, the identification of new potential pain targets, and the development of novel analgesics, the pharmacological treatment options available for pain management remain limited, and most novel pain medications have failed to achieve advanced clinical development, leaving many patients with unbearable and undermanaged pain. Sex-specific susceptibility to chronic pain conditions as well as sex differences in pain sensitivity, pain tolerance and analgesic efficacy are increasingly recognized in the literature and have thus prompted scientists to seek mechanistic explanations. Hence, recent findings have highlighted that the signaling mechanisms underlying pain hypersensitivity are sexually dimorphic, which sheds light on the importance of conducting preclinical and clinical pain research on both sexes and of developing sex-specific pain medications. This review thus focuses on the clinical and preclinical evidence supporting the existence of sex differences in pain neurobiology. Attention is drawn to the sexually dimorphic role of glial and immune cells, which are both recognized as key players in neuroglial maladaptive plasticity at the origin of the transition from acute pain to chronic pathological pain. Growing evidence notably attributes to microglial cells a pivotal role in the sexually dimorphic pain phenotype and in the sexually dimorphic analgesic efficacy of opioids. This review also summarizes the recent advances in understanding the pathobiology underpinning the development of pain hypersensitivity in both males and females in different types of pain conditions, with particular emphasis on the mechanistic signaling pathways driving sexually dimorphic pain responses.
Collapse
Affiliation(s)
- Élora Midavaine
- Department of Pharmacology-Physiology, Institut de pharmacologie de Sherbrooke, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada; Centre de recherche du Centre hospitalier universitaire de Sherbrooke, CIUSSS de l'Estrie - CHUS, Sherbrooke, Québec, Canada.
| | - Jérôme Côté
- Department of Pharmacology-Physiology, Institut de pharmacologie de Sherbrooke, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada; Centre de recherche du Centre hospitalier universitaire de Sherbrooke, CIUSSS de l'Estrie - CHUS, Sherbrooke, Québec, Canada
| | - Serge Marchand
- Department of Pharmacology-Physiology, Institut de pharmacologie de Sherbrooke, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada; Centre de recherche du Centre hospitalier universitaire de Sherbrooke, CIUSSS de l'Estrie - CHUS, Sherbrooke, Québec, Canada
| | - Philippe Sarret
- Department of Pharmacology-Physiology, Institut de pharmacologie de Sherbrooke, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada; Centre de recherche du Centre hospitalier universitaire de Sherbrooke, CIUSSS de l'Estrie - CHUS, Sherbrooke, Québec, Canada.
| |
Collapse
|
15
|
ATEŞ Y, ÜNLÜER NÖ. An investigation of knee position sense, balance, and dual task performance in different phases of menstrual cycle in females with multiple sclerosis: a pilot study. Mult Scler Relat Disord 2020; 44:102235. [DOI: 10.1016/j.msard.2020.102235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 05/19/2020] [Accepted: 05/21/2020] [Indexed: 10/24/2022]
|
16
|
Metzger-Peter K, Kremer LD, Edan G, Loureiro De Sousa P, Lamy J, Bagnard D, Mensah-Nyagan AG, Tricard T, Mathey G, Debouverie M, Berger E, Kerbrat A, Meyer N, De Seze J, Collongues N. The TOTEM RRMS (Testosterone Treatment on neuroprotection and Myelin Repair in Relapsing Remitting Multiple Sclerosis) trial: study protocol for a randomized, double-blind, placebo-controlled trial. Trials 2020; 21:591. [PMID: 32600454 PMCID: PMC7322908 DOI: 10.1186/s13063-020-04517-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 06/15/2020] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Central nervous system damage in multiple sclerosis (MS) is responsible for serious deficiencies. Current therapies are focused on the treatment of inflammation; however, there is an urgent need for innovative therapies promoting neuroregeneration, particularly myelin repair. It is demonstrated that testosterone can act through neural androgen receptors and several clinical observations stimulated an interest in the potential protective effects of testosterone treatment for MS. Here, we sought to demonstrate the effects of a testosterone supplementation in testosterone-deficient men with relapsing-remitting MS. METHODS/DESIGN This report presents the rationale and methodology of TOTEM RRMS, a French, phase 2, multicenter, randomized, placebo-controlled, and double-blind trial, which aims to prevent the progression of MS in men with low testosterone levels by administration of testosterone undecanoate, who were kept under natalizumab (Tysabri®) to overcome the anti-inflammatory effect of testosterone. Forty patients will be randomized into two groups receiving either a testosterone treatment (Nebido®) or a matching placebo. The intervention period for each group will last 66 weeks (treatment will be injected at baseline, week 6, and then every 12 weeks). The main objective is to determine the neuroprotective and remyelinating effects of testosterone using tensor diffusion imaging techniques and thalamic atrophy analyses. As secondary objectives, impacts of the testosterone supplementation will be studied using other conventional and unconventional MRI parameters and with clinical outcomes. DISCUSSION The action of testosterone is observed in different experimental autoimmune encephalomyelitis models and epidemiological studies in humans. However, despite several preclinical data and some small clinical trials in MS, clear evidence for a therapeutic effect of hormone therapy is still missing. Therefore, our goal is to demonstrate the effects of testosterone therapies in MS. As there is no effective treatment currently available on fatigue in MS, careful attention should also be paid to secondary endpoints: fatigue, cognitive functions, and other symptoms that may improve life quality. Assuming a positive outcome of the trial, this treatment could be considered as a new neuroprotective and remyelinating therapy in relapsing-remitting MS and could be applicable to other demyelinating diseases. TRIAL REGISTRATION ClinicalTrials.gov NCT03910738. Registered on 10 April 2019.
Collapse
Affiliation(s)
| | - Laurent Daniel Kremer
- Departement of Neurology, Hôpital de Hautepierre, University Hospital of Strasbourg, Strasbourg, France
| | - Gilles Edan
- Departement of Neurology, Hôpital Pontchaillou, University Hospital of Rennes, Rennes, France
| | - Paulo Loureiro De Sousa
- Laboratory of Engineering Sciences, Computer Science and Imagery (ICube), CNRS, Institute of Biological Physics, University of Strasbourg, Strasbourg, France
| | - Julien Lamy
- Laboratory of Engineering Sciences, Computer Science and Imagery (ICube), CNRS, Institute of Biological Physics, University of Strasbourg, Strasbourg, France
| | - Dominique Bagnard
- Departement of Myelin Biopathology, Neuroprotection and Therapeutic Strategies, UMR_S Inserm 1119, Strasbourg, France
| | - Ayikoe-Guy Mensah-Nyagan
- Departement of Myelin Biopathology, Neuroprotection and Therapeutic Strategies, UMR_S Inserm 1119, Strasbourg, France
| | - Thibault Tricard
- Departement of Urological Surgery, Nouvel Hôpital Civil, University Hospital of Strasbourg, Strasbourg, France
| | - Guillaume Mathey
- Departement of Neurology, Hôpital Central, University Hospital of Nancy, Nancy, France
| | - Marc Debouverie
- Departement of Neurology, Hôpital Central, University Hospital of Nancy, Nancy, France
| | - Eric Berger
- Departement of Neurology, Hôpital Jean Minjoz, University Hospital of Besançon, Besançon, France
| | - Anne Kerbrat
- Department of Neurology, Hôpital de Pontchaillou, University Hospital of Rennes, Rennes, France
| | - Nicolas Meyer
- Departement of Public Health, GMRC University Hospital of Strasbourg, Strasbourg, France
| | - Jérôme De Seze
- Centre d᾿Investigation Clinique INSERM 1434, Strasbourg, France.,Departement of Neurology, Hôpital de Hautepierre, University Hospital of Strasbourg, Strasbourg, France
| | - Nicolas Collongues
- Centre d᾿Investigation Clinique INSERM 1434, Strasbourg, France.,Departement of Neurology, Hôpital de Hautepierre, University Hospital of Strasbourg, Strasbourg, France
| |
Collapse
|
17
|
Al Wutayd O. Association of infections with multiple sclerosis in Gulf Cooperation Council countries: a review. J Int Med Res 2019; 48:300060519884151. [PMID: 31880177 PMCID: PMC7607054 DOI: 10.1177/0300060519884151] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disease that affects the central nervous system, causing inflammation, demyelination, and neurodegeneration. Infection can play a role in its etiology. Herein, a review is presented of studies that have reported an association between infection and MS risk in countries of the Arabian Gulf region. Searches of the PubMed, Google Scholar, and Science Direct databases were carried out using various search terms, and relevant studies published through January 2019 on the epidemiology of MS in Gulf Cooperation Council countries identified. MS has been found to be associated with measles in Saudi Arabia and Epstein–Barr virus in Kuwait whereas no association has been identified between risk of MS and varicella-zoster virus, mumps, or human herpesvirus-6. However, few epidemiological studies on this topic have been conducted in countries of the Gulf region. Longitudinal and serological studies to establish robust evidence between infection and risk of MS are highly recommended, and a regional MS registry is needed.
Collapse
Affiliation(s)
- O Al Wutayd
- O Al Wutayd, Department of Family and Community Medicine, Unaizah College of Medicine, Qassim University, P.O. Box 3174, Unaizah 51911, Saudi Arabia.
| |
Collapse
|
18
|
|
19
|
Metabolomic Signature in Sera of Multiple Sclerosis Patients during Pregnancy. Int J Mol Sci 2018; 19:ijms19113589. [PMID: 30441762 PMCID: PMC6274842 DOI: 10.3390/ijms19113589] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 11/05/2018] [Accepted: 11/11/2018] [Indexed: 12/14/2022] Open
Abstract
Multiple sclerosis (MuS) is an autoimmune disease of the central nervous system characterized by neuroinflammation, neurodegeneration, and degradation of the myelin sheath. Epidemiological studies have shown that the female gender is more susceptible than the male gender to MuS development, with a female-to-male ratio of 2:1. Despite this high onset, women have a better prognosis than men, and the frequency of the relapsing phase decreases during pregnancy, while it increases soon after birth. Therefore, it is interesting to investigate hormonal fluctuations during pregnancy and whether they correlate with metabolic signatures. To gain a deeper inside into the biochemical mechanism of such a multifactorial disease, we adopted targeted metabolomics approaches for the determination of many serum metabolites in 12 pregnant women affected by MuS by mass spectrometry analysis. Our data show a characteristic hormonal fluctuation for estrogens and progesterone, as expected. They also highlight other interesting hormonal alterations for cortisol, corticosterone, 11-deoxycortisol, 4-androstene-3,17-dione, testosterone, and 17α-hydroxyprogesterone. Furthermore, a negative correlation with progesterone levels was observed for amino acids and for acylcarnitines, while an imbalance of different sphingolipids pathways was found during pregnancy. In conclusion, these data are in agreement with the characteristic clinical signs of MuS patients during pregnancy and, if confirmed, they may add an important tessera in the complex mosaic of maternal neuroprotection.
Collapse
|
20
|
Badihian S, Manouchehri N, Mirmosayyeb O, Ashtari F, Shaygannejad V. Neuromyelitis optica spectrum disorder and menstruation. Rev Neurol (Paris) 2018; 174:716-721. [PMID: 30049560 DOI: 10.1016/j.neurol.2018.01.373] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 12/13/2017] [Accepted: 01/10/2018] [Indexed: 01/01/2023]
Abstract
BACKGROUND Gender issues and the female preponderance in neuromyelitis optica spectrum disorder (NMOSD) have been investigated before, yet the interplay between NMOSD and menstrual characteristics has remained unknown. Thus, the aim was to compare menstrual cycle patterns and their symptoms in NMOSD patients and healthy women. METHODS This cross-sectional study was conducted during 2015-2016 in Isfahan, Iran, and included female patients aged>14years with a diagnosis of NMOSD and healthy subjects as controls. Data regarding age at menarche, menstrual characteristics, history of premenstrual syndrome (PMS) and possible perimenstrual symptoms were collected. Also, NMOSD patients were asked to report changes in their menstrual cycles after onset of the disorder. RESULTS The final study population included 32 NMOSD and 33 healthy controls. These groups did not differ regarding their demographics (P>0.05), and age at menarche in the NMOSD and control groups was 13.31±1.49 years and 13.48±1.44 years, respectively (P=0.637). The controls experienced PMS more frequently (78.8% vs. 40.6% in the NMOSD patients; P=0.03), with no significant differences in other menstrual features between groups (P>0.05). However, changes in menstruation after NMOSD onset were reported by 43.8% of patients, with an increase in menstrual irregularities from 15.6% to 43.7% (P=0.012); other menstrual characteristics did not differ after disease onset (P>0.05). CONCLUSION Menstruation do not differ between healthy controls and NMOSD patients before the onset of disease whereas, after its onset, those affected experienced more irregularities in their menstrual cycles. This may be an effect of NMOSD and its underlying disorders on menstruation and suggests that further interventions may be required for affected women.
Collapse
Affiliation(s)
- Shervin Badihian
- Isfahan Neurosciences Research Center, Alzahra Research Institute, Isfahan University of Medical Sciences, 81746-73461 Isfahan, Iran; Student Research Committee, Isfahan University of Medical Sciences, 81746-73461 Isfahan, Iran
| | - Navid Manouchehri
- Isfahan Neurosciences Research Center, Alzahra Research Institute, Isfahan University of Medical Sciences, 81746-73461 Isfahan, Iran; Student Research Committee, Isfahan University of Medical Sciences, 81746-73461 Isfahan, Iran
| | - Omid Mirmosayyeb
- Isfahan Neurosciences Research Center, Alzahra Research Institute, Isfahan University of Medical Sciences, 81746-73461 Isfahan, Iran; Student Research Committee, Isfahan University of Medical Sciences, 81746-73461 Isfahan, Iran
| | - Fereshteh Ashtari
- Isfahan Neurosciences Research Center, Alzahra Research Institute, Isfahan University of Medical Sciences, 81746-73461 Isfahan, Iran; Department of Neurology, School of Medicine, Isfahan University of Medical Sciences, 81746-73461 Isfahan, Iran
| | - Vahid Shaygannejad
- Isfahan Neurosciences Research Center, Alzahra Research Institute, Isfahan University of Medical Sciences, 81746-73461 Isfahan, Iran; Department of Neurology, School of Medicine, Isfahan University of Medical Sciences, 81746-73461 Isfahan, Iran.
| |
Collapse
|
21
|
Collongues N, Patte-Mensah C, De Seze J, Mensah-Nyagan AG, Derfuss T. Testosterone and estrogen in multiple sclerosis: from pathophysiology to therapeutics. Expert Rev Neurother 2018; 18:515-522. [PMID: 29799288 DOI: 10.1080/14737175.2018.1481390] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Neuroprotection and remyelination are two unmet needs in the treatment of multiple sclerosis (MS). Therapeutic potential has been identified with sexual hormones, supported in women by a decrease in MS activity during the pregnancy, in men by a greater severity of symptoms and a faster progression than in women. Areas covered: The therapeutic effect of testosterone and estrogens is reviewed. Both hormones have demonstrated an anti-inflammatory effect. Testosterone has an effect in protecting neurons in culture against glutamate-induced toxicity and oxidative stress, and stimulates myelin formation and regeneration mediated through the neural androgen receptor. In experimental autoimmune encephalomyelitis model, estrogens significantly decrease inflammation in the central nervous system via ERα, while its action on ERβ leads to myelin and axon reparation. Estriol therapy in two phase 2 trials showed a decrease in clinical disease activity and inflammatory parameters in MRI. However, evidence of a therapeutic effect of testosterone is scarce. Expert commentary: Phase 3 trials with estriol as an add-on supplementation are now mandatory. Testosterone is another candidate to be tested in phase 2 trials. These hormones should be considered as an adjunctive therapy. New validated tools are needed to assess their effect on neuroprotection and remyelination.
Collapse
Affiliation(s)
- Nicolas Collongues
- a Biopathology of Myelin, Neuroprotection and Therapeutic Strategies , INSERM U1119, University Hospital of Strasbourg , Strasbourg , France.,b Department of Neurology , University Hospital of Strasbourg , Strasbourg , France.,c Clinical Investigation Center , INSERM U1434, University Hospital of Strasbourg , Strasbourg , France
| | - Christine Patte-Mensah
- a Biopathology of Myelin, Neuroprotection and Therapeutic Strategies , INSERM U1119, University Hospital of Strasbourg , Strasbourg , France
| | - Jérôme De Seze
- a Biopathology of Myelin, Neuroprotection and Therapeutic Strategies , INSERM U1119, University Hospital of Strasbourg , Strasbourg , France.,b Department of Neurology , University Hospital of Strasbourg , Strasbourg , France.,c Clinical Investigation Center , INSERM U1434, University Hospital of Strasbourg , Strasbourg , France
| | - Ayikoe-Guy Mensah-Nyagan
- a Biopathology of Myelin, Neuroprotection and Therapeutic Strategies , INSERM U1119, University Hospital of Strasbourg , Strasbourg , France
| | - Tobias Derfuss
- d Departments of Neurology and Biomedicine , University Hospital Basel , Basel , Switzerland
| |
Collapse
|
22
|
The interplay of multiple sclerosis and menstrual cycle: Which one affects the other one? Mult Scler Relat Disord 2018; 21:46-50. [DOI: 10.1016/j.msard.2018.01.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 01/05/2018] [Accepted: 01/21/2018] [Indexed: 01/09/2023]
|
23
|
Ritzel RM, Patel AR, Spychala M, Verma R, Crapser J, Koellhoffer EC, Schrecengost A, Jellison ER, Zhu L, Venna VR, McCullough LD. Multiparity improves outcomes after cerebral ischemia in female mice despite features of increased metabovascular risk. Proc Natl Acad Sci U S A 2017; 114:E5673-E5682. [PMID: 28645895 PMCID: PMC5514696 DOI: 10.1073/pnas.1607002114] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Females show a varying degree of ischemic sensitivity throughout their lifespan, which is not fully explained by hormonal or genetic factors. Epidemiological data suggest that sex-specific life experiences such as pregnancy increase stroke risk. This work evaluated the role of parity on stroke outcome. Age-matched virgin (i.e., nulliparous) and multiparous mice were subjected to 60 min of reversible middle cerebral artery occlusion and evaluated for infarct volume, behavioral recovery, and inflammation. Using an established mating paradigm, fetal microchimeric cells present in maternal mice were also tracked after parturition and stroke. Parity was associated with sedentary behavior, weight gain, and higher triglyceride and cholesterol levels. The multiparous brain exhibited features of immune suppression, with dampened baseline microglial activity. After acute stroke, multiparous mice had smaller infarcts, less glial activation, and less behavioral impairment in the critical recovery window of 72 h. Behavioral recovery was significantly better in multiparous females compared with nulliparous mice 1 mo after stroke. This recovery was accompanied by an increase in poststroke angiogenesis that was correlated with improved performance on sensorimotor and cognitive tests. Multiparous mice had higher levels of VEGF, both at baseline and after stroke. GFP+ fetal cells were detected in the blood and migrated to areas of tissue injury where they adopted endothelial morphology 30 d after injury. Reproductive experience has profound and complex effects on neurovascular health and disease. Inclusion of female mice with reproductive experience in preclinical studies may better reflect the life-long patterning of ischemic stroke risk in women.
Collapse
Affiliation(s)
- Rodney M Ritzel
- Department of Anesthesiology, Center for Shock, Trauma, and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Anita R Patel
- Neuroscience Department, University of Connecticut Health Center, Farmington, CT 06030
| | - Monica Spychala
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Rajkumar Verma
- Neuroscience Department, University of Connecticut Health Center, Farmington, CT 06030
| | - Joshua Crapser
- Neuroscience Department, University of Connecticut Health Center, Farmington, CT 06030
| | - Edward C Koellhoffer
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Anna Schrecengost
- Neuroscience Department, University of Connecticut Health Center, Farmington, CT 06030
| | - Evan R Jellison
- Immunology Department, University of Connecticut Health Center, Farmington, CT 06030
| | - Liang Zhu
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Venugopal Reddy Venna
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Louise D McCullough
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030;
| |
Collapse
|
24
|
Borisow N, Kleiter I, Gahlen A, Fischer K, Wernecke KD, Pache F, Ruprecht K, Havla J, Krumbholz M, Kümpfel T, Aktas O, Ringelstein M, Geis C, Kleinschnitz C, Berthele A, Hemmer B, Angstwurm K, Weissert R, Stellmann JP, Schuster S, Stangel M, Lauda F, Tumani H, Mayer C, Zeltner L, Ziemann U, Linker RA, Schwab M, Marziniak M, Then Bergh F, Hofstadt-van Oy U, Neuhaus O, Winkelmann A, Marouf W, Rückriem L, Faiss J, Wildemann B, Paul F, Jarius S, Trebst C, Hellwig K. Influence of female sex and fertile age on neuromyelitis optica spectrum disorders. Mult Scler 2016; 23:1092-1103. [PMID: 27758954 DOI: 10.1177/1352458516671203] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Gender and age at onset are important epidemiological factors influencing prevalence, clinical presentation, and treatment response in autoimmune diseases. OBJECTIVE To evaluate the impact of female sex and fertile age on aquaporin-4-antibody (AQP4-ab) status, attack localization, and response to attack treatment in patients with neuromyelitis optica (NMO) and its spectrum disorders (neuromyelitis optica spectrum disorder (NMOSD)). METHODS Female-to-male ratios, diagnosis at last visit (NMO vs NMOSD), attack localization, attack treatment, and outcome were compared according to sex and age at disease or attack onset. RESULTS A total of 186 NMO/SD patients (82% female) were included. In AQP4-ab-positive patients, female predominance was most pronounced during fertile age (female-to-male ratio 23:1). Female patients were more likely to be positive for AQP4-abs (92% vs 55%; p < 0.001). Interval between onset and diagnosis of NMO/SD was longer in women than in men (mean 54 vs 27 months; p = 0.023). In women, attacks occurring ⩽40 years of age were more likely to show complete remission ( p = 0.003) and better response to high-dose intravenous steroids ( p = 0.005) compared to woman at >40 years. CONCLUSION Our data suggest an influence of sex and age on susceptibility to AQP4-ab-positive NMO/SD. Genetic and hormonal factors might contribute to pathophysiology of NMO/SD.
Collapse
Affiliation(s)
- Nadja Borisow
- NeuroCure Clinical Research Center and Clinical and Experimental Multiple Sclerosis Research Center, Department of Neurology, Charité University Medicine Berlin, Berlin, Germany
| | - Ingo Kleiter
- Department of Neurology, St. Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - Anna Gahlen
- Department of Neurology, St. Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - Katrin Fischer
- Department of Neurology, Asklepios Fachklinikum Teupitz, Teupitz, Germany
| | | | - Florence Pache
- NeuroCure Clinical Research Center and Clinical and Experimental Multiple Sclerosis Research Center, Department of Neurology, Charité University Medicine Berlin, Berlin, Germany
| | - Klemens Ruprecht
- Department of Neurology and Clinical and Experimental Multiple Sclerosis Research Center, Charité University Medicine Berlin, Berlin, Germany
| | - Joachim Havla
- Institute of Clinical Neuroimmunology, Medical Campus Grosshadern, Ludwig Maximilians University of Munich, Munich, Germany
| | - Markus Krumbholz
- Institute of Clinical Neuroimmunology, Medical Campus Grosshadern, Ludwig Maximilians University of Munich, Munich, Germany
| | - Tania Kümpfel
- Institute of Clinical Neuroimmunology, Medical Campus Grosshadern, Ludwig Maximilians University of Munich, Munich, Germany
| | - Orhan Aktas
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Marius Ringelstein
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Christian Geis
- Department of Neurology, University Hospital of Würzburg, Würzburg, Germany/Hans-Berger Department of Neurology and Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany
| | | | - Achim Berthele
- Department of Neurology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Bernhard Hemmer
- Department of Neurology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany/Munich Cluster for Systems Neurology (SyNergy), Technische Universität München, Munich, Germany
| | - Klemens Angstwurm
- Department of Neurology, University Hospital Regensburg, Regensburg, Germany
| | - Robert Weissert
- Department of Neurology, University Hospital Regensburg, Regensburg, Germany
| | - Jan-Patrick Stellmann
- Institute of Neuroimmunology and MS (INIMS), University Medical Center Hamburg-Eppendorf, Hamburg, Germany/Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Simon Schuster
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Martin Stangel
- Department of Clinical Neuroimmunology and Neurochemistry and Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Florian Lauda
- Department of Neurology, University of Ulm, Ulm, Germany
| | - Hayrettin Tumani
- Department of Neurology at RKU and Specialty Clinic of Neurology Dietenbronn, University of Ulm, Ulm, Germany
| | - Christoph Mayer
- Department of Neurology, Goethe University Frankfurt, Frankfurt, Germany
| | - Lena Zeltner
- Department of Neurology and Stroke and Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Ulf Ziemann
- Department of Neurology and Stroke and Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Ralf A Linker
- Department of Neurology, Friedrich-Alexander University Erlangen-Nüremberg, Erlangen, Germany
| | - Matthias Schwab
- Hans-Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Martin Marziniak
- Department of Neurology, University of Münster, Münster, Germany/Department of Neurology and Neurological Intensive Care, Isar-Amper-Clinic, Munich-East, Haar, Germany
| | | | - Ulrich Hofstadt-van Oy
- Department of Neurology, Klinikum Bayreuth, Bayreuth, Germany/Department of Neurology, Klinikum Westfalen, Dortmund, Germany
| | - Oliver Neuhaus
- Department of Neurology, SRH Krankenhaus Sigmaringen, Sigmaringen, Germany
| | | | - Wael Marouf
- Department of Neurology, HELIOS Hanseklinikum Stralsund, Stralsund, Germany
| | - Lioba Rückriem
- Department of Neurology, MediClin Hedon Klinik, Lingen, Germany
| | - Jürgen Faiss
- Department of Neurology, Asklepios Fachklinikum Teupitz, Teupitz, Germany
| | | | - Friedemann Paul
- NeuroCure Clinical Research Center and Clinical and Experimental Multiple Sclerosis Research Center, Department of Neurology, Charité University Medicine Berlin, Berlin, Germany/Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité University Medicine Berlin, Berlin, Germany
| | - Sven Jarius
- Department of Neurology, Heidelberg University, Heidelberg, Germany
| | - Corinna Trebst
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Kerstin Hellwig
- Department of Neurology, St. Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | | |
Collapse
|
25
|
Levels and actions of neuroactive steroids in the nervous system under physiological and pathological conditions: Sex-specific features. Neurosci Biobehav Rev 2016; 67:25-40. [DOI: 10.1016/j.neubiorev.2015.09.023] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 09/15/2015] [Accepted: 09/16/2015] [Indexed: 01/21/2023]
|
26
|
Sorge RE, Totsch SK. Sex Differences in Pain. J Neurosci Res 2016; 95:1271-1281. [PMID: 27452349 DOI: 10.1002/jnr.23841] [Citation(s) in RCA: 261] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 05/23/2016] [Accepted: 06/29/2016] [Indexed: 12/17/2022]
Abstract
Females greatly outnumber males as sufferers of chronic pain. Although social and psychological factors certainly play a role in the differences in prevalence and incidence, biological differences in the functioning of the immune system likely underlie these observed effects. This Review examines the current literature on biological sex differences in the functioning of the innate and adaptive immune systems as they relate to pain experience. With rodent models, we and others have observed that male mice utilize microglia in the spinal cord to mediate pain, whereas females preferentially use T cells in a similar manner. The difference can be traced to differences in cell populations, differences in suppression by hormones, and disparate cellular responses in males and females. These sex differences also translate into human cellular responses and may be the mechanism by which the disproportionate chronic pain experience is based. Recognition of the evidence underlying sex differences in pain will guide development of treatments and provide better options for patients that are tailored to their physiology. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Robert E Sorge
- Department of Psychology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Stacie K Totsch
- Department of Psychology, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
27
|
van den Berg R, Laman JD, van Meurs M, Hintzen RQ, Hoogenraad CC. Rotarod motor performance and advanced spinal cord lesion image analysis refine assessment of neurodegeneration in experimental autoimmune encephalomyelitis. J Neurosci Methods 2016; 262:66-76. [PMID: 26784021 DOI: 10.1016/j.jneumeth.2016.01.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Revised: 01/07/2016] [Accepted: 01/07/2016] [Indexed: 12/28/2022]
Abstract
BACKGROUND Experimental autoimmune encephalomyelitis (EAE) is a commonly used experimental model for multiple sclerosis (MS). Experience with this model mainly comes from the field of immunology, while data on its use in studying the neurodegenerative aspects of MS is scarce. NEW METHOD The aim of this study is to improve and refine methods to assess neurodegeneration and function in EAE. Using the rotarod, a tool used in neuroscience to monitor motor performance, we evaluated the correlation between motor performance, disease severity as measured using a clinical scale and area covered by inflammatory lesions. RESULTS The included parameters are highly correlated in a non-linear manner, with motor performance rapidly decreasing in the intermediate values of the clinical scale. The relation between motor performance and histopathological damage is exclusively determined by lesions in the ventral and lateral columns, based on a new method of analysis of the entire spinal cord. Using a set of definitions for distinct disease milestones, we quantified disease duration as well as severity. COMPARISON WITH EXISTING METHODS The rotarod measures motor performance in a more objective and quantitative manner compared to using a clinical score. The outcome shows a strong correlation to the surface area of inflammatory lesions in the motor systems of the spinal cord. CONCLUSIONS These results provide an improved workflow for interpreting the outcome of EAE from a neurological point of view, with the eventual goal of dissecting neurodegeneration and evaluating neuroprotective drugs in EAE for application in MS.
Collapse
Affiliation(s)
- Robert van den Berg
- Cell Biology, Utrecht University, Utrecht, The Netherlands; Department of Neurology, Erasmus MC, Rotterdam, The Netherlands
| | - Jon D Laman
- Department of Neuroscience, University Groningen, University Medical Center Groningen, The Netherlands
| | - Marjan van Meurs
- Department of Immunology, Erasmus MC, Rotterdam, The Netherlands
| | | | | |
Collapse
|
28
|
Sato F, Omura S, Jaffe S, Tsunoda I. Role of CD4+ T Cells in the Pathophysiology of Multiple Sclerosis. MULTIPLE SCLEROSIS 2016. [PMCID: PMC7150304 DOI: 10.1016/b978-0-12-800763-1.00004-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system. Although the precise etiology of MS remains unclear, CD4+ T cells have been proposed to play not only effector but also regulatory roles in MS. CD4+ T cells can be divided into four subsets: pro-inflammatory helper T (Th) 1 and Th17 cells, anti-inflammatory Th2 cells and regulatory T cells (Tregs). The roles of CD4+ T cells in MS have been clarified by either “loss-of-function” or “gain-of-function” methods, which have been carried out mainly in autoimmune and viral models of MS: experimental autoimmune encephalomyelitis and Theiler's murine encephalomyelitis virus infection, respectively. Observations in MS patients were consistent with the mechanisms found in the MS models, that is, increased pro-inflammatory Th1 and Th17 activity is associated with disease exacerbation, while anti-inflammatory Th2 cells and Tregs appear to play a protective role.
Collapse
|
29
|
The impact of gender on stroke pathology and treatment. Neurosci Biobehav Rev 2015; 67:119-24. [PMID: 26657813 DOI: 10.1016/j.neubiorev.2015.08.020] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 08/13/2015] [Accepted: 08/25/2015] [Indexed: 11/21/2022]
Abstract
Cerebral ischemic stroke is a leading cause of mortality and functional disability. However, unfortunately few effective treatments exist to counteract the deleterious pathological mechanisms triggered following an ischemic event. Epidemiological and experimental studies have revealed a significant difference in the vulnerability of males versus females to both the incidence of stroke and amount of resulting pathology following an ischemic stroke which is also dependent on the stage of lifespan. Here we review the evidence for gender differences in both the overall pathology and cellular mechanisms of injury following ischemic stroke. In addition, we discuss the evidence for any gender differences that may occur in the effectiveness of treatments and how this supports the need for the investigation and development of gender-specific therapies.
Collapse
|
30
|
Giatti S, Garcia-Segura LM, Melcangi RC. New steps forward in the neuroactive steroid field. J Steroid Biochem Mol Biol 2015; 153:127-34. [PMID: 25797031 DOI: 10.1016/j.jsbmb.2015.03.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Revised: 03/07/2015] [Accepted: 03/17/2015] [Indexed: 12/12/2022]
Abstract
Evidence accumulated in recent years suggests that the systemic treatment with neuroactive steroids, or the pharmacological modulation of its production by brain cells, represent therapeutic options to promote neuroprotection. However, new findings, which are reviewed in this paper, suggest that the factors to be considered for the design of possible therapies based on neuroactive steroids are more complex than previously thought. Thus, although as recently reported, the nervous system regulates neuroactive steroid synthesis and metabolism in adaptation to modifications in peripheral steroidogenesis, the neuroactive steroid levels in the brain do not fully reflect its levels in plasma. Even, in some cases, neuroactive steroid level modifications occurring in the nervous tissues, under physiological and pathological conditions, are in the opposite direction than in the periphery. This suggests that the systemic treatment with these molecules may have unexpected outcomes on neural steroid levels. In addition, the multiple metabolic pathways and signaling mechanisms of neuroactive steroids, which may change from one brain region to another, together with the existence of regional and sex differences in its neural levels are additional sources of complexity that should be clarified. This complexity in the levels and actions of these molecules may explain why in some cases these molecules have detrimental rather than beneficial actions for the nervous system. This article is part of a Special Issue entitled 'Steroid Perspectives'.
Collapse
Affiliation(s)
- Silvia Giatti
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | | | - Roberto Cosimo Melcangi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
31
|
Acaz-Fonseca E, Duran JC, Carrero P, Garcia-Segura LM, Arevalo MA. Sex differences in glia reactivity after cortical brain injury. Glia 2015; 63:1966-1981. [DOI: 10.1002/glia.22867] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 05/12/2015] [Accepted: 05/13/2015] [Indexed: 12/11/2022]
Affiliation(s)
| | - Juan C. Duran
- Consejo Superior De Investigaciones Cientificas (CSIC); Instituto Cajal; Madrid Spain
| | - Paloma Carrero
- Consejo Superior De Investigaciones Cientificas (CSIC); Instituto Cajal; Madrid Spain
| | - Luis M. Garcia-Segura
- Consejo Superior De Investigaciones Cientificas (CSIC); Instituto Cajal; Madrid Spain
| | - M. Angeles Arevalo
- Consejo Superior De Investigaciones Cientificas (CSIC); Instituto Cajal; Madrid Spain
| |
Collapse
|
32
|
Kalakh S, Mouihate A. The promyelinating properties of androstenediol in gliotoxin-induced demyelination in rat corpus callosum. Neuropathol Appl Neurobiol 2015; 41:964-82. [DOI: 10.1111/nan.12237] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2014] [Accepted: 03/06/2015] [Indexed: 12/12/2022]
Affiliation(s)
- Samah Kalakh
- Department of Physiology, Faculty of Medicine; Kuwait University; Safat Kuwait
| | - Abdeslam Mouihate
- Department of Physiology, Faculty of Medicine; Kuwait University; Safat Kuwait
| |
Collapse
|
33
|
Bastani F, Sobhani M, Emamzadeh Ghasemi HS. Effect of acupressure on fatigue in women with multiple sclerosis. Glob J Health Sci 2015; 7:375-81. [PMID: 25946938 PMCID: PMC4802202 DOI: 10.5539/gjhs.v7n4p375] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 11/23/2014] [Indexed: 11/25/2022] Open
Abstract
Introduction: Multiple sclerosis (MS) is the most common cause of progressive neurological disability. The prevalence of MS is much more common in women than men. The women are exposed to a variety of symptoms including fatigue. Acupressure is a noninvasive procedure that can be used to control symptoms including fatigue. The aim of the study was to evaluate the effect of acupressure on fatigue in women with multiple sclerosis. Methods: A randomized clinical trial was conducted on 100 women with MS at Tehran MS Association. The subjects were equally allocated to experimental group and a placebo group (50 women per group) by blocking randomization method. The experimental group were received acupressure, at the true points (ST36, SP6, LI4) and the placebo group, were received touching at the same points. Fatigue was measured by a Fatigue Severity Scale (FSS) in the groups at immediately prior to, two and four weeks after the beginning of the intervention. The data was analyzed using descriptive and inferential statistics by SPSS version 17. Results: The findings indicated no differences in demographic characteristics and the severity of fatigue at the baseline in two groups (p=0.54). But there were significant reductions of the mean score of fatigue in the experimental group compared to the placebo group immediately, two and four weeks after the intervention respectively (p=0.03, p≤0/001, p=0.04). Conclusion: According to the findings, the study provided an alternative method for health care providers including nurses to train acupressure to the clients with MS to managing their fatigue.
Collapse
|
34
|
Li R, Singh M. Sex differences in cognitive impairment and Alzheimer's disease. Front Neuroendocrinol 2014; 35:385-403. [PMID: 24434111 PMCID: PMC4087048 DOI: 10.1016/j.yfrne.2014.01.002] [Citation(s) in RCA: 363] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Revised: 12/31/2013] [Accepted: 01/06/2014] [Indexed: 12/15/2022]
Abstract
Studies have shown differences in specific cognitive ability domains and risk of Alzheimer's disease between the men and women at later age. However it is important to know that sex differences in cognitive function during adulthood may have their basis in both organizational effects, i.e., occurring as early as during the neuronal development period, as well as in activational effects, where the influence of the sex steroids influence brain function in adulthood. Further, the rate of cognitive decline with aging is also different between the sexes. Understanding the biology of sex differences in cognitive function will not only provide insight into Alzheimer's disease prevention, but also is integral to the development of personalized, gender-specific medicine. This review draws on epidemiological, translational, clinical, and basic science studies to assess the impact of sex differences in cognitive function from young to old, and examines the effects of sex hormone treatments on Alzheimer's disease in men and women.
Collapse
Affiliation(s)
- Rena Li
- Center for Hormone Advanced Science and Education (CHASE), Roskamp Institute, Sarasota, FL 34243, United States.
| | - Meharvan Singh
- Department of Pharmacology and Neuroscience, Institute for Aging and Alzheimer's Disease Research (IAADR), Center FOR HER, University of North Texas, Health Science Center, Fort Worth, TX 76107, United States
| |
Collapse
|
35
|
Iannitti T, Kerr BJ, Taylor BK. Mechanisms and pharmacology of neuropathic pain in multiple sclerosis. Curr Top Behav Neurosci 2014; 20:75-97. [PMID: 24590824 PMCID: PMC4464806 DOI: 10.1007/7854_2014_288] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The neuropathic pain of multiple sclerosis is quite prevalent and severely impacts quality of life. A few randomized, placebo-controlled, blinded clinical trials suggest that cannabis- and anticonvulsant-based treatments provide partial pain relief, but at the expense of adverse events. An even smaller, but emerging, number of translational studies are using rodent models of experimental autoimmune encephalomyelitis (EAE), which exhibit pain-like behaviors resembling those of Multiple sclerosis (MS) patients. These studies not only support the possible effectiveness of anticonvulsants, but also compel further clinical trials with serotonin-norepinephrine reuptake inhibitors, the immunosuppressant drug rapamycin, or drugs which interfere with glutamatergic neurotransmission. Future behavioral studies in EAE models are essential toward a new pharmacotherapy of multiple sclerosis pain.
Collapse
Affiliation(s)
- T Iannitti
- Department of Physiology, School of Medicine, University of Kentucky Medical Center, Lexington, KY, 40536-0298, USA
| | | | | |
Collapse
|
36
|
Melcangi RC, Giatti S, Calabrese D, Pesaresi M, Cermenati G, Mitro N, Viviani B, Garcia-Segura LM, Caruso D. Levels and actions of progesterone and its metabolites in the nervous system during physiological and pathological conditions. Prog Neurobiol 2014; 113:56-69. [DOI: 10.1016/j.pneurobio.2013.07.006] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2013] [Revised: 07/17/2013] [Accepted: 07/31/2013] [Indexed: 12/12/2022]
|
37
|
|
38
|
Naegele M, Martin R. The good and the bad of neuroinflammation in multiple sclerosis. HANDBOOK OF CLINICAL NEUROLOGY 2014; 122:59-87. [PMID: 24507513 DOI: 10.1016/b978-0-444-52001-2.00003-0] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Multiple sclerosis (MS) is the most common inflammatory, demyelinating, neurodegenerative disorder of the central nervous system (CNS). It is widely considered a T-cell mediated autoimmune disease that develops in genetically susceptible individuals, possibly under the influence of certain environmental trigger factors. The invasion of autoreactive CD4+ T-cells into the CNS is thought to be a central step that initiates the disease. Several other cell types, including CD8+ T-cells, B-cells and phagocytes appear to be involved in causing inflammation and eventually neurodegeneration. But inflammation is not entirely deleterious in MS. Evidence has accumulated in the recent years that show the importance of regulatory immune mechanisms which restrain tissue damage and initiate regeneration. More insight into the beneficial aspects of neuroinflammation might allow us to develop new treatment strategies for this enigmatic disease.
Collapse
Affiliation(s)
- Matthias Naegele
- Institute for Neuroimmunology and Clinical Multiple Sclerosis Research, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Roland Martin
- Neuroimmunology and MS Research, Neurology Clinic, University Hospital, Zurich, Switzerland.
| |
Collapse
|
39
|
Tatar C, Bessert D, Tse H, Skoff RP. Determinants of central nervous system adult neurogenesis are sex, hormones, mouse strain, age, and brain region. Glia 2012; 61:192-209. [PMID: 23027402 DOI: 10.1002/glia.22426] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Accepted: 08/30/2012] [Indexed: 11/11/2022]
Abstract
Multiple sclerosis is a sexually dimorphic (SD) disease that causes oligodendrocyte death, but SD of glial cells is poorly studied. Here, we analyze SD of neural progenitors in 6-8 weeks and 6-8 months normal C57BL/6, SJL/J, and BALB/c mice in the subventricular zone (SVZ), dorsolateral horn (DLC), corpus callosum (CC), and parenchyma. With a short 2-h bromodeoxyuridine (BrdU) pulse, no gender and strain differences are present at 6-8 weeks. At 6-8 months, the number of BrdU(+) cells decreases twofold in each sex, strain, and region, indicating that a common aging mechanism regulates BrdU incorporation. Strikingly, 2× more BrdU(+) cells are found in all brain regions in 6-8 months C57BL/6 females versus males, no gender differences in 6-8 months SJL/J, and fewer BrdU(+) cells in females versus males in BALB/cs. The number of BrdU(+) cells modestly fluctuates throughout the estrous cycle in C57BL/6 and SJLs. Castration causes a dramatic increase in BrdU(+) cells in SVZ and DLC. These findings indicate that testosterone is a major regulator of adult neural proliferation. At 6-8 months, the ratio of PDGFRα(+) cells in the CC to BrdU(+) cells in the DLC of both strains, sexes, estrous cycle, and castrated mice was essentially the same, suggesting that BrdU(+) cells in the DLC differentiate into CC oligodendrocytes. The ratio of TUNEL(+) to BrdU(+) cells does not match proliferation, indicating that these events are differentially regulated. Differential regulation of these two processes leads to the variation in glial numbers between gender and strain. Explanations of neural proliferation based upon data from one sex or strain may be very misleading.
Collapse
Affiliation(s)
- Carrie Tatar
- Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, Michigan 48201, USA
| | | | | | | |
Collapse
|
40
|
Multimodal Analysis in Acute and Chronic Experimental Autoimmune Encephalomyelitis. J Neuroimmune Pharmacol 2012; 8:238-50. [DOI: 10.1007/s11481-012-9385-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Accepted: 06/19/2012] [Indexed: 01/01/2023]
|
41
|
Giatti S, Caruso D, Boraso M, Abbiati F, Ballarini E, Calabrese D, Pesaresi M, Rigolio R, Santos-Galindo M, Viviani B, Cavaletti G, Garcia-Segura LM, Melcangi RC. Neuroprotective effects of progesterone in chronic experimental autoimmune encephalomyelitis. J Neuroendocrinol 2012; 24:851-61. [PMID: 22283602 DOI: 10.1111/j.1365-2826.2012.02284.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Observations so far obtained in experimental autoimmune encephalomyelitis (EAE) have revealed the promising neuroprotective effects exerted by progesterone (PROG). The findings suggest that this neuroactive steroid may potentially represent a therapeutic tool for multiple sclerosis (MS). However, up to now, the efficacy of PROG has been only tested in the acute phase of the disease, whereas it is well known that MS expresses different features depending on the phase of the disease. Accordingly, we have evaluated the effect of PROG treatment in EAE induced in Dark Agouti rats (i.e. an experimental model showing a protracted relapsing EAE). Data obtained 45 days after EAE induction show that PROG treatment exerts a beneficial effect on clinical score, confirming surrogate parameters of spinal cord damage in chronic EAE (i.e. reactive microglia, cytokine levels, activity of the Na(+) ,K(+) -ATPase pump and myelin basic protein expression). An increase of the levels of dihydroprogesterone and isopregnanolone (i.e. two PROG metabolites) was also observed in the spinal cord after PROG treatment. Taken together, these results indicate that PROG is effective in reducing the severity of chronic EAE and, consequently, may have potential with respect to MS treatment.
Collapse
Affiliation(s)
- S Giatti
- Department of Endocrinology, Pathophysiology and Applied Biology, Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Milano, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Massella A, D'Intino G, Fernández M, Sivilia S, Lorenzini L, Giatti S, Melcangi RC, Calzà L, Giardino L. Gender effect on neurodegeneration and myelin markers in an animal model for multiple sclerosis. BMC Neurosci 2012; 13:12. [PMID: 22272832 PMCID: PMC3282645 DOI: 10.1186/1471-2202-13-12] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Accepted: 01/24/2012] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Multiple sclerosis (MS) varies considerably in its incidence and progression in females and males. In spite of clinical evidence, relatively few studies have explored molecular mechanisms possibly involved in gender-related differences. The present study describes possible cellular- and molecular-involved markers which are differentially regulated in male and female rats and result in gender-dependent EAE evolution and progression. Attention was focused on markers of myelination (MBP and PDGFαR) and neuronal distress and/or damage (GABA synthesis enzymes, GAD65 and GAD67, NGF, BDNF and related receptors), in two CNS areas, i.e. spinal cord and cerebellum, which are respectively severely and mildly affected by inflammation and demyelination. Tissues were sampled during acute, relapse/remission and chronic phases and results were analysed by two-way ANOVA. RESULTS 1. A strong gender-dependent difference in myelin (MBP) and myelin precursor (PDGFαR) marker mRNA expression levels is observed in control animals in the spinal cord, but not in the cerebellum. This is the only gender-dependent difference in the expression level of the indicated markers in healthy animals; 2. both PDGFαR and MBP mRNAs in the spinal cord and MBP in the cerebellum are down-regulated during EAE in gender-dependent manner; 3. in the cerebellum, the expression profile of neuron-associated markers (GAD65, GAD67) is characterized by a substantial down-regulation during the inflammatory phase of the disease, which does not differ between male and female rats (two-way ANOVA); 4. there is an up-regulation of NGF, trkA and p75 mRNA expression in the early phases of the disease (14 and 21 days post-immunization), which is not different between male and female. CONCLUSIONS It is reported herein that the regulation of markers involved in demyelination and neuroprotection processes occurring during EAE, a well-established MS animal model, is gender- and time-dependent. These findings might contribute to gender- and phase disease-based therapy strategies.
Collapse
Affiliation(s)
- Alessandro Massella
- Department of Veterinary Medicine, University of Bologna, Via Tolara di Sopra 50, 40064 Ozzano Emilia, Italy
| | - Giulia D'Intino
- Department of Veterinary Medicine, University of Bologna, Via Tolara di Sopra 50, 40064 Ozzano Emilia, Italy
| | - Mercedes Fernández
- Health Sciences and Technology - Interdepartmental Center for Industrial Research (HST-ICIR), University of Bologna, Via Tolara di Sopra 50, 40064 Ozzano Emilia, Italy
| | - Sandra Sivilia
- Department of Veterinary Medicine, University of Bologna, Via Tolara di Sopra 50, 40064 Ozzano Emilia, Italy
| | - Luca Lorenzini
- Department of Veterinary Medicine, University of Bologna, Via Tolara di Sopra 50, 40064 Ozzano Emilia, Italy
| | - Silvia Giatti
- Dept. of Endocrinology, Pathophysiology and Applied Biology - Center of Excellence on Neurodegenerative Diseases, University of Milan, Via Balzaretti 9, 20133 Milano, Italy
| | - Roberto C Melcangi
- Dept. of Endocrinology, Pathophysiology and Applied Biology - Center of Excellence on Neurodegenerative Diseases, University of Milan, Via Balzaretti 9, 20133 Milano, Italy
| | - Laura Calzà
- Health Sciences and Technology - Interdepartmental Center for Industrial Research (HST-ICIR), University of Bologna, Via Tolara di Sopra 50, 40064 Ozzano Emilia, Italy
- Department of Veterinary Medicine, University of Bologna, Via Tolara di Sopra 50, 40064 Ozzano Emilia, Italy
| | - Luciana Giardino
- Health Sciences and Technology - Interdepartmental Center for Industrial Research (HST-ICIR), University of Bologna, Via Tolara di Sopra 50, 40064 Ozzano Emilia, Italy
- Department of Veterinary Medicine, University of Bologna, Via Tolara di Sopra 50, 40064 Ozzano Emilia, Italy
| |
Collapse
|
43
|
Irizar H, Muñoz-Culla M, Zuriarrain O, Goyenechea E, Castillo-Triviño T, Prada A, Saenz-Cuesta M, De Juan D, Lopez de Munain A, Olascoaga J, Otaegui D. HLA-DRB1*15:01 and multiple sclerosis: a female association? Mult Scler 2011; 18:569-77. [PMID: 22127897 DOI: 10.1177/1352458511426813] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
BACKGROUND The association between multiple sclerosis (MS) and the HLA-DRB1*15:01 haplotype has been proven to be strong, but its molecular basis remains unclear. Vitamin D receptor (VDR) gene variants and sex have been proposed to modulate this association. OBJECTIVES 1) Test the association of MS with *15:01 and VDR variants; 2) check whether VDR variants and/or sex modulate the risk conferred by *15:01; 3) study whether *15:01, VDR variants and/or sex affect HLA II gene expression. METHODS Peripheral blood from 364 MS patients and 513 healthy controls was obtained and DNA and total RNA were extracted from leukocytes. HLA-DRB1, DRB5 and DQA1 gene expression measurements and *15:01 genotyping were performed by qPCR. VDR variants were genotyped by PCR-RFLP. RESULTS Our data confirms that the *15:01 haplotype confers a higher risk of suffering from MS (OR = 1.364; 95% CI = 1.107-1.681). No association was found between VDR variants and MS, but they were shown to moderately modulate the risk conferred by *15:01. Sex confers a much stronger modulation and the *15:01-MS association seems to be female specific. A higher *15:01 frequency has been observed in Basques (45.1%). *15:01 positive samples showed a significant overexpression of DRB1 (p < 0.001), DRB5 (p < 0.001) and DQA1 (p = 0.004) in patients. DRB1 (p = 0.004) and DRB5 (p < 0.001) were also overexpressed in *15:01 controls. CONCLUSIONS We confirm the *15:01-MS association and support that it is female specific. The relevance of ethnic origin on association studies has also been highlighted. HLA-DRB1*15:01 seems to be a haplotype consistently linked to high HLA II gene expression.
Collapse
Affiliation(s)
- Haritz Irizar
- Multiple Sclerosis Unit, Neuroscience Area, Biodonostia Health Research Institute, Donostia-San Sebastian, Spain.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Sato F, Tanaka H, Hasanovic F, Tsunoda I. Theiler's virus infection: Pathophysiology of demyelination and neurodegeneration. ACTA ACUST UNITED AC 2011; 18:31-41. [PMID: 20537875 DOI: 10.1016/j.pathophys.2010.04.011] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2009] [Revised: 02/28/2010] [Accepted: 04/08/2010] [Indexed: 02/08/2023]
Abstract
Multiple sclerosis (MS) has been suggested to be an autoimmune demyelinating disease of the central nervous system (CNS), whose primary target is either myelin itself, or myelin-forming cells, the oligodendrocytes. Although axonal damage occurs in MS, it is regarded as a secondary event to the myelin damage. Here, the lesion develops from the myelin (outside) to the axons (inside) "Outside-In model". The Outside-In model has been supported by an autoimmune model for MS, experimental autoimmune (allergic) encephalomyelitis (EAE). However, recently, (1) EAE-like disease has also been shown to be induced by immune responses against axons, and (2) immune responses against axons and neurons as well as neurodegeneration independent of inflammatory demyelination have been reported in MS, which can not be explained by the Outside-In model. Theiler's murine encephalomyelitis virus (TMEV)-induced demyelinating disease (TMEV-IDD) is a viral model for MS. In TMEV infection, axonal injury precedes demyelination, where the lesion develops from the axons (inside) to the myelin (outside) "Inside-Out model". The initial axonal damage could result in the release of neuroantigens, inducing autoimmune responses against myelin antigens, which potentially attack the myelin from outside the nerve fiber. Thus, the Inside-Out and Outside-In models can make a "vicious" immunological cycle or initiate an immune cascade.
Collapse
Affiliation(s)
- Fumitaka Sato
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, School of Medicine in Shreveport, Shreveport, LA 71130, USA
| | | | | | | |
Collapse
|
45
|
Luchetti S, Huitinga I, Swaab DF. Neurosteroid and GABA-A receptor alterations in Alzheimer's disease, Parkinson's disease and multiple sclerosis. Neuroscience 2011; 191:6-21. [PMID: 21514366 DOI: 10.1016/j.neuroscience.2011.04.010] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2010] [Revised: 04/03/2011] [Accepted: 04/05/2011] [Indexed: 01/17/2023]
Abstract
Steroid hormones (e.g. estrogens, androgens, progestagens) which are synthesized de novo or metabolized within the CNS are called neurosteroids. There is substantial evidence from animal studies suggesting that these steroids can affect brain function by modulating neurotransmission, and influence neuronal survival, neuronal and glial differentiation and myelination in the CNS by regulating gene expression of neurotrophic factors and anti-inflammatory molecules. Indeed, evidence is emerging that expression of the enzymes responsible for the synthesis of neurosteroids changes in neurodegenerative diseases. Some of these changes may contribute to the pathology, while others, conversely, may represent an attempted rescue program in the diseased brain. Here we review the data on changes in neurosteroid levels and neurosteroid synthesis pathways in the human brain in three neurodegenerative conditions, Alzheimers's (AD) and Parkinson's (PD) diseases and Multiple Sclerosis (MS) and the extent to which these findings may implicate protective or pathological roles for neurosteroids in the course of these diseases.Some neurosteroids can modulate neurotransmitter activity, for example, the pregnane steroids allopregnanolone and 3α5α-tetrahydro-deoxycorticosterone which are potent positive allosteric modulators of ionotropic GABA-A receptors. Therefore, neurosteroid-modulated GABA-A receptor subunit alterations found in AD and PD will also be discussed. These data imply an involvement of neurosteroid changes in the neurodegenerative and neuroinflammatory processes and suggest that they may deserve further investigation as potential therapeutic agents in AD, PD and MS. Finally, suggestions for therapeutic strategies will be included. This article is part of a Special Issue entitled: Neuroactive Steroids: Focus on Human Brain.
Collapse
Affiliation(s)
- S Luchetti
- Netherlands Institute for Neuroscience (NIN), an Institute of the Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA, Amsterdam, The Netherlands.
| | | | | |
Collapse
|
46
|
Jobin C, Larochelle C, Parpal H, Coyle PK, Duquette P. Gender issues in multiple sclerosis: an update. ACTA ACUST UNITED AC 2011; 6:797-820. [PMID: 21118039 DOI: 10.2217/whe.10.69] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Although multiple sclerosis (MS) affects both women and men, women are more susceptible to MS than men. Accumulating evidence indicates that the incidence and prevalence of MS is increasing, more so in women than in men. Owing to pregnancy, differing hormonal states and distinct social roles, the impact of MS differs between women and men. Since Patricia K Coyle published a review on gender issues in MS, multiple studies have added to the body of knowledge. This update will summarize the current thinking on gender-related issues in MS and we will address incidence and prevalence, hormonal factors, pregnancy and breastfeeding, genetics, course and prognosis, imaging, treatment and psychosocial aspects. Future progression within this field will help elucidate the cause of and define the treatment of MS.
Collapse
|
47
|
Melcangi RC, Garcia-Segura LM. Sex differences in the injured brain. Horm Mol Biol Clin Investig 2011; 7:385-91. [DOI: 10.1515/hmbci.2011.112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2011] [Accepted: 08/15/2011] [Indexed: 11/15/2022]
Abstract
AbstractObservations obtained in human and in experimental models clearly demonstrate sex differences in degenerative events occurring in the central nervous system. The present review focuses on potential factors that may contribute to these sex-dimorphic features; in particular, morphological organization of the central nervous system and functional influence by neuroactive steroids, genes, and immune system are considered.
Collapse
|
48
|
Majde JA. Neuroinflammation resulting from covert brain invasion by common viruses - a potential role in local and global neurodegeneration. Med Hypotheses 2010; 75:204-13. [PMID: 20236772 PMCID: PMC2897933 DOI: 10.1016/j.mehy.2010.02.023] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2010] [Accepted: 02/21/2010] [Indexed: 11/22/2022]
Abstract
Neurodegenerative diseases are a horrendous burden for their victims, their families, and society as a whole. For half a century scientists have pursued the hypothesis that these diseases involve a chronic viral infection in the brain. However, efforts to consistently detect a specific virus in brains of patients with such diseases as Alzheimer's or multiple sclerosis have generally failed. Neuropathologists have become increasingly aware that most patients with neurodegenerative diseases demonstrate marked deterioration of the brain olfactory bulb in addition to brain targets that define the specific disease. In fact, the loss of the sense of smell may precede overt neurological symptoms by many years. This realization that the olfactory bulb is a common target in neurodegenerative diseases suggests the possibility that microbes and/or toxins in inhaled air may play a role in their pathogenesis. With regard to inhaled viruses, neuropathologists have focused on those viruses that infect and kill neurons. However, a recent study shows that a respiratory virus with no neurotropic properties can rapidly invade the mouse olfactory bulb from the nasal cavity. Available data suggest that this strain of influenza is passively transported to the bulb via the olfactory nerves (mechanism unknown), and is taken up by glial cells in the outer layers of the bulb. The infected glial cells appear to be activated by the virus, secrete proinflammatory cytokines, and block further spread of virus within the brain. At the time that influenza symptoms become apparent (15 h post-infection), but not prior to symptom onset (10 h post-infection), proinflammatory cytokine-expressing neurons are increased in olfactory cortical pathways and hypothalamus as well as in the olfactory bulb. The mice go on to die of pneumonitis with severe acute phase and respiratory disease symptoms but no classical neurological symptoms. While much remains to be learned about this intranasal influenza-brain invasion model, it suggests the hypothesis that common viruses encountered in our daily life may initiate neuroinflammation via olfactory neural networks. The numerous viruses that we inhale during a lifetime might cause the death of only a few neurons per infection, but this minor damage would accumulate over time and contribute to age-related brain shrinkage and/or neurodegenerative diseases. Elderly individuals with a strong innate inflammatory system, or ongoing systemic inflammation (or both), might be most susceptible to these outcomes. The evidence for the hypothesis that common respiratory viruses may contribute to neurodegenerative processes is developed in the accompanying article.
Collapse
Affiliation(s)
- Jeannine A Majde
- Department of VCAPP, College of Veterinary Medicine, Washington State University, Pullman, WA 99164-6520, USA.
| |
Collapse
|
49
|
Caruso D, D’Intino G, Giatti S, Maschi O, Pesaresi M, Calabrese D, Garcia-Segura LM, Calza L, Melcangi RC. Sex-dimorphic changes in neuroactive steroid levels after chronic experimental autoimmune encephalomyelitis. J Neurochem 2010; 114:921-32. [DOI: 10.1111/j.1471-4159.2010.06825.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
50
|
Cosimo Melcangi R, Garcia-Segura LM. Sex-specific therapeutic strategies based on neuroactive steroids: In search for innovative tools for neuroprotection. Horm Behav 2010; 57:2-11. [PMID: 19524584 DOI: 10.1016/j.yhbeh.2009.06.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2009] [Revised: 06/02/2009] [Accepted: 06/04/2009] [Indexed: 02/07/2023]
Abstract
Different pathologies of the central and peripheral nervous system show sex differences in their incidence, symptomatology and/or neurodegenerative outcome. These include Parkinson's disease, Alzheimer's disease, Huntington's disease, multiple sclerosis, traumatic brain injury, stroke, autism, schizophrenia, depression, anxiety disorders, eating disorders and peripheral neuropathy. These sex differences reveal the need for sex-specific neuroprotective strategies. This review article and other manuscripts published in this issue of Hormones and Behavior analyze possible sex-specific therapeutic strategies based on neuroactive steroids. In particular in our introductory article, the possibility that sex differences in the levels or in the action of neuroactive steroids may represent causative factors for sex differences in the incidence or manifestation of pathologies of the nervous system is considered.
Collapse
Affiliation(s)
- Roberto Cosimo Melcangi
- Department of Endocrinology, Pathophysiology and Applied Biology, Center of Excellence on Neurodegenerative Diseases, University of Milan, Milano, Italy.
| | | |
Collapse
|