51
|
Rafiq M, Hayward A, Warren-Gash C, Denaxas S, Gonzalez-Izquierdo A, Lyratzopoulos G, Thomas S. Socioeconomic deprivation and regional variation in Hodgkin's lymphoma incidence in the UK: a population-based cohort study of 10 million individuals. BMJ Open 2019; 9:e029228. [PMID: 31542744 PMCID: PMC6756616 DOI: 10.1136/bmjopen-2019-029228] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 04/18/2019] [Accepted: 07/12/2019] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVES Hodgkin's lymphoma (HL) is the the most common cancer in teenagers and young adults. This nationwide study conducted over a 25-year period in the UK investigates variation in HL incidence by age, sex, region and deprivation to identify trends and high-risk populations for HL development. DESIGN Population-based cohort study. SETTING Clinical Practice Research Datalink (CPRD) electronic primary care records linked to Hospital Episode Statistics and Index of Multiple Deprivation data were used. PARTICIPANTS Data on 10 million individuals in the UK from 1992 to 2016 were analysed. PRIMARY AND SECONDARY OUTCOME MEASURES Poisson models were used to explore differences in HL incidence by age, sex, region and deprivation. Age-specific HL incidence rates by sex and directly age-standardised incidence rates by region and deprivation group were calculated. RESULTS A total of 2402 new cases of HL were identified over 78 569 436 person-years. There was significant variation in HL incidence by deprivation group. Individuals living in the most affluent areas had HL incidence 60% higher than those living in the most deprived (incidence rate ratios (IRR) 1.60, 95% CI 1.40 to 1.83), with strong evidence of a marked linear trend towards increasing HL incidence with decreasing deprivation (p=<0.001). There was significant regional variation in HL incidence across the UK, which persisted after adjusting for age, sex and deprivation (IRR 0.80-1.42, p=<0.001). CONCLUSIONS This study identified high-risk regions for HL development in the UK and observed a trend towards higher incidence of HL in individuals living in less deprived areas. Consistent with findings from other immune-mediated diseases, this study supports the hypothesis that an affluent childhood environment may predispose to development of immune-related neoplasms, potentially through fewer immune challenges interfering with immune maturation in early life. Understanding the mechanisms behind this immune dysfunction could inform prevention, detection and treatment of HL and other immune diseases.
Collapse
Affiliation(s)
- Meena Rafiq
- Institute of Health Informatics, University College London, London, UK
| | - Andrew Hayward
- Institute of Epidemiology and Health Care, University College London, London, UK
| | - Charlotte Warren-Gash
- Non-communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
| | - S Denaxas
- Institute of Health Informatics, University College London, London, UK
| | | | - Georgios Lyratzopoulos
- Department of Behavioural Science and Health, ECHO (Epidemiology of Cancer Healthcare & Outcomes) Research Group, University College London, London, UK
| | - Sara Thomas
- Non-communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
| |
Collapse
|
52
|
ImmGen report: sexual dimorphism in the immune system transcriptome. Nat Commun 2019; 10:4295. [PMID: 31541153 PMCID: PMC6754408 DOI: 10.1038/s41467-019-12348-6] [Citation(s) in RCA: 145] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 08/29/2019] [Indexed: 11/24/2022] Open
Abstract
Sexual dimorphism in the mammalian immune system is manifested as more frequent and severe infectious diseases in males and, on the other hand, higher rates of autoimmune disease in females, yet insights underlying those differences are still lacking. Here we characterize sex differences in the immune system by RNA and ATAC sequence profiling of untreated and interferon-induced immune cell types in male and female mice. We detect very few differentially expressed genes between male and female immune cells except in macrophages from three different tissues. Accordingly, very few genomic regions display differences in accessibility between sexes. Transcriptional sexual dimorphism in macrophages is mediated by genes of innate immune pathways, and increases after interferon stimulation. Thus, the stronger immune response of females may be due to more activated innate immune pathways prior to pathogen invasion. Sexual dimorphism is observed frequently in immune disorders, but the underlying insights are still unclear. Here the authors analyze transcriptome and epigenome changes induced by interferon in various mouse immune cell types, and find only a restricted set of sexual dimorphism genes in innate immunity and macrophages.
Collapse
|
53
|
Wu FL, Lai DY, Ding HH, Tang YJ, Xu ZW, Ma ML, Guo SJ, Wang JF, Shen N, Zhao XD, Qi H, Li H, Tao SC. Identification of Serum Biomarkers for Systemic Lupus Erythematosus Using a Library of Phage Displayed Random Peptides and Deep Sequencing. Mol Cell Proteomics 2019; 18:1851-1863. [PMID: 31308251 PMCID: PMC6731078 DOI: 10.1074/mcp.ra119.001582] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 07/01/2019] [Indexed: 12/26/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is one of the most serious autoimmune diseases, characterized by highly diverse clinical manifestations. A biomarker is still needed for accurate diagnostics. SLE serum autoantibodies were discovered and validated using serum samples from independent sample cohorts encompassing 306 participants divided into three groups, i.e. healthy, SLE patients, and other autoimmune-related diseases. To discover biomarkers for SLE, a phage displayed random peptide library (Ph.D. 12) and deep sequencing were applied to screen specific autoantibodies in a total of 100 serum samples from 50 SLE patients and 50 healthy controls. A statistical analysis protocol was set up for the identification of peptides as potential biomarkers. For validation, 10 peptides were analyzed using enzyme-linked immunosorbent assays (ELISA). As a result, four peptides (SLE2018Val001, SLE2018Val002, SLE2018Val006, and SLE2018Val008) were discovered with high diagnostic power to differentiate SLE patients from healthy controls. Among them, two peptides, i.e. SLE2018Val001 and SLE2018Val002, were confirmed between SLE with other autoimmune patients. The procedure we established could be easily adopted for the identification of autoantibodies as biomarkers for many other diseases.
Collapse
Affiliation(s)
- Fan-Lin Wu
- ‡Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China; ¶School of Agriculture, Ludong University, Yantai 264025, China
| | - Dan-Yun Lai
- ‡Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Hui-Hua Ding
- ‖Shanghai Institute of Rheumatology, Department of rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 145 Shan Dong Road (c), Shanghai 200240, China
| | - Yuan-Jia Tang
- ‖Shanghai Institute of Rheumatology, Department of rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 145 Shan Dong Road (c), Shanghai 200240, China
| | - Zhao-Wei Xu
- ‡Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Ming-Liang Ma
- ‡Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Shu-Juan Guo
- ‡Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Jing-Fang Wang
- ‡Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Nan Shen
- ‖Shanghai Institute of Rheumatology, Department of rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 145 Shan Dong Road (c), Shanghai 200240, China; **State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, 2200 Lane 25 Xietu Road, Shanghai 200240, China
| | - Xiao-Dong Zhao
- ‡Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Huan Qi
- ‡Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China.
| | - Hua Li
- §Bio-ID Center, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Sheng-Ce Tao
- ‡Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China; §Bio-ID Center, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
54
|
Stumper A, Moriarity DP, Coe CL, Ellman LM, Abramson LY, Alloy LB. Pubertal Status and Age are Differentially Associated with Inflammatory Biomarkers in Female and Male Adolescents. J Youth Adolesc 2019; 49:1379-1392. [PMID: 31410721 DOI: 10.1007/s10964-019-01101-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 08/01/2019] [Indexed: 12/21/2022]
Abstract
A better understanding of the maturational correlates of inflammatory activity during adolescence is needed to more appropriately study both normal and abnormal development. Inflammation is the immune system's first response to infection, injury, or psychological stress, and it has been shown to be elevated in individuals with both physical and psychological conditions. This study examined unique associations between (1) pubertal status and inflammatory biomarkers, and (2) age and inflammatory biomarkers, and whether these relationships differed by sex in a diverse sample of 155 adolescents (54.2% female, 45.8% male; Mage = 16.22) from a northeastern city in the US. A more advanced pubertal status was uniquely associated with lower levels of tumor necrosis factor alpha (TNF-α) and interleukin-8 (IL-8). Chronological age was uniquely associated with lower IL-8 levels. The association between pubertal status and C-reactive protein (CRP) levels differed by sex: more mature females had higher CRP, whereas pubertal status and CRP were not significantly associated in males. These findings highlight an important relation between pubertal development and inflammatory activity during adolescence.
Collapse
|
55
|
Seeliger T, Prenzler NK, Gingele S, Seeliger B, Körner S, Thiele T, Bönig L, Sühs KW, Witte T, Stangel M, Skripuletz T. Neuro-Sjögren: Peripheral Neuropathy With Limb Weakness in Sjögren's Syndrome. Front Immunol 2019; 10:1600. [PMID: 31354737 PMCID: PMC6637792 DOI: 10.3389/fimmu.2019.01600] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 06/26/2019] [Indexed: 11/23/2022] Open
Abstract
Objective: Sjögren's syndrome is a heterogeneous inflammatory disorder frequently involving peripheral nerves with a wide spectrum of sensory modalities and distribution patterns. The objective of this cross-sectional study was to determine characteristics of Sjögren's syndrome as a cause for severe neuropathy with limb weakness. Methods: One hundred and eighty four patients with polyneuropathy associated with limb weakness underwent routine diagnostics including investigations for Sjögren's syndrome. Forty-four patients with Sjögren's syndrome (ACR-EULAR classification criteria) and severe neuropathy were identified. Results: Sjögren's syndrome was found at a median age of 63 years and the gender distribution showed a balanced female-male ratio of 1:1. Anti-SSA(Ro) antibodies were detected in 48% while seronegative patients were diagnosed with Sjögren's syndrome based on sialadenitis on minor salivary gland biopsy with a focus score ≥1. The majority of patients (93%) were diagnosed with Sjögren's syndrome after neurological symptoms appeared. Limbs were symmetrically involved in 84% of patients (57% tetraparesis, 27% paraparesis). Sensory function was not affected in 11% of patients indicating that Sjögren's syndrome associated neuropathy can present as a pure motor syndrome. Electrophysiological measurements did not reveal pathognomonic findings (23% demyelinating pattern, 36% axonal pattern, 41% both demyelinating and axonal damage signs). More than half of our patients fulfilled the European Federation of Neurological Societies (EFNS) diagnostic criteria for CIDP indicating that distinction between Neuro-Sjögren and other causes of neuropathy such as CIDP is challenging. Interpretation: Our findings show that severe neuropathy with limb weakness is often associated with Sjögren's syndrome. This is of great importance in identifying and understanding the causes of immune mediated polyneuropathy.
Collapse
Affiliation(s)
- Tabea Seeliger
- Department of Neurology, Hanover Medical School, Hanover, Germany
| | - Nils K. Prenzler
- Department of Otolaryngology, Hanover Medical School, Hanover, Germany
| | - Stefan Gingele
- Department of Neurology, Hanover Medical School, Hanover, Germany
| | - Benjamin Seeliger
- Department of Respiratory Medicine, Hanover Medical School, Hanover, Germany
| | - Sonja Körner
- Department of Neurology, Hanover Medical School, Hanover, Germany
| | - Thea Thiele
- Department of Clinical Immunology and Rheumatology, Hanover Medical School, Hanover, Germany
| | - Lena Bönig
- Department of Neurology, Hanover Medical School, Hanover, Germany
| | | | - Torsten Witte
- Department of Clinical Immunology and Rheumatology, Hanover Medical School, Hanover, Germany
| | - Martin Stangel
- Department of Neurology, Hanover Medical School, Hanover, Germany
| | | |
Collapse
|
56
|
Shepard BD. Sex differences in diabetes and kidney disease: mechanisms and consequences. Am J Physiol Renal Physiol 2019; 317:F456-F462. [PMID: 31241989 DOI: 10.1152/ajprenal.00249.2019] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Type 1 and type 2 diabetes, along with their accompanying hyperglycemia, are associated with a multitude of comorbidities including the development of diabetic kidney disease. Although the hallmarks of these metabolic disorders have been well characterized in population and animal studies, it is becoming increasingly apparent that diabetes manifests itself differently in men and women. This review summarizes the recent diabetic literature with a focus on known sex differences in clinical and preclinical studies. It explores the physiological differences of glucose handling and the development of diabetes between men and women. This review also uncovers potential mechanisms for these differences, honing in on the vital role that sex hormone signaling plays in the progression of diabetes and renal complications.
Collapse
Affiliation(s)
- Blythe D Shepard
- Department of Human Science, Georgetown University, Washington, District of Columbia
| |
Collapse
|
57
|
He X, Yang L, Liu X, Wei W, Shi C, Li B, Li J. Ginsenoside Rb1 Upregulating AQP5 Protein Expression and Alleviating Salivary Secretion Impairment in Ovariectomized Sjögren’s Syndrome Mice. Chem Res Chin Univ 2019. [DOI: 10.1007/s40242-019-9056-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
58
|
Sibanda E, Makaza N. Health effects of diesel engine exhaust emissions exposure (DEEE) can mimic allergic asthma and rhinitis. ALLERGY, ASTHMA, AND CLINICAL IMMUNOLOGY : OFFICIAL JOURNAL OF THE CANADIAN SOCIETY OF ALLERGY AND CLINICAL IMMUNOLOGY 2019; 15:31. [PMID: 31168306 PMCID: PMC6489272 DOI: 10.1186/s13223-019-0342-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 04/15/2019] [Indexed: 01/28/2023]
Abstract
BACKGROUND Patients presenting to Accident and Emergency (A&E) facilities with dyspnoea, coughing, wheezing and nasal blockage are presumed to have allergic asthma and/or rhinitis. Occupational asthma (OA), which has similar symptoms is rarely considered. Triggers of OA include exposure to diesel engine exhaust emissions exposure (DEEEE) that are carcinogenic. We report the case of a patient who presented to an A&E facility with asthma-like symptoms, was treated for allergic asthma. Frequent exacerbations were experienced. Upon investigations it was shown that were symptoms triggered by DEEE exposure. CASE PRESENTATION A 36-year-old female bank employee was referred for the evaluation of suspected asthma. She reported a 3-month history of symptoms suggestive of asthma and rhinitis, for which she had previously required A&E treatment. There was no history of atopy. The symptoms only occurred at work or after work. Their onset had coincided with changing offices to one located proximal to a diesel-powered electricity generator. A diagnosis of asthma had been made at the A&E facility and the appropriately used inhaled fluticasone and salbutamol provided limited relief. Skin prick testing was weakly positive for seasonal pollen and house dust mite allergens. Allergen specific IgE tests for 16 regionally relevant aeroallergens were negative. Tests to exclude connective tissue diseases were positive for the anti-Ro-52/TRIM-21 autoantibody. Baseline spirometry values were markedly reduced and bronchodilator administration showed limited reversibility, FEV1 (+ 8%), PEF (+ 5%). Following a 10-day discontinuation of work exposure, the symptoms abated and FEV1 and PEF increased by 10-14% from baseline. The recent onset of asthma, in a non-atopic adult, with workday related symptoms and improvement upon discontinuation of exposure were attributed to passive occupational exposure to DEEE. The diesel generator was relocated, a short course of inhaled fluticasone and oral prednisolone was prescribed and symptoms resolved. This is the first report of the health effects of DEEE mimicking asthma and rhinitis in Zimbabwe. CONCLUSIONS Atypical presentations of adult onset asthma in the absence of a history of either atopy or allergen specific IgE antibody sensitization should trigger in-depth evaluation of occupational exposure in all cases including office workers. Serial monitoring of lung function values should be used for diagnostic and monitoring of the patients.
Collapse
Affiliation(s)
- Elopy Sibanda
- Asthma, Allergy and Immune Dysfunction Clinic, Twin Palms Medical Centre, 113 Kwame Nkrumah Avenue, Harare, Zimbabwe
- Department of Pathology, Medical School, National University of Science and Technology, Bulawayo, Zimbabwe
- Division of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Nancy Makaza
- Asthma, Allergy and Immune Dysfunction Clinic, Twin Palms Medical Centre, 113 Kwame Nkrumah Avenue, Harare, Zimbabwe
| |
Collapse
|
59
|
Zheng P, Li Z, Zhou Z. Gut microbiome in type 1 diabetes: A comprehensive review. Diabetes Metab Res Rev 2018; 34:e3043. [PMID: 29929213 PMCID: PMC6220847 DOI: 10.1002/dmrr.3043] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 06/06/2018] [Accepted: 06/14/2018] [Indexed: 12/17/2022]
Abstract
Type 1 diabetes (T1D) is an autoimmune disease, which is characterized by the destruction of islet β cells in the pancreas triggered by genetic and environmental factors. In past decades, extensive familial and genome-wide association studies have revealed more than 50 risk loci in the genome. However, genetic susceptibility cannot explain the increased incidence of T1D worldwide, which is very likely attributed by the growing impact of environmental factors, especially gut microbiome. Recently, the role of gut microbiome in the pathogenesis of T1D has been uncovered by the increasing evidence from both human subjects and animal models, strongly indicating that gut microbiome might be a pivotal hub of T1D-triggering factors, especially environmental factors. In this review, we summarize the current aetiological and mechanism studies of gut microbiome in T1D. A better understanding of the role of gut microbiome in T1D may provide us with powerful prognostic and therapeutic tools in the near future.
Collapse
Affiliation(s)
- Peilin Zheng
- Department of Metabolism and Endocrinology, The Second Xiangya HospitalCentral South UniversityChangshaChina
- Key Laboratory of Diabetes Immunology (Central South University), Ministry of EducationNational Clinical Research Center for Metabolic DiseasesChangshaChina
| | - Zhixia Li
- Department of Metabolism and Endocrinology, The Second Xiangya HospitalCentral South UniversityChangshaChina
- Key Laboratory of Diabetes Immunology (Central South University), Ministry of EducationNational Clinical Research Center for Metabolic DiseasesChangshaChina
| | - Zhiguang Zhou
- Department of Metabolism and Endocrinology, The Second Xiangya HospitalCentral South UniversityChangshaChina
- Key Laboratory of Diabetes Immunology (Central South University), Ministry of EducationNational Clinical Research Center for Metabolic DiseasesChangshaChina
| |
Collapse
|
60
|
Pinares-Garcia P, Stratikopoulos M, Zagato A, Loke H, Lee J. Sex: A Significant Risk Factor for Neurodevelopmental and Neurodegenerative Disorders. Brain Sci 2018; 8:E154. [PMID: 30104506 PMCID: PMC6120011 DOI: 10.3390/brainsci8080154] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 08/08/2018] [Accepted: 08/10/2018] [Indexed: 12/11/2022] Open
Abstract
Males and females sometimes significantly differ in their propensity to develop neurological disorders. Females suffer more from mood disorders such as depression and anxiety, whereas males are more susceptible to deficits in the dopamine system including Parkinson's disease (PD), attention-deficit hyperactivity disorder (ADHD) and autism. Despite this, biological sex is rarely considered when making treatment decisions in neurological disorders. A better understanding of the molecular mechanism(s) underlying sex differences in the healthy and diseased brain will help to devise diagnostic and therapeutic strategies optimal for each sex. Thus, the aim of this review is to discuss the available evidence on sex differences in neuropsychiatric and neurodegenerative disorders regarding prevalence, progression, symptoms and response to therapy. We also discuss the sex-related factors such as gonadal sex hormones and sex chromosome genes and how these might help to explain some of the clinically observed sex differences in these disorders. In particular, we highlight the emerging role of the Y-chromosome gene, SRY, in the male brain and its potential role as a male-specific risk factor for disorders such as PD, autism, and ADHD in many individuals.
Collapse
Affiliation(s)
- Paulo Pinares-Garcia
- Brain and Gender laboratory, Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia.
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria 3168, Australia.
| | - Marielle Stratikopoulos
- Brain and Gender laboratory, Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia.
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria 3168, Australia.
| | - Alice Zagato
- Brain and Gender laboratory, Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia.
- School of Life and Environmental Sciences, Deakin University, Burwood, Victoria 3125, Australia.
| | - Hannah Loke
- Brain and Gender laboratory, Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia.
| | - Joohyung Lee
- Brain and Gender laboratory, Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia.
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria 3168, Australia.
| |
Collapse
|
61
|
Abstract
The practice of autoimmune neurology focuses on the diagnosis and treatment of a wide spectrum of neurological conditions driven by abnormal immune responses directed against neural tissues. These include autoimmune, paraneoplastic, postinfectious, and iatrogenic conditions. Symptoms of autoimmune neurologic disorders can be diverse and often difficult to recognize in their early stages, complicating the diagnosis. This review discusses the classification and management of common autoimmune neurological conditions, placing an emphasis on the rapid identification of autoimmune etiology and mechanism of immune dysfunction to allow for the timely institution of appropriate treatment.
Collapse
|
62
|
Marks ES, Bonnemaison ML, Brusnahan SK, Zhang W, Fan W, Garrison JC, Boesen EI. Renal iron accumulation occurs in lupus nephritis and iron chelation delays the onset of albuminuria. Sci Rep 2017; 7:12821. [PMID: 28993663 PMCID: PMC5634457 DOI: 10.1038/s41598-017-13029-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 09/19/2017] [Indexed: 12/11/2022] Open
Abstract
Proteins involved in iron homeostasis have been identified as biomarkers for lupus nephritis, a serious complication of systemic lupus erythematosus (SLE). We tested the hypothesis that renal iron accumulation occurs and contributes to renal injury in SLE. Renal non-heme iron levels were increased in the (New Zealand Black x New Zealand White) F1 (NZB/W) mouse model of lupus nephritis compared with healthy New Zealand White (NZW) mice in an age- and strain-dependent manner. Biodistribution studies revealed increased transferrin-bound iron accumulation in the kidneys of albuminuric NZB/W mice, but no difference in the accumulation of non-transferrin bound iron or ferritin. Transferrin excretion was significantly increased in albuminuric NZB/W mice, indicating enhanced tubular exposure and potential for enhanced tubular uptake following filtration. Expression of transferrin receptor and 24p3R were reduced in tubules from NZB/W compared to NZW mice, while ferroportin expression was unchanged and ferritin expression increased, consistent with increased iron accumulation and compensatory downregulation of uptake pathways. Treatment of NZB/W mice with the iron chelator deferiprone significantly delayed the onset of albuminuria and reduced blood urea nitrogen concentrations. Together, these findings suggest that pathological changes in renal iron homeostasis occurs in lupus nephritis, contributing to the development of kidney injury.
Collapse
Affiliation(s)
- Eileen S Marks
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Mathilde L Bonnemaison
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Susan K Brusnahan
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Wenting Zhang
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Wei Fan
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Jered C Garrison
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Erika I Boesen
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
63
|
Neuroimmunology and neuroepigenetics in the establishment of sex differences in the brain. Nat Rev Neurosci 2017. [PMID: 28638119 DOI: 10.1038/nrn.2017.61] [Citation(s) in RCA: 171] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The study of sex differences in the brain is a topic of neuroscientific study that has broad reaching implications for culture, society and biomedical science. Recent research in rodent models has led to dramatic shifts in our views of the mechanisms underlying the sexual differentiation of the brain. These include the surprising discoveries of a role for immune cells and inflammatory mediators in brain masculinization and a role for epigenetic suppression in brain feminization. How and to what degree these findings will translate to human brain development will be questions of central importance in future research in this field.
Collapse
|
64
|
Ko HH, Chien WC, Lin YH, Chung CH, Cheng SJ. Examining the correlation between diabetes and odontogenic infection: A nationwide, retrospective, matched-cohort study in Taiwan. PLoS One 2017; 12:e0178941. [PMID: 28594848 PMCID: PMC5464582 DOI: 10.1371/journal.pone.0178941] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Accepted: 05/22/2017] [Indexed: 12/24/2022] Open
Abstract
More than 90% of head and neck infections are caused by pathological changes originating in the teeth. When odontogenic infections are not properly treated, infections may spread to distant spaces and cause more serious infections in fascial spaces, ultimately leading to deep neck infections. Clinical experience has indicated that patients with diabetes mellitus (DM) may be more susceptible to facial cellulitis and deep neck infections caused by odontogenic infections. This study used the Taiwan National Health Insurance Database (NHIRD) to analyze and examine the correlation between DM and odontogenic infections in patients. To this end, this study analyzed 1 million NHIRD individual datasets from 2005, of which 964,182 individuals had medical treatment records. The insurance database also recorded related factors such as age, sex, duration of hospital stays, season, and whether patients were low income. We also analyzed the correlation between urbanization and the studied diseases. The results indicated that the correlation between facial cellulitis and DM patients was confirmed; facial cellulitis was most likely to occur 2 years after the initial DM diagnosis, with a risk occurrence 1.409 times greater than that of the control group. Facial cellulitis is more likely to occur in patients originating from poorer socioeconomic backgrounds, and female DM patients are more likely to experience this condition. These conclusions may facilitate the establishment of clinical guidelines for preventative education and treatment. Oral prevention and health education for high-risk patients, as well as early-stage surgical intervention and antibiotic usage in early-stage odontogenic infections, can prevent disease progression, improve patient recovery rates, and reduce the use and waste of medical resources.
Collapse
Affiliation(s)
- Hui-Hsin Ko
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan
- Department of Dentistry, National Taiwan University Hospital, College of Medicine, Taipei, Taiwan
- Department of Dentistry, National Taiwan University Hospital, Hsin-Chu Branch, Taiwan
| | - Wu-Chien Chien
- School of Public Health, National Defense Medical Center, Taipei, Taiwan
| | - Yen-Hung Lin
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan
- Department of Oral and Maxillofacial Surgery, Linkou Chang Gung Memorial Hospital
| | - Chi-Hsiang Chung
- School of Public Health, National Defense Medical Center, Taipei, Taiwan
| | - Shih-Jung Cheng
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan
- Department of Dentistry, National Taiwan University Hospital, College of Medicine, Taipei, Taiwan
- School of Dentistry, National Taiwan University, Taipei, Taiwan
- Graduate Institute of Oral Biology, School of Dentistry, National Taiwan University, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
65
|
Zhang Y, Zhou Z, Si Y. The role of parental health care utilization in children's unnecessary utilization in China: evidence from Shaanxi province. Int J Equity Health 2017; 16:47. [PMID: 28279211 PMCID: PMC5345193 DOI: 10.1186/s12939-017-0544-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 02/28/2017] [Indexed: 12/02/2022] Open
Abstract
Background China has a large population of children under 18 years of age, whose health is of great concern to the Chinese health care system. However, few studies have been conducted to analyze the factors associated with children’s unnecessary health care utilization in China. The objective of this study is to provide some empirical evidence on this issue by investigating the role of parental health care utilization in children’s unnecessary health care use. Methods The data were obtained from the fifth Health Service Survey of Shaanxi province in 2013. We employed three dependent variables to measure children’s health care utilization: the number of children’s outpatient visits during the past 2 weeks, whether or not infusion was used if the child had any outpatient visits during the past 2 weeks, and the number of children’s inpatient visits during last year. Based on specific characteristics of these outcome variables, negative binomial models were used for the non-negative numbers of outpatient and inpatient visits, while a probit model was used for the zero-one indicator variable showing whether infusion was used during outpatient visits. Results Based on a sample of 11,024 children, our results of multivariate analysis showed that children whose parents used outpatient care were estimated to have a larger number of outpatient visits than those whose parents did not have outpatient visits in the past 2 weeks (with a difference of 0.0393 visits). Among children having outpatient visits in the last 2 weeks, the probability of obtaining infusion was 57.01 percentage points higher for children whose parents used infusion in the past 2 weeks than the probability for those whose parents did not use infusion. The predicted number of inpatient visits was higher for children whose parents used inpatient services in the last year, compared with the group whose parents did not use (with a difference of 0.0567 visits). Moreover, we noted that the positive association between parental and children’s health care use was more prominent among younger children. Conclusions Chinese children whose parents were high health care users were more likely to overuse health care services, holding other factors constant. Parents can play an important role in reducing children’s unnecessary outpatient visits, infusion use, and inpatient visits. The results suggest that interventions aimed at affecting patterns of parental use may be helpful in improving appropriate health care utilization for children.
Collapse
Affiliation(s)
- Yi Zhang
- Jinhe Center for Economic Research, Xi'an Jiaotong University, No. 28 Xianning West Road, Xi'an, Shaanxi, 710049, China
| | - Zhongliang Zhou
- School of Public Policy and Administration, Xi'an Jiaotong University, No. 28 Xianning West Road, Xi'an, Shaanxi, 710049, China.
| | - Yafei Si
- School of Public Policy and Administration, Xi'an Jiaotong University, No. 28 Xianning West Road, Xi'an, Shaanxi, 710049, China
| |
Collapse
|
66
|
Mauvais-Jarvis F. Role of Sex Steroids in β Cell Function, Growth, and Survival. Trends Endocrinol Metab 2016; 27:844-855. [PMID: 27640750 PMCID: PMC5116277 DOI: 10.1016/j.tem.2016.08.008] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 08/27/2016] [Accepted: 08/29/2016] [Indexed: 01/08/2023]
Abstract
The gonads have long been considered endocrine glands, producing sex steroids such as estrogens, androgens, and progesterone (P4) for the sole purpose of sexual differentiation, puberty, and reproduction. Reproduction and energy metabolism are tightly linked, however, and gonadal steroids play an important role in sex-specific aspects of energy metabolism in various physiological conditions. In that respect, gonadal steroids also influence the secretion of insulin in a sex-specific manner. This review presents a perspective on the physiological roles of estrogens, androgens, and P4 via their receptors in pancreatic β cells in the gender-specific tuning of insulin secretion. I also discuss potential gender-specific therapeutic avenues that this knowledge may open in the future.
Collapse
Affiliation(s)
- Franck Mauvais-Jarvis
- Diabetes Discovery and Gender Medicine Laboratory, Section of Endocrinology and Metabolism, Department of Medicine, Tulane University Health Sciences Center, School of Medicine, New Orleans, LA, USA.
| |
Collapse
|
67
|
Lereim RR, Oveland E, Xiao Y, Torkildsen Ø, Wergeland S, Myhr KM, Sun SC, Berven FS. The Brain Proteome of the Ubiquitin Ligase Peli1 Knock-Out Mouse during Experimental Autoimmune Encephalomyelitis. ACTA ACUST UNITED AC 2016; 9:209-219. [PMID: 27746629 PMCID: PMC5061044 DOI: 10.4172/jpb.1000408] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The ubiquitin ligase Peli1 has previously been suggested as a potential treatment target in multiple sclerosis. In the multiple sclerosis disease model, experimental autoimmune encephalomyelitis, Peli1 knock-out led to less activated microglia and less inflammation in the central nervous system. Despite being important in microglia, Peli1 expression has also been detected in glial and neuronal cells. In the present study the overall brain proteomes of Peli1 knock-out mice and wild-type mice were compared prior to experimental autoimmune encephalomyelitis induction, at onset of the disease and at disease peak. Brain samples from the frontal hemisphere, peripheral from the extensive inflammatory foci, were analyzed using TMT-labeling of sample pools, and the discovered proteins were verified in individual mice using label-free proteomics. The greatest proteomic differences between Peli1 knock-out and wild-type mice were observed at the disease peak. In Peli1 knock-out a higher degree of antigen presentation, increased activity of adaptive and innate immune cells and alterations to proteins involved in iron metabolism were observed during experimental autoimmune encephalomyelitis. These results unravel global effects to the brain proteome when abrogating Peli1 expression, underlining the importance of Peli1 as a regulator of the immune response also peripheral to inflammatory foci during experimental autoimmune encephalomyelitis. The proteomics data is available in PRIDE with accession PXD003710.
Collapse
Affiliation(s)
- Ragnhild Reehorst Lereim
- Proteomics Unit, Department of Biomedicine, University of Bergen, Norway; Kristian Gerhard Jebsen MS Research Centre, Department of Clinical Medicine, University of Bergen, Bergen Norway
| | - Eystein Oveland
- Proteomics Unit, Department of Biomedicine, University of Bergen, Norway; Kristian Gerhard Jebsen MS Research Centre, Department of Clinical Medicine, University of Bergen, Bergen Norway
| | - Yichuan Xiao
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA; Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University, Shanghai 200031, China
| | - Øivind Torkildsen
- Kristian Gerhard Jebsen MS Research Centre, Department of Clinical Medicine, University of Bergen, Bergen Norway; Norwegian Multiple Sclerosis Competence Centre, Department of Neurology, Haukeland University Hospital, Bergen, Norway
| | - Stig Wergeland
- Kristian Gerhard Jebsen MS Research Centre, Department of Clinical Medicine, University of Bergen, Bergen Norway; Norwegian Multiple Sclerosis Competence Centre, Department of Neurology, Haukeland University Hospital, Bergen, Norway
| | - Kjell-Morten Myhr
- Kristian Gerhard Jebsen MS Research Centre, Department of Clinical Medicine, University of Bergen, Bergen Norway; Norwegian Multiple Sclerosis Competence Centre, Department of Neurology, Haukeland University Hospital, Bergen, Norway
| | - Shao-Cong Sun
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Frode S Berven
- Proteomics Unit, Department of Biomedicine, University of Bergen, Norway; Kristian Gerhard Jebsen MS Research Centre, Department of Clinical Medicine, University of Bergen, Bergen Norway; Norwegian Multiple Sclerosis Competence Centre, Department of Neurology, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
68
|
Abstract
Female/male ratios of autoimmune diseases range from 10: 1 to 1: 3, with similar severity between the sexes. Men and women respond similarly to the infection and to vaccination, arguing against intrinsic sex differences in immune response. In autoimmune-like illnesses caused by environmental agents sex discrepancy is explained by differences in exposure. Endogenous hormones could cause sex discrepancy if their effect is a threshold off-on switch rather than quantitatively variable. X-inactivation and imprinting could cause sex discrepancy. Other possibilities include chronobiologic differences and pregnancy and menstruation biologies in which men differ from women.
Collapse
Affiliation(s)
- M D Lockshin
- Barbara Volcker Center for Women and Rheumatic Disease Joan and Sanford Weill Medical College of Cornell University, Hospital for Special Surgery, New York, NY 10021, USA.
| |
Collapse
|
69
|
Whitacre CC. Summary and Future Directions. Mult Scler 2016. [DOI: 10.1177/135245859800400614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Caroline C. Whitacre
- Medical Microbiology and Immunology, Pathology, and Internal Medicine at The Ohio State University College of Medicine
| |
Collapse
|
70
|
Klil-Drori AJ, Tascilar K, Yin H, Aprikian A, Bitton A, Azoulay L. Androgen Deprivation Therapy and the Incidence of Inflammatory Bowel Disease in Patients With Prostate Cancer. Am J Epidemiol 2016; 184:15-22. [PMID: 27268031 PMCID: PMC4929242 DOI: 10.1093/aje/kwv307] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 11/03/2015] [Indexed: 12/12/2022] Open
Abstract
Androgen deprivation therapy (ADT) is the mainstay treatment for advanced prostate cancer. By lowering androgen levels, ADT inhibits the progression of prostate cancer, but it may also affect gut autoimmunity. We investigated the association between ADT and the incidence of inflammatory bowel disease using a cohort of 31,842 men newly diagnosed with prostate cancer between 1988 and 2014, identified in the United Kingdom Clinical Practice Research Datalink. Exposure to ADT was treated as a time-varying variable and lagged by 1 year to account for diagnostic delays, with nonuse as the reference category. During 133,018 person-years of follow-up, 48 men were newly diagnosed with ulcerative colitis (incidence rate (IR) = 36/100,000 person-years (PY)) and 12 were diagnosed with Crohn's disease (IR = 9/100,000 PY). In Cox proportional hazards models, ADT was associated with a decreased risk of ulcerative colitis (IR = 24/100,000 PY vs. IR = 50/100,000 PY; hazard ratio = 0.52, 95% confidence interval: 0.28, 0.99) and a nonsignificant decreased risk of Crohn's disease (hazard ratio = 0.38, 95% confidence interval: 0.11, 1.37). These findings indicate that the use of ADT may be associated with intestinal autoimmunity. Further research is warranted to replicate these findings and assess their clinical significance.
Collapse
Affiliation(s)
| | | | | | | | | | - Laurent Azoulay
- Correspondence to Dr. Laurent Azoulay, Center for Clinical Epidemiology, Lady Davis Institute, Jewish General Hospital, 3755 Côte Sainte-Catherine, H-425.1, Montreal, QC H3T 1E2, Canada (e-mail: )
| |
Collapse
|
71
|
Zhu ML, Bakhru P, Conley B, Nelson JS, Free M, Martin A, Starmer J, Wilson EM, Su MA. Sex bias in CNS autoimmune disease mediated by androgen control of autoimmune regulator. Nat Commun 2016; 7:11350. [PMID: 27072778 PMCID: PMC5512610 DOI: 10.1038/ncomms11350] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 03/16/2016] [Indexed: 12/19/2022] Open
Abstract
Male gender is protective against multiple sclerosis and other T-cell-mediated autoimmune diseases. This protection may be due, in part, to higher androgen levels in males. Androgen binds to the androgen receptor (AR) to regulate gene expression, but how androgen protects against autoimmunity is not well understood. Autoimmune regulator (Aire) prevents autoimmunity by promoting self-antigen expression in medullary thymic epithelial cells, such that developing T cells that recognize these self-antigens within the thymus undergo clonal deletion. Here we show that androgen upregulates Aire-mediated thymic tolerance to protect against autoimmunity. Androgen recruits AR to Aire promoter regions, with consequent enhancement of Aire transcription. In mice and humans, thymic Aire expression is higher in males compared with females. Androgen administration and male gender protect against autoimmunity in a multiple sclerosis mouse model in an Aire-dependent manner. Thus, androgen control of an intrathymic Aire-mediated tolerance mechanism contributes to gender differences in autoimmunity.
Collapse
MESH Headings
- Androgens/pharmacology
- Animals
- Antigens/metabolism
- Autoimmune Diseases/metabolism
- Autoimmune Diseases/pathology
- Central Nervous System/pathology
- Dihydrotestosterone/pharmacology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/prevention & control
- Female
- Fluorescent Antibody Technique
- Humans
- Male
- Mice, Inbred C57BL
- Myelin-Oligodendrocyte Glycoprotein/genetics
- Myelin-Oligodendrocyte Glycoprotein/metabolism
- Promoter Regions, Genetic/genetics
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Androgen/metabolism
- Sexism
- Thymus Gland/drug effects
- Thymus Gland/metabolism
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Up-Regulation/drug effects
- AIRE Protein
Collapse
Affiliation(s)
- Meng-Lei Zhu
- Division of Endocrinology, Department of Pediatrics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Pearl Bakhru
- Division of Endocrinology, Department of Pediatrics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Bridget Conley
- Division of Endocrinology, Department of Pediatrics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Jennifer S. Nelson
- Division of Cardiothoracic Surgery, Department of Surgery, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Meghan Free
- Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Aaron Martin
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Joshua Starmer
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Elizabeth M. Wilson
- Division of Endocrinology, Department of Pediatrics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- Department of Biochemistry and Biophysics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Maureen A. Su
- Division of Endocrinology, Department of Pediatrics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| |
Collapse
|
72
|
Elderman M, van Beek A, Brandsma E, de Haan B, Savelkoul H, de Vos P, Faas M. Sex impacts Th1 cells, Tregs, and DCs in both intestinal and systemic immunity in a mouse strain and location-dependent manner. Biol Sex Differ 2016; 7:21. [PMID: 27051505 PMCID: PMC4820953 DOI: 10.1186/s13293-016-0075-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 03/30/2016] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Males and females have a different predisposition for the development of intestinal disorders, like inflammatory bowel disease (IBD). We hypothesized that sex specific differences in intestinal immune responses may underlie this bias. To test this hypothesis, we studied sex differences in immune cell populations in the Peyer's patches (PP). For comparison with systemic immunity, we studied spleen cells. METHODS Two mouse strains with different susceptibility for developing colitis (BALB/c and C57Bl/6) were used. Using flow cytometry, we measured the percentage of T cells, Th1, Th17, and Treg cells in the PP and spleen. In addition, we measured the percentages of NK cells, macrophages, myeloid, and lymphoid dendritic cells (DCs) and their expression of CD80 and CD103. Moreover, we measured percentages of monocyte subsets in the peripheral circulation. Results were tested using two-way ANOVA, p < 0.05. RESULTS Males had a lower percentage of T cells in both PP and spleen (PP BALB/c 22.1 %, B6 13.6 %; spleen BALB/c 4.7 %, B6 19.9 %) but a higher percentage of Th1 cell in both tissues (PP BALB/c 350 %, B6 109.5 %; spleen BALB/c 48.7 %, B6 41.9 %) than females. They also had a higher percentage of Tregs in the spleen than females (BALB/c 20.5 %, B6 4.5 %). Furthermore, males had a higher percentage of CD80(+) DCs in both the PP and spleen (lymphoid DCs in PP BALB/c 104.7 %, B6 29.6 %; spleen BALB/c 72.2 %, B6 44.2 %; myeloid DCs in PP BALB/c 80.5 %, B6 93.3 %; spleen BALB/c 88.5 %, B6 50.8 %) and a higher percentage of lymphoid CD103(+) DCs in the spleen than females (BALB/c 41.5 %, B6 28.3 %). The percentage of NK cells was decreased in the spleen (BALB/c 12.5 %, B6 25.1 %) but increased in the PP (BALB/c 75.7 %, B6 78.6 %) of males as compared with females. Strain differences were also found in the PP; BALB/c mice had a higher percentage of T cells (males 58.1 %, females 75.5 %), a higher Th/Tc ratio (males 81.0 %, females 134.2 %), less FoxP3(+)CD25(-) T cells (males 14.6 %, females 30.0 %), more DCs (males 14.8 %, females 15.7 %) and macrophages (males 67.9 %, females 141.2 %), and more NK cells (males 160 %, females164.3 %) than BALB/c mice. CONCLUSIONS In this study, we show sex differences in intestinal and peripheral immune populations. These differences may underlie sex differences in intestinal disorders like IBD, and this information may be an important knowledge for the treatment of intestinal-related diseases.
Collapse
Affiliation(s)
- Marlies Elderman
- Top Institute Food and Nutrition, Wageningen, The Netherlands ; Division of Medical Biology, Department of Pathology and Medical Biology, University Medical Centre Groningen, Groningen, The Netherlands
| | - Adriaan van Beek
- Top Institute Food and Nutrition, Wageningen, The Netherlands ; Cell Biology and Immunology Group, Wageningen University, Wageningen, The Netherlands
| | - Eelke Brandsma
- Top Institute Food and Nutrition, Wageningen, The Netherlands ; Division of Medical Biology, Department of Pathology and Medical Biology, University Medical Centre Groningen, Groningen, The Netherlands
| | - Bart de Haan
- Division of Medical Biology, Department of Pathology and Medical Biology, University Medical Centre Groningen, Groningen, The Netherlands
| | - Huub Savelkoul
- Cell Biology and Immunology Group, Wageningen University, Wageningen, The Netherlands
| | - Paul de Vos
- Top Institute Food and Nutrition, Wageningen, The Netherlands ; Division of Medical Biology, Department of Pathology and Medical Biology, University Medical Centre Groningen, Groningen, The Netherlands
| | - Marijke Faas
- Division of Medical Biology, Department of Pathology and Medical Biology, University Medical Centre Groningen, Groningen, The Netherlands ; Department of Obstetrics and Gynaecology, University of Groningen and University Medical Centre Groningen, Groningen, The Netherlands
| |
Collapse
|
73
|
Gender-specific incidence of autoimmune diseases from national registers. J Autoimmun 2016; 69:102-6. [PMID: 26994904 DOI: 10.1016/j.jaut.2016.03.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 03/08/2016] [Accepted: 03/08/2016] [Indexed: 12/12/2022]
Abstract
BACKGROUND It is widely believed that autoimmune diseases affect predominantly in women, but the available evidence came from case control study with potential selection and recall bias. We aimed to examine the gender-specific incidence of autoimmune diseases by using national wide registers in Sweden. METHODS Swedish Hospital Discharge Register and Outpatient Register were used to identify a set of autoimmune diseases between 1987 and 2010. Gender-specific incidence rate was standardized directly according to the Swedish age distribution in 2000. RESULTS A total of 403,757 individuals were diagnosed with autoimmune diseases between 1987 and 2010 in Sweden. The overall incidence of 32 autoimmune disease was 60% higher in women than men. Female predominance was noted in 18 specific diseases, whereas the rest of them showed no difference or male predominance. The age of onset was different between men and women in 27 autoimmune diseases. CONCLUSIONS Our study suggested that the classical view of female predominance of autoimmune diseases may be far from striking than previously believed. Further studies are needed to examine whether there is true difference between men and women.
Collapse
|
74
|
Khan D, Ansar Ahmed S. The Immune System Is a Natural Target for Estrogen Action: Opposing Effects of Estrogen in Two Prototypical Autoimmune Diseases. Front Immunol 2016; 6:635. [PMID: 26779182 PMCID: PMC4701921 DOI: 10.3389/fimmu.2015.00635] [Citation(s) in RCA: 243] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 12/03/2015] [Indexed: 12/11/2022] Open
Abstract
Analogous to other physiological systems, the immune system also demonstrates remarkable sex differences. Although the reasons for sex differences in immune responses are not precisely understood, it potentially involves differences in sex hormones (estrogens, androgens, and differential sex hormone receptor-mediated events), X-chromosomes, microbiome, epigenetics among others. Overall, females tend to have more responsive and robust immune system compared to their male counterparts. It is therefore not surprising that females respond more aggressively to self-antigens and are more susceptible to autoimmune diseases. Female hormone (estrogen or 17β-estradiol) can potentially act on all cellular subsets of the immune system through estrogen receptor-dependent and -independent mechanisms. This minireview highlights differential expression of estrogen receptors on immune cells, major estrogen-mediated signaling pathways, and their effect on immune cells. Since estrogen has varied effects in female-predominant autoimmune diseases such as multiple sclerosis and systemic lupus erythematosus, we will mechanistically postulate the potential differential role of estrogen in these chronic debilitating diseases.
Collapse
Affiliation(s)
- Deena Khan
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech , Blacksburg, VA , USA
| | - S Ansar Ahmed
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech , Blacksburg, VA , USA
| |
Collapse
|
75
|
Forger NG, Strahan JA, Castillo-Ruiz A. Cellular and molecular mechanisms of sexual differentiation in the mammalian nervous system. Front Neuroendocrinol 2016; 40:67-86. [PMID: 26790970 PMCID: PMC4897775 DOI: 10.1016/j.yfrne.2016.01.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 12/31/2015] [Accepted: 01/09/2016] [Indexed: 01/16/2023]
Abstract
Neuroscientists are likely to discover new sex differences in the coming years, spurred by the National Institutes of Health initiative to include both sexes in preclinical studies. This review summarizes the current state of knowledge of the cellular and molecular mechanisms underlying sex differences in the mammalian nervous system, based primarily on work in rodents. Cellular mechanisms examined include neurogenesis, migration, the differentiation of neurochemical and morphological cell phenotype, and cell death. At the molecular level we discuss evolving roles for epigenetics, sex chromosome complement, the immune system, and newly identified cell signaling pathways. We review recent findings on the role of the environment, as well as genome-wide studies with some surprising results, causing us to re-think often-used models of sexual differentiation. We end by pointing to future directions, including an increased awareness of the important contributions of tissues outside of the nervous system to sexual differentiation of the brain.
Collapse
Affiliation(s)
- Nancy G Forger
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, United States.
| | - J Alex Strahan
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, United States.
| | | |
Collapse
|
76
|
Yadav A, Kumar A, Das M, Tripathi A. Sodium benzoate, a food preservative, affects the functional and activation status of splenocytes at non cytotoxic dose. Food Chem Toxicol 2015; 88:40-7. [PMID: 26706697 DOI: 10.1016/j.fct.2015.12.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Revised: 12/11/2015] [Accepted: 12/14/2015] [Indexed: 12/31/2022]
Abstract
Sodium benzoate (SB) is a widely used food preservative due to its bacteriostatic and fungistatic properties. The acceptable daily intake of SB is 5 mg/kg-bw, however, it has been found to be used in the food commodities at relatively high levels (2119 mg/kg). Earlier studies on SB have shown its immunosuppressive properties, but comprehensive immunotoxicity data is lacking. Our studies have shown that SB was non cytotoxic in splenocytes up to 1000 μg/ml for 72 h, however at 2500 μg/ml it was found to be cytotoxic. Thus, 1000 μg/ml dose of SB was chosen for the subsequent experiments. SB significantly suppresses the proliferation of Con A and LPS stimulated splenocytes at 72 h, while allogenic response of T cells was significantly decreased after 96 h. SB did not affect the relative expression of CD3e or CD4 molecules following 72 h exposure, however, it downregulated the relative expression of CD8 co-receptor. Further, exposure of splenocytes to SB for 72 h led to reduced expression of CD28 and CD95, which play a vital role in T cell activation. SB also suppresses the relative expression of CD19, CD40 and CD95 receptors on B cells after 72 h. In addition to the functional responses, SB lowered the expression of IL4, IL6, IFNγ and IL17 cytokines in Con A stimulated splenocytes; and IL6, IFNγ and TNFα in LPS stimulated splenocytes following 48 h of exposure. Taken together, the present study is suggestive of the immunomodulatory potential of SB.
Collapse
Affiliation(s)
- Ashish Yadav
- Food Toxicology Lab, Food, Drug and Chemical Toxicology Group, CSIR- Indian Institute of Toxicology Research (CSIR-IITR), M.G. Marg, Lucknow, 226001, Uttar Pradesh, India; Molecular Immunology Lab, School of Biotechnology, Faculty of Science, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India
| | - Arvind Kumar
- Molecular Immunology Lab, School of Biotechnology, Faculty of Science, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India
| | - Mukul Das
- Food Toxicology Lab, Food, Drug and Chemical Toxicology Group, CSIR- Indian Institute of Toxicology Research (CSIR-IITR), M.G. Marg, Lucknow, 226001, Uttar Pradesh, India.
| | - Anurag Tripathi
- Food Toxicology Lab, Food, Drug and Chemical Toxicology Group, CSIR- Indian Institute of Toxicology Research (CSIR-IITR), M.G. Marg, Lucknow, 226001, Uttar Pradesh, India.
| |
Collapse
|
77
|
Brandt JE, Priori R, Valesini G, Fairweather D. Sex differences in Sjögren's syndrome: a comprehensive review of immune mechanisms. Biol Sex Differ 2015; 6:19. [PMID: 26535108 PMCID: PMC4630965 DOI: 10.1186/s13293-015-0037-7] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 09/17/2015] [Indexed: 02/03/2023] Open
Abstract
Autoimmune diseases (ADs) are estimated to affect between 5 and 8 % of the US population, and approximately 80 % of these patients are women. Sjögren’s syndrome (SS) is an AD that occurs predominately in women over men (16:1). The hallmark characteristic of SS is diminished secretory production from the primary exocrine gland and the lacrimal or salivary glands resulting in symptoms of dry eye and mouth. The disease is believed to be mediated by an inflammatory and autoantibody response directed against salivary and lacrimal gland tissues. This review will examine the literature on sex differences in the immune response of patients and animal models of Sjögren’s syndrome in order to gain a better understanding of disease pathogenesis.
Collapse
Affiliation(s)
- Jessica E Brandt
- Department of Environmental Health Sciences, Johns Hopkins University, Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD 21205 USA ; Reumatologia, Dipartimento di Medicina Interna e Specialita Mediche, Sapienza Universita di Roma, 00161 Rome, Italy
| | - Roberta Priori
- Reumatologia, Dipartimento di Medicina Interna e Specialita Mediche, Sapienza Universita di Roma, 00161 Rome, Italy
| | - Guido Valesini
- Reumatologia, Dipartimento di Medicina Interna e Specialita Mediche, Sapienza Universita di Roma, 00161 Rome, Italy
| | - DeLisa Fairweather
- Department of Environmental Health Sciences, Johns Hopkins University, Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD 21205 USA ; Department of Cardiovascular Diseases, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| |
Collapse
|
78
|
McDonald G, Cabal N, Vannier A, Umiker B, Yin RH, Orjalo AV, Johansson HE, Han JH, Imanishi-Kari T. Female Bias in Systemic Lupus Erythematosus is Associated with the Differential Expression of X-Linked Toll-Like Receptor 8. Front Immunol 2015; 6:457. [PMID: 26441962 PMCID: PMC4561825 DOI: 10.3389/fimmu.2015.00457] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 08/24/2015] [Indexed: 11/28/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease characterized by the production of anti-nuclear antibodies. SLE is one of many autoimmune disorders that have a strong gender bias, with 70–90% of SLE patients being female. Several explanations have been postulated to account for the severity of autoimmune diseases in females, including hormonal, microbiota, and gene dosage differences. X-linked toll-like receptors (TLRs) have recently been implicated in disease progression in females. Our previous studies using the 564Igi mouse model of SLE on a Tlr7 and Tlr9 double knockout background showed that the presence of Tlr8 on both X chromosomes was required for the production of IgG autoantibodies, Ifn-I expression and granulopoiesis in females. Here, we show the results of our investigation into the role of Tlr8 expression in SLE pathogenesis in 564Igi females. Female mice have an increase in serum pathogenic anti-RNA IgG2a and IgG2b autoantibodies. 564Igi mice have also been shown to have an increase in neutrophils in vivo, which are major contributors to Ifn-α expression. Here, we show that neutrophils from C57BL/6 mice express Ifn-α in response to 564 immune complexes and TLR8 activation. Bone marrow-derived macrophages from 564Igi females have a significant increase in Tlr8 expression compared to male-derived cells, and RNA fluorescence in situ hybridization data suggest that Tlr8 may escape X-inactivation in female-derived macrophages. These results propose a model by which females may be more susceptible to SLE pathogenesis due to inefficient inactivation of Tlr8.
Collapse
Affiliation(s)
- Gabrielle McDonald
- Department of Integrative Physiology and Pathobiology, Tufts University , Boston, MA , USA
| | - Nicholas Cabal
- Department of Integrative Physiology and Pathobiology, Tufts University , Boston, MA , USA
| | - Augustin Vannier
- Department of Integrative Physiology and Pathobiology, Tufts University , Boston, MA , USA
| | - Benjamin Umiker
- Department of Integrative Physiology and Pathobiology, Tufts University , Boston, MA , USA
| | | | | | | | - Jin-Hwan Han
- Merck Research Laboratories , Palo Alto, CA , USA
| | - Thereza Imanishi-Kari
- Department of Integrative Physiology and Pathobiology, Tufts University , Boston, MA , USA
| |
Collapse
|
79
|
de Vries GJ, Forger NG. Sex differences in the brain: a whole body perspective. Biol Sex Differ 2015; 6:15. [PMID: 26279833 PMCID: PMC4536872 DOI: 10.1186/s13293-015-0032-z] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 07/28/2015] [Indexed: 12/29/2022] Open
Abstract
Most writing on sexual differentiation of the mammalian brain (including our own) considers just two organs: the gonads and the brain. This perspective, which leaves out all other body parts, misleads us in several ways. First, there is accumulating evidence that all organs are sexually differentiated, and that sex differences in peripheral organs affect the brain. We demonstrate this by reviewing examples involving sex differences in muscles, adipose tissue, the liver, immune system, gut, kidneys, bladder, and placenta that affect the nervous system and behavior. The second consequence of ignoring other organs when considering neural sex differences is that we are likely to miss the fact that some brain sex differences develop to compensate for differences in the internal environment (i.e., because male and female brains operate in different bodies, sex differences are required to make output/function more similar in the two sexes). We also consider evidence that sex differences in sensory systems cause male and female brains to perceive different information about the world; the two sexes are also perceived by the world differently and therefore exposed to differences in experience via treatment by others. Although the topic of sex differences in the brain is often seen as much more emotionally charged than studies of sex differences in other organs, the dichotomy is largely false. By putting the brain firmly back in the body, sex differences in the brain are predictable and can be more completely understood.
Collapse
Affiliation(s)
- Geert J. de Vries
- Neuroscience Institute, Georgia State University, P.O. Box 5030, Atlanta, GA 30302-5030 USA
| | - Nancy G. Forger
- Neuroscience Institute, Georgia State University, P.O. Box 5030, Atlanta, GA 30302-5030 USA
| |
Collapse
|
80
|
Mark S, Scott GBI, Donoviel DB, Leveton LB, Mahoney E, Charles JB, Siegel B. The impact of sex and gender on adaptation to space: executive summary. J Womens Health (Larchmt) 2015; 23:941-7. [PMID: 25401937 DOI: 10.1089/jwh.2014.4914] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
This review article is a compendium of six individual manuscripts, a Commentary, and an Executive Summary. This body of work is entitled "The Impact of Sex and Gender on Adaptation to Space" and was developed in response to a recommendation from the 2011 National Academy of Sciences Decadal Survey, "Recapturing a Future for Space Exploration: Life and Physical Sciences for a New Era," which emphasized the need to fully understand sex and gender differences in space. To ensure the health and safety of male and female astronauts during long-duration space missions, it is imperative to examine and understand the influences that sex and gender have on physiological and psychological changes that occur during spaceflight. In this collection of manuscripts, six workgroups investigated and summarized the current body of published and unpublished human and animal research performed to date related to sex- and gender-based differences in the areas of cardiovascular, immunological, sensorimotor, musculoskeletal, reproductive, and behavioral adaptations to human spaceflight. Each workgroup consisted of scientists and clinicians from academia, the National Aeronautics and Space Administration (NASA), and other federal agencies and was co-chaired by one representative from NASA and one from the external scientific community. The workgroups met via telephone and e-mail over 6 months to review literature and data from space- and ground-based studies to identify sex and gender factors affecting crew health. In particular, the Life Sciences Data Archive and the Lifetime Surveillance of Astronaut Health were extensively mined. The groups identified certain sex-related differences that impact the risks and the optimal medical care required by space-faring women and men. It represents innovative research in sex and gender-based biology that impacts those individuals that are at the forefront of space exploration.
Collapse
Affiliation(s)
- Saralyn Mark
- 1 Advanced Exploration Systems Division, Human Exploration and Operations Mission Directorate, National Aeronautics and Space Administration, Washington, DC
| | | | | | | | | | | | | |
Collapse
|
81
|
Kennedy AR, Crucian B, Huff JL, Klein SL, Morens D, Murasko D, Nickerson CA, Sonnenfeld G. Effects of sex and gender on adaptation to space: immune system. J Womens Health (Larchmt) 2015; 23:956-8. [PMID: 25401940 DOI: 10.1089/jwh.2014.4913] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
This review is focused on sex and gender effects on immunological alterations occurring during space flight. Sex differences in immune function and the outcome of inflammatory, infectious, and autoimmune diseases are well documented. The work of the Immunology Workgroup identified numerous reasons why there could be sex and/or gender differences observed during and after spaceflight, but thus far, there has been very little investigation in this area of research. In most cases, this is due to either a low total number of subjects or the minimal number of female flight crew members available for these studies. Thus, the availability of a sufficient number of female subjects to enable statistical analysis of the data has been a limiting factor. As the inclusion of female crew members has increased in the recent past, such studies should be possible in the future. It is very difficult to obtain immunologic and infectious data in small animals that can be usefully extrapolated to humans undergoing spaceflight. Thus, it is recommended by the Immunology Workgroup that a greater emphasis be placed on studying astronauts themselves, with a focus on long-term evaluations of specific, known infectious risks.
Collapse
Affiliation(s)
- Ann R Kennedy
- 1 Department of Radiation Oncology, University of Pennsylvania Perelman School of Medicine , Philadelphia, Pennsylvania
| | | | | | | | | | | | | | | |
Collapse
|
82
|
Crampton SP, Morawski PA, Bolland S. Linking susceptibility genes and pathogenesis mechanisms using mouse models of systemic lupus erythematosus. Dis Model Mech 2015; 7:1033-46. [PMID: 25147296 PMCID: PMC4142724 DOI: 10.1242/dmm.016451] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Systemic lupus erythematosus (SLE) represents a challenging autoimmune disease from a clinical perspective because of its varied forms of presentation. Although broad-spectrum steroids remain the standard treatment for SLE, they have many side effects and only provide temporary relief from the symptoms of the disease. Thus, gaining a deeper understanding of the genetic traits and biological pathways that confer susceptibility to SLE will help in the design of more targeted and effective therapeutics. Both human genome-wide association studies (GWAS) and investigations using a variety of mouse models of SLE have been valuable for the identification of the genes and pathways involved in pathogenesis. In this Review, we link human susceptibility genes for SLE with biological pathways characterized in mouse models of lupus, and discuss how the mechanistic insights gained could advance drug discovery for the disease.
Collapse
Affiliation(s)
- Steve P Crampton
- Laboratory of Immunogenetics, National Institute of Allergic and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Peter A Morawski
- Laboratory of Immunogenetics, National Institute of Allergic and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Silvia Bolland
- Laboratory of Immunogenetics, National Institute of Allergic and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| |
Collapse
|
83
|
Chang D, Gao F, Slavney A, Ma L, Waldman YY, Sams AJ, Billing-Ross P, Madar A, Spritz R, Keinan A. Accounting for eXentricities: analysis of the X chromosome in GWAS reveals X-linked genes implicated in autoimmune diseases. PLoS One 2014; 9:e113684. [PMID: 25479423 PMCID: PMC4257614 DOI: 10.1371/journal.pone.0113684] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 10/30/2014] [Indexed: 12/12/2022] Open
Abstract
Many complex human diseases are highly sexually dimorphic, suggesting a potential contribution of the X chromosome to disease risk. However, the X chromosome has been neglected or incorrectly analyzed in most genome-wide association studies (GWAS). We present tailored analytical methods and software that facilitate X-wide association studies (XWAS), which we further applied to reanalyze data from 16 GWAS of different autoimmune and related diseases (AID). We associated several X-linked genes with disease risk, among which (1) ARHGEF6 is associated with Crohn's disease and replicated in a study of ulcerative colitis, another inflammatory bowel disease (IBD). Indeed, ARHGEF6 interacts with a gastric bacterium that has been implicated in IBD. (2) CENPI is associated with three different AID, which is compelling in light of known associations with AID of autosomal genes encoding centromere proteins, as well as established autosomal evidence of pleiotropy between autoimmune diseases. (3) We replicated a previous association of FOXP3, a transcription factor that regulates T-cell development and function, with vitiligo; and (4) we discovered that C1GALT1C1 exhibits sex-specific effect on disease risk in both IBDs. These and other X-linked genes that we associated with AID tend to be highly expressed in tissues related to immune response, participate in major immune pathways, and display differential gene expression between males and females. Combined, the results demonstrate the importance of the X chromosome in autoimmunity, reveal the potential of extensive XWAS, even based on existing data, and provide the tools and incentive to properly include the X chromosome in future studies.
Collapse
Affiliation(s)
- Diana Chang
- Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, New York, United States of America
- Program in Computational Biology and Medicine, Cornell University, Ithaca, New York, United States of America
| | - Feng Gao
- Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, New York, United States of America
| | - Andrea Slavney
- Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, New York, United States of America
- Graduate Field of Genetics, Genomics and Development, Cornell University, Ithaca, New York, United States of America
| | - Li Ma
- Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, New York, United States of America
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland, United States of America
| | - Yedael Y. Waldman
- Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, New York, United States of America
| | - Aaron J. Sams
- Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, New York, United States of America
| | - Paul Billing-Ross
- Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, New York, United States of America
- Graduate Field of Genetics, Genomics and Development, Cornell University, Ithaca, New York, United States of America
| | - Aviv Madar
- Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, New York, United States of America
| | - Richard Spritz
- Human Medical Genetics and Genomics Program, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Alon Keinan
- Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, New York, United States of America
- Program in Computational Biology and Medicine, Cornell University, Ithaca, New York, United States of America
- Graduate Field of Genetics, Genomics and Development, Cornell University, Ithaca, New York, United States of America
| |
Collapse
|
84
|
Multiple sclerosis at menopause: Potential neuroprotective effects of estrogen. Maturitas 2014; 80:133-9. [PMID: 25544310 DOI: 10.1016/j.maturitas.2014.11.013] [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: 10/16/2014] [Revised: 11/19/2014] [Accepted: 11/20/2014] [Indexed: 12/17/2022]
Abstract
Multiple sclerosis (MS) is an autoimmune demyelinating and neurodegenerative condition of the central nervous system that preferentially afflicts women more than men. Low estrogen states such as menopause and the postpartum period favor exacerbations of multiple sclerosis in women with the disease. Existing and emerging evidence suggests a role for estrogen in the alleviation of symptoms and reversal of pathology associated with MS. While clinical evidence is sparse regarding the benefit of estrogen therapy for women at risk for MS exacerbations, scientific data demonstrates that estrogen potentiates numerous neuroprotective effects on the central nervous system (CNS). Estrogens play a wide range of roles involved in MS disease pathophysiology, including increasing antiinflammatory cytokines, decreasing demyelination, and enhancing oxidative and energy producing processes in CNS cells.
Collapse
|
85
|
McDougle CJ, Landino SM, Vahabzadeh A, O'Rourke J, Zurcher NR, Finger BC, Palumbo ML, Helt J, Mullett JE, Hooker JM, Carlezon WA. Toward an immune-mediated subtype of autism spectrum disorder. Brain Res 2014; 1617:72-92. [PMID: 25445995 DOI: 10.1016/j.brainres.2014.09.048] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Revised: 09/15/2014] [Accepted: 09/20/2014] [Indexed: 11/25/2022]
Abstract
A role for immunological involvement in autism spectrum disorder (ASD) has long been hypothesized. This review includes four sections describing (1) evidence for a relationship between familial autoimmune disorders and ASD; (2) results from post-mortem and neuroimaging studies that investigated aspects of neuroinflammation in ASD; (3) findings from animal model work in ASD involving inflammatory processes; and (4) outcomes from trials of anti-inflammatory/immune-modulating drugs in ASD that have appeared in the literature. Following each section, ideas are provided for future research, suggesting paths forward in the continuing effort to define the role of immune factors and inflammation in the pathophysiology of a subtype of ASD. This article is part of a Special Issue entitled SI: Neuroimmunology in Health And Disease.
Collapse
Affiliation(s)
- Christopher J McDougle
- Lurie Center for Autism, Lexington, MA, United States; Massachusetts General Hospital, Boston, MA, United States; Harvard Medical School, Boston, MA, United States.
| | - Samantha M Landino
- Behavioral Genetics Laboratory, Belmont, MA, United States; McLean Hospital, Belmont, MA, United States
| | - Arshya Vahabzadeh
- Massachusetts General Hospital, Boston, MA, United States; McLean Hospital, Belmont, MA, United States; Harvard Medical School, Boston, MA, United States
| | - Julia O'Rourke
- Lurie Center for Autism, Lexington, MA, United States; Massachusetts General Hospital, Boston, MA, United States; Harvard Medical School, Boston, MA, United States
| | - Nicole R Zurcher
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, United States; Massachusetts General Hospital, Boston, MA, United States; Harvard Medical School, Boston, MA, United States
| | - Beate C Finger
- Behavioral Genetics Laboratory, Belmont, MA, United States; McLean Hospital, Belmont, MA, United States; Harvard Medical School, Boston, MA, United States
| | - Michelle L Palumbo
- Lurie Center for Autism, Lexington, MA, United States; Massachusetts General Hospital, Boston, MA, United States; Harvard Medical School, Boston, MA, United States
| | - Jessica Helt
- Lurie Center for Autism, Lexington, MA, United States; Massachusetts General Hospital, Boston, MA, United States
| | - Jennifer E Mullett
- Lurie Center for Autism, Lexington, MA, United States; Massachusetts General Hospital, Boston, MA, United States
| | - Jacob M Hooker
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, United States; Massachusetts General Hospital, Boston, MA, United States; Harvard Medical School, Boston, MA, United States
| | - William A Carlezon
- Behavioral Genetics Laboratory, Belmont, MA, United States; McLean Hospital, Belmont, MA, United States; Harvard Medical School, Boston, MA, United States
| |
Collapse
|
86
|
Meleine M, Matricon J. Gender-related differences in irritable bowel syndrome: Potential mechanisms of sex hormones. World J Gastroenterol 2014; 20:6725-6743. [PMID: 24944465 PMCID: PMC4051914 DOI: 10.3748/wjg.v20.i22.6725] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 02/08/2014] [Accepted: 03/19/2014] [Indexed: 02/06/2023] Open
Abstract
According to epidemiological studies, twice as many women as men are affected by irritable bowel syndrome (IBS) in western countries, suggesting a role for sex hormones in IBS pathophysiology. Despite growing evidence about the implications of sex hormones in IBS symptom modulation, data on mechanisms by which they influence disease development are sparse. This review aims to determine the state of knowledge about the role of sex hormones in sensorimotor dysfunctions and to address the possible interplay of sex hormones with common risk factors associated with IBS. The scientific bibliography was searched using the following keywords: irritable bowel syndrome, sex, gender, ovarian hormone, estradiol, progesterone, testosterone, symptoms, pain, sensitivity, motility, permeability, stress, immune system, brain activity, spinal, supraspinal, imaging. Ovarian hormones variations along the menstrual cycle affect sensorimotor gastrointestinal function in both healthy and IBS populations. They can modulate pain processing by interacting with neuromodulator systems and the emotional system responsible for visceral pain perception. These hormones can also modulate the susceptibility to stress, which is a pivotal factor in IBS occurrence and symptom severity. For instance, estrogen-dependent hyper-responsiveness to stress can promote immune activation or impairments of gut barrier function. In conclusion, whereas it is important to keep in mind that ovarian hormones cannot be considered as a causal factor of IBS, they arguably modulate IBS onset and symptomatology. However, our understanding of the underlying mechanisms remains limited and studies assessing the link between IBS symptoms and ovarian hormone levels are needed to improve our knowledge of the disease evolution with regard to gender. Further studies assessing the role of male hormones are also needed to understand fully the role of sex hormones in IBS. Finally, investigation of brain-gut interactions is critical to decipher how stress, ovarian hormones, and female brain processing of pain can translate into gut dysfunctions.
Collapse
|
87
|
Pisto L, Vadén A, Sillanmäki L, Mattila K. Childhood Adversities are Associated with Diabetes Management in Working Age in Finland. INTERNATIONAL JOURNAL OF FAMILY MEDICINE 2014; 2014:864572. [PMID: 24868461 PMCID: PMC4020449 DOI: 10.1155/2014/864572] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2013] [Revised: 02/08/2014] [Accepted: 03/27/2014] [Indexed: 06/03/2023]
Abstract
Backgrounds. Research findings suggest that the mind can cause physical disease. To plan the best quality of care, general practitioner needs to understand an individual's health problems in physical, social, and psychological dimensions. This study sought to establish whether adverse life events occurring in childhood and adolescence are associated with diabetes. Methods. The cohort was collected from the health and social support (HeSSup) study-a postal follow-up survey of randomized working-aged Finns initiated in 1998. The response rate was 40.0% and the final cohort size 24057. Data on reimbursed diabetes medication during the years 1998-2006 were obtained from the Social Insurance Institute of Finland registers. Subjects were divided into insulin, tablet, combination therapy, and drug-naive groups together with a control group without diabetes. The prevalence of childhood adversities was assessed based on answers to six survey questions. Results. Childhood adversities showed predominant linkage to diabetes type 2 groups, especially to the combination therapy group requiring combined insulin and tablet treatment. No connection was found between childhood adversities and insulin use. Cumulative adversities did not markedly increase the association. Conclusions. Stressful events in childhood are associated with diabetes combination therapy in working age. The meaning of the relationship remains unsolved.
Collapse
Affiliation(s)
- Lauri Pisto
- Department of General Practice, Medical School, University of Tampere, Lääkärinkatu 1, 33520 Tampere, Finland
| | | | | | - Kari Mattila
- Department of General Practice, Medical School, University of Tampere, Lääkärinkatu 1, 33520 Tampere, Finland
- Centre for General Practice, Pirkanmaa Hospital District, Tampere, Finland
| |
Collapse
|
88
|
Abstract
The intra-uterine environment provides the first regulatory connection for the developing fetus and shapes its physiological responses in preparation for postnatal life. Psychological stress acts as a programming determinant by setting functional parameters to abnormal levels, thus inducing postnatal maladaptation. The effects of prenatal maternal stress (PNMS) on the developing immune system have been documented mostly through animal studies, but inconsistent results and methodological differences have hampered the complete understanding of these findings. As the immune system follows a similar ontogenic pattern in all mammals, a translational framework based on the developmental windows of vulnerability proposed by immunotoxicology studies was created to integrate these findings. The objective of this review is to examine the available literature on PNMS and immune function in the offspring through the above framework and gain a better understanding of these results by elucidating the moderating influence of the stressor type, timing and duration, and the offspring species, sex and age at assessment. The evaluation of the literature through this framework showed that the effects of PNMS are parameter specific: the moderating effects of timing in gestation were relevant for lymphocyte population numbers, Natural Killer cell function and mitogen-induced proliferation. The presence of an important and directional sexual dimorphism was evident and the influence of the type or duration of PNMS paralleled that of stress in non-pregnant animals. In conclusion, PNMS is a relevant factor in the programming of immune function. Its consequences may be related to disorders with an important immune component such as allergies.
Collapse
Affiliation(s)
- Franz Veru
- Department of Psychiatry, McGill University , Montreal, QC , Canada and
| | | | | | | |
Collapse
|
89
|
Dagley LF, Emili A, Purcell AW. Application of quantitative proteomics technologies to the biomarker discovery pipeline for multiple sclerosis. Proteomics Clin Appl 2014; 7:91-108. [PMID: 23112123 DOI: 10.1002/prca.201200104] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Revised: 10/04/2012] [Accepted: 10/11/2012] [Indexed: 11/08/2022]
Abstract
Multiple sclerosis is an inflammatory-mediated demyelinating disorder most prevalent in young Caucasian adults. The various clinical manifestations of the disease present several challenges in the clinic in terms of diagnosis, monitoring disease progression and response to treatment. Advances in MS-based proteomic technologies have revolutionized the field of biomarker research and paved the way for the identification and validation of disease-specific markers. This review focuses on the novel candidates discovered by the application of quantitative proteomics to relevant disease-affected tissues in both the human context and within the animal model of the disease known as experimental autoimmune encephalomyelitis. The role of targeted MS approaches for biomarker validation studies, such as multiple reaction monitoring will also be discussed.
Collapse
Affiliation(s)
- Laura F Dagley
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia
| | | | | |
Collapse
|
90
|
Sex-biased chromatin and regulatory cross-talk between sex chromosomes, autosomes, and mitochondria. Biol Sex Differ 2014; 5:2. [PMID: 24422881 PMCID: PMC3907150 DOI: 10.1186/2042-6410-5-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Accepted: 12/29/2013] [Indexed: 02/07/2023] Open
Abstract
Several autoimmune and neurological diseases exhibit a sex bias, but discerning the causes and mechanisms of these biases has been challenging. Sex differences begin to manifest themselves in early embryonic development, and gonadal differentiation further bifurcates the male and female phenotypes. Even at this early stage, however, there is evidence that males and females respond to environmental stimuli differently, and the divergent phenotypic responses may have consequences later in life. The effect of prenatal nutrient restriction illustrates this point, as adult women exposed to prenatal restrictions exhibited increased risk factors of cardiovascular disease, while men exposed to the same condition did not. Recent research has examined the roles of sex-specific genes, hormones, chromosomes, and the interactions among them in mediating sex-biased phenotypes. Such research has identified testosterone, for example, as a possible protective agent against autoimmune disorders and an XX chromosome complement as a susceptibility factor in murine models of lupus and multiple sclerosis. Sex-biased chromatin is an additional and likely important component. Research suggesting a role for X and Y chromosome heterochromatin in regulating epigenetic states of autosomes has highlighted unorthodox mechanisms of gene regulation. The crosstalk between the Y chromosomes and autosomes may be further mediated by the mitochondria. The organelles have solely maternal transmission and exert differential effects on males and females. Altogether, research supports the notion that the interaction between sex-biased elements might exert novel regulatory functions in the genome and contribute to sex-specific susceptibilities to autoimmune and neurological diseases.
Collapse
|
91
|
Shah K, McCormack CE, Bradbury NA. Do you know the sex of your cells? Am J Physiol Cell Physiol 2014; 306:C3-18. [PMID: 24196532 PMCID: PMC3919971 DOI: 10.1152/ajpcell.00281.2013] [Citation(s) in RCA: 143] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Accepted: 10/31/2013] [Indexed: 11/22/2022]
Abstract
Do you know the sex of your cells? Not a question that is frequently heard around the lab bench, yet thanks to recent research is probably one that should be asked. It is self-evident that cervical epithelial cells would be derived from female tissue and prostate cells from a male subject (exemplified by HeLa and LnCaP, respectively), yet beyond these obvious examples, it would be true to say that the sex of cell lines derived from non-reproductive tissue, such as lung, intestine, kidney, for example, is given minimal if any thought. After all, what possible impact could the presence of a Y chromosome have on the biochemistry and cell biology of tissues such as the exocrine pancreatic acini? Intriguingly, recent evidence has suggested that far from being irrelevant, genes expressed on the sex chromosomes can have a marked impact on the biology of such diverse tissues as neurons and renal cells. It is also policy of AJP-Cell Physiology that the source of all cells utilized (species, sex, etc.) should be clearly indicated when submitting an article for publication, an instruction that is rarely followed (http://www.the-aps.org/mm/Publications/Info-For-Authors/Composition). In this review we discuss recent data arguing that the sex of cells being used in experiments can impact the cell's biology, and we provide a table outlining the sex of cell lines that have appeared in AJP-Cell Physiology over the past decade.
Collapse
Affiliation(s)
- Kalpit Shah
- Department of Physiology and Biophysics, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois
| | | | | |
Collapse
|
92
|
Subcutaneous interferon β-1a may protect against cognitive impairment in patients with relapsing-remitting multiple sclerosis: 5-year follow-up of the COGIMUS study. PLoS One 2013; 8:e74111. [PMID: 24137499 PMCID: PMC3796707 DOI: 10.1371/journal.pone.0074111] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Accepted: 07/31/2013] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE To assess the effects of subcutaneous (sc) interferon (IFN) -1a on cognition over 5 years in mildly disabled patients with relapsing-remitting multiple sclerosis (RRMS). METHODS Patients aged 18-50 years with RRMS (Expanded Disability Status Scale score ≤4.0) who had completed the 3-year COGIMUS study underwent standardized magnetic resonance imaging, neurological examination, and neuropsychological testing at years 4 and 5. Predictors of cognitive impairment at year 5 were identified using multivariate analysis. RESULTS Of 331 patients who completed the 3-year COGIMUS study, 265 participated in the 2-year extension study, 201 of whom (75.8%; sc IFN β-1a three times weekly: 44 µg, n = 108; 22 µg, n = 93) completed 5 years' follow-up. The proportion of patients with cognitive impairment in the study population overall remained stable between baseline (18.0%) and year 5 (22.6%). The proportion of patients with cognitive impairment also remained stable in both treatment groups between baseline and year 5, and between year 3 and year 5. However, a significantly higher proportion of men than women had cognitive impairment at year 5 (26.5% vs 14.4%, p = 0.046). Treatment with the 22 versus 44 µg dose was predictive of cognitive impairment at year 5 (hazard ratio 0.68; 95% confidence interval 0.48-0.97). CONCLUSIONS This study suggests that sc IFN β-1a dose-dependently stabilizes or delays cognitive impairment over a 5-year period in most patients with mild RRMS. Women seem to be more protected against developing cognitive impairment, which may indicate greater response to therapy or the inherently better prognosis associated with female sex in MS.
Collapse
|
93
|
Fernández Liguori N, Rojas JI, Klajn DS, Ciapponi A. Intravenous immunoglobulin to prevent relapses during pregnancy and postpartum in multiple sclerosis. Hippokratia 2013. [DOI: 10.1002/14651858.cd010601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Nora Fernández Liguori
- Hospital Enrique Tornú; Multiple Sclerosis Clinic, Neurology Section; Combatiente de Malvinas 3002 Buenos Aires Argentina 1427
| | - Juan Ignacio Rojas
- Hospital Italiano Buenos Aires; Neurology Department; Gascon 450 Buenos Aires Buenos Aires Argentina 1411
| | - Diana S Klajn
- Hospital Tornu; Academic and Research Department; Combatientes de Malvinas 3002 Buenos Aires Argentina 1431
| | - Agustín Ciapponi
- Southern American Branch of the Iberoamerican Cochrane Centre; Argentine Cochrane Centre IECS, Institute for Clinical Effectiveness and Health Policy; Dr. Emilio Ravignani 2024 Buenos Aires Capital Federal Argentina C1414CPV - C1181ACH
| |
Collapse
|
94
|
Sankaran-Walters S, Macal M, Grishina I, Nagy L, Goulart L, Coolidge K, Li J, Fenton A, Williams T, Miller MK, Flamm J, Prindiville T, George M, Dandekar S. Sex differences matter in the gut: effect on mucosal immune activation and inflammation. Biol Sex Differ 2013; 4:10. [PMID: 23651648 PMCID: PMC3652739 DOI: 10.1186/2042-6410-4-10] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 04/11/2013] [Indexed: 01/11/2023] Open
Abstract
Background Women and men have diverse responses to many infectious diseases. These differences are amplified following menopause. However, despite extensive information regarding the effects of sex hormones on immune cells, our knowledge is limited regarding the effects of sex and gender on the function of the mucosal immune system. Sex differences also manifest in the prevalence of gut associated inflammatory and autoimmune disorders, including Crohn’s disease, ulcerative colitis and Celiac disease. It is thus hypothesized that a baseline sex-associated difference in immune activation may predispose women to inflammation-associated disease. Methods Peripheral blood samples and small intestinal biopsies were obtained from 34 healthy men and women. Immunophenotypic analysis of isolated lymphocytes was performed by flow cytometry. Oligonucleotide analysis was used to study the transcriptional profile in the gut mucosal microenvironment while real-time PCR analysis was utilized to identify differential gene expression in isolated CD4+ T cells. Transcriptional analysis was confirmed by protein expression levels for genes of interest using fluorescent immunohistochemistry. Data was analyzed using the GraphPad software package. Results Women had higher levels of immune activation and inflammation-associated gene expression in gut mucosal samples. CD4+ and CD8+ T cells had a significantly higher level of immune activation-associated phenotype in peripheral blood as well as in gut associated lymphoid tissue along with higher levels of proliferating T cells. CD4+ T cells that showed upregulation of IL1β as well as the TH17 pathway-associated genes contributed a large part of the inflammatory profile. Conclusion In this study, we demonstrated an upregulation in gene expression related to immune function in the gut microenvironment of women compared to men, in the absence of disease or pathology. Upon closer investigation, CD4+ T cell activation levels were higher in the LPLs in women than in men. Sex differences in the mucosal immune system may predispose women to inflammation-associated diseases that are exacerbated following menopause. Our study highlights the need for more detailed analysis of the effects of sex differences in immune responses at mucosal effector sites.
Collapse
Affiliation(s)
- Sumathi Sankaran-Walters
- Department of Medical Microbiology and Immunology, University of CA Davis Health System, 5605A GBSF, 451 Health Sciences Drive, Davis, CA 95616, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
95
|
Impact of microbes on autoimmune diseases. Arch Immunol Ther Exp (Warsz) 2013; 61:175-86. [PMID: 23417246 DOI: 10.1007/s00005-013-0216-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Accepted: 02/01/2013] [Indexed: 12/15/2022]
Abstract
Autoimmune and autoinflammatory diseases arise as a consequence of complex interactions of environmental factors with genetic traits. Although specific allelic variations cluster in predisposed individuals and promote the generation and/or expansion of autoreactive T and B lymphocytes, autoimmunity appears in various disease phenotypes and localizes to diverging tissues. Furthermore, the discovery that allelic variations within genes encoding components of the innate immune system drive self-reactive immune responses as well, led to the distinction of immune responses against host tissues into autoimmune and autoinflammatory diseases. In both categories of disorders, different pathogenic mechanisms and/or subsequent orders of tissue assaults may underlie the target cell specificity of the respective autoimmune attack. Furthermore, the transition from the initial tissue assault to the development of full-blown disease is likely driven by several factors. Thus, the development of specific forms of autoimmunity and autoinflammation reflects a multi-factorial process. The delineation of the specific factors involved in the pathogenic process is hampered by the fact that certain symptoms are assembled under the umbrella of a specific disease, although they might originate from diverging pathogenic pathways. These multi-factorial triggers and pathogenic pathways may also explain the inter-individual divergent courses and outcomes of diseases among humans. Here, we will discuss the impact of different environmental factors in general and microbial pathogens in particular on the regulation/expression of genes encoded within susceptibility alleles, and its consequences on subsequent autoimmune and/or autoinflammatory tissue damage utilizing primarily the chronic cholestatic liver disease primary biliary cirrhosis as model.
Collapse
|
96
|
Gomez FP, Steelman AJ, Young CR, Welsh CJ. Hormone and immune system interactions in demyelinating disease. Horm Behav 2013; 63:315-21. [PMID: 23137721 DOI: 10.1016/j.yhbeh.2012.10.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Revised: 10/25/2012] [Accepted: 10/29/2012] [Indexed: 11/17/2022]
Abstract
The immune, endocrine and nervous systems communicate with each other through a myriad of molecules including cytokines, hormones and neurotransmitters. Alterations in the balance of the products of these systems affect susceptibility to autoimmune disease and also the progression of disease. One of the most intensely studied autoimmune diseases is multiple sclerosis (MS). The purpose of this review is to explore the relationships between sex hormones and MS disease progression and to attempt to understand the paradox that although women are more likely to develop MS, female sex hormones appear to be beneficial in symptom amelioration. The proposed mechanisms of the therapeutic action of estrogens will be discussed with respect to T cell polarization and also on CNS cell populations. Investigations into the pathogenesis of multiple sclerosis (MS) and animal models of MS have given insights into the interactions between the neuroendocrine systems and provide important potential therapeutic venues that may be expanded to other autoimmune and neurodegenerative conditions.
Collapse
Affiliation(s)
- Francisco P Gomez
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843-4458, USA
| | | | | | | |
Collapse
|
97
|
Can decision-making skills affect responses to psychological stress in healthy women? Psychoneuroendocrinology 2012; 37:1912-21. [PMID: 22560372 DOI: 10.1016/j.psyneuen.2012.04.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Revised: 04/02/2012] [Accepted: 04/03/2012] [Indexed: 11/22/2022]
Abstract
In recent studies showing how stress can affect an individual's decision-making process, the cognitive component of decision-making could also be considered a coping resource available to individuals when faced with a stressful situation. The Iowa Gambling Task (IGT) constitutes the standard test for the assessment of decision-making skills under conditions of uncertainty. Responses of the hypothalamic-pituitary-adrenal (HPA) axis to psychosocial stress, in turn, have been estimated by means of cortisol measurements. Our main objective in this study was to test if good and bad IGT performers show distinct HPA axis responses, when challenged in a classic psychosocial stress test. Because women have been shown to outperform men on the IGT under the influence of psychosocial stress, we chose a sample of 40 women to take the IGT before they were exposed to a public speaking task in a virtual environment. The activation of the HPA axis, involved in the stress response, was assessed by examining the levels of cortisol in the subjects' saliva at the following four stages: before the challenge, after the challenge, and 10 and 20 min after the task. Participants were divided into two groups according to their level of performance, good or poor, on the IGT. Results showed statistically significant differences between the groups for pre-exposure cortisol levels and for cortisol levels 20 min after exposure. Overall cortisol levels were significantly higher in the group with poor performance on the IGT. It appears that good decision-making, which may be an important resource for coping with stress, is associated with a lower HPA axis response to a psychosocial stressor.
Collapse
|
98
|
Trune DR, Canlon B. Corticosteroid therapy for hearing and balance disorders. Anat Rec (Hoboken) 2012; 295:1928-43. [PMID: 23044978 PMCID: PMC3999710 DOI: 10.1002/ar.22576] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 07/24/2012] [Indexed: 12/20/2022]
Abstract
This review addresses the current status of steroid therapies for hearing and vestibular disorders and how certain misconceptions may be undermining the efficacy in restoring normal ear function, both experimentally and clinically. Specific misconceptions addressed are that steroid therapy is not effective, steroid-responsive hearing loss proves an underlying inflammatory problem in the ear, and steroids only have application to the hearing disorders listed below. Glucocorticoid therapy for hearing and balance disorders has been employed for over 60 years. It is recommended in cases of sudden hearing loss, Meniére's disease, immune-mediated hearing loss, and any vestibular dysfunction suspected of having an inflammatory etiology. The predominant steroids employed today are dexamethasone, prednisone, prednisolone, and methylprednisolone. Despite years of use, little is known of the steroid responsive mechanisms in the ear that are influenced by glucocorticoid therapy. Furthermore, meta-analyses and clinical study reviews occasionally question whether steroids offer any benefit at all. Foremost in the minds of clinicians is the immune suppression and anti-inflammatory functions of steroids because of their efficacy for autoimmune hearing loss. However, glucocorticoids have a strong binding affinity for the mineralocorticoid (aldosterone) and glucocorticoid receptors, both of which are prominent in the ear. Because the auditory and vestibular end organs require tightly regulated endolymph and perilymph fluids, this ion homeostasis role of the mineralocorticoid receptor cannot be overlooked in both normal and pathologic functions of the ear. The function of the glucocorticoid receptor is to provide anti-inflammatory and antiapoptotic signals by mediating survival factors.
Collapse
Affiliation(s)
- Dennis R Trune
- Oregon Hearing Research Center, Department of Otolaryngology/Head & Neck Surgery, Oregon Health & Science University, Portland, Oregon 97239-3098, USA.
| | | |
Collapse
|
99
|
Bakalov VK, Gutin L, Cheng CM, Zhou J, Sheth P, Shah K, Arepalli S, Vanderhoof V, Nelson LM, Bondy CA. Autoimmune disorders in women with turner syndrome and women with karyotypically normal primary ovarian insufficiency. J Autoimmun 2012; 38:315-21. [PMID: 22342295 PMCID: PMC3358475 DOI: 10.1016/j.jaut.2012.01.015] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Revised: 01/24/2012] [Accepted: 01/28/2012] [Indexed: 01/15/2023]
Abstract
The higher prevalence of autoimmune diseases in women compared to men could be due to effects of ovarian hormones, pregnancy and/or the presence of a second X chromosome. To elucidate the role of these factors, we investigated the prevalence and spectrum of autoimmune diagnoses in women with primary ovarian insufficiency associated with X chromosome monosomy (Turner syndrome, TS, n = 244) and women with karyotypically normal (46,XX) primary ovarian insufficiency (POI, n = 457) in a prospective study, conducted at the National Institutes of Health. We compared the study group prevalence to normative data for the U.S. population of women. Chronic lymphocytic (Hashimoto's) thyroiditis (HT) occurred in 37% of women with TS vs. 15% with POI (P < 0.0001); HT prevalence in both ovarian insufficiency groups significantly exceeded that in U.S. population of women (5.8%). Inflammatory bowel (IBD, 4%) and celiac disease (CD, 2.7%) were significantly increased in TS, but not in POI. No other autoimmune diagnosis, including Graves' disease or Type 1 diabetes appears to be significantly increased in either group. Women with TS had higher pro-inflammatory IL6 and TGF β1 levels (p < 0.0001 for both), and lower anti-inflammatory IL10 and TGF β2 levels (p < 0.005 for both) compared to POI and to normal volunteers. Lifetime estrogen exposure and parity were significantly lower in TS compared to POI, which were in turn lower than the general population of women. The finding that lymphocytic thyroiditis is greatly increased in both women with TS and POI suggests that factors associated with ovarian insufficiency per se promote this form of autoimmunity. The absence of a normal second X-chromosome further contributes to increased autoimmunity in TS.
Collapse
Affiliation(s)
- Vladimir K Bakalov
- Section on Epigenetics & Development, Program on Developmental Endocrinology and Genetics, National Institute of Child Health and Human Development, 10 Center Dr. CRC 1-3330; Bethesda, MD 20892-1103, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
100
|
Peroxisome proliferator-activated receptor (PPAR)α and -γ regulate IFNγ and IL-17A production by human T cells in a sex-specific way. Proc Natl Acad Sci U S A 2012; 109:9505-10. [PMID: 22647601 DOI: 10.1073/pnas.1118458109] [Citation(s) in RCA: 154] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Women develop certain autoimmune diseases more often than men. It has been hypothesized that this may relate to the development of more robust T-helper (Th)1 responses in women. To test whether women exhibit a Th1 bias, we isolated naïve cluster of differentiation (CD)4(+) T cells from peripheral blood of healthy women and men and measured the proliferation and cytokine production by these cells in response to submaximal amounts of anti-CD3 and anti-CD28. We observed that CD4(+) T cells from women produced higher levels of IFNγ as well as tended to proliferate more than male CD4(+) T cells. Intriguingly, male CD4(+) T cells instead had a predilection toward IL-17A production. This sex dichotomy in Th cytokine production was found to be even more striking in the Swiss/Jackson Laboratory (SJL) mouse. Studies in mice and humans indicated that the sexual dimorphism in Th1 and Th17 cytokine production was dependent on the androgen status and the T-cell expression of peroxisome proliferator activated receptor (PPAR)α and PPARγ. Androgens increased PPARα and decreased PPARγ expression by human CD4(+) T cells. PPARα siRNA-mediated knockdown had the effect of increasing IFNγ by male CD4(+) T cells, while transfection of CD4(+) T cells with PPARγ siRNAs increased IL-17A production uniquely by female T cells. Together, our observations indicate that human T cells exhibit a sex difference in the production of IFNγ and IL-17A that may be driven by expressions of PPARα and PPARγ.
Collapse
|