1
|
Rousselle D, Silveyra P. Acute Exposure to Ozone Affects Circulating Estradiol Levels and Gonadotropin Gene Expression in Female Mice. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2025; 22:222. [PMID: 40003448 PMCID: PMC11855596 DOI: 10.3390/ijerph22020222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 01/29/2025] [Accepted: 02/03/2025] [Indexed: 02/27/2025]
Abstract
Ozone, a critical air pollutant, has been shown to lead to systemic inflammation that can alter bodily functions, including hormone secretion, fertility, and the hypothalamic-pituitary-gonadal (HPG) axis. This study aimed to quantify changes in hormone production and follicle development after acute exposure to ozone using an animal model to identify the potential mechanisms underlying the observed effects of air pollution exposures on fertility and hormone secretion. To accomplish this, regularly cycling 8-week-old female C57BL/6J mice were exposed to 2 ppm of ozone or filtered air (control) for 3 h on the day of proestrus. Blood, ovaries, brain tissues, and pituitary glands were collected at 4 h after exposure to evaluate hormone levels, ovarian follicle distribution, and gene expression. Ovaries were also harvested at 24 h post-exposure. We found that at 4 h after ozone exposure, mice had significantly higher (30%) circulating estradiol levels than mice exposed to filtered air. This effect was accompanied by a decrease in mRNA expression of gonadotropin genes (LH, FSH) and gonadotropin-releasing hormone in the pituitary gland. Analysis of ovarian tissue at 4 h and 24 h after exposure showed no significant changes in follicle composition or the expression of steroidogenesis genes. We conclude that acute ozone exposure affects sex hormone levels and disrupts the HPG axis. Future studies addressing chronic or long-term effects of air pollution exposure are needed to elucidate the mechanisms by which ambient ozone affects endocrine function.
Collapse
Affiliation(s)
- Dustin Rousselle
- Department of Environmental and Occupational Health, School of Public Health, Indiana University, Bloomington, IN 47405, USA;
| | - Patricia Silveyra
- Department of Environmental and Occupational Health, School of Public Health, Indiana University, Bloomington, IN 47405, USA;
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
2
|
Duarte-Silva M, Oliveira CNS, Fuzo C, Silva-Neto PV, Toro DM, Pimentel VE, Pérez MM, Fraga-Silva TFC, Carvalho JCS, Neto FMS, Júnior RBM, Arruda E, Vilar FC, Degiovani AM, Ostini FM, Feitosa MR, Parra RS, Gaspar GG, Rocha JJR, Feres O, Fernandes APM, Maruyama SR, Russo EMS, Bonato VLD, Santos IKFM, Sorgi CA, Dias-Baruffi M, Faccioli LH, Cardoso CRB. Divergent androgenic modulation of SARS-CoV-2 infection cooperates with dysregulated immune response to dictate worse COVID-19 outcomes in men. Brain Behav Immun 2023; 114:275-286. [PMID: 37648004 DOI: 10.1016/j.bbi.2023.08.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 08/22/2023] [Accepted: 08/26/2023] [Indexed: 09/01/2023] Open
Abstract
BACKGROUND Sex-determined differences are rarely addressed in the management of diseases, despite well-known contrasting outcomes between female and male patients. In COVID-19 there is a remarkable disparity, with higher rates of mortality and more severe acute disease in men compared to women, who are mostly affected by long COVID-19. Furthermore, whether androgens play a protective or detrimental role in COVID-19 is still a matter of debate. Hence, the adequate management of the disease, especially regarding men presenting acute disease aggravation, still needs important data to elucidate the interplay between sex hormones and host immune responses that drive the worse evolution in male patients. METHODS A cohort of 92 controls and 198 non-severe and severe COVID-19 patients, from both sexes, was assessed for clinical outcomes, plasma steroids, gonadotropins, sex hormone binding globulin (SHBG) and immune mediators, before vaccination. These data were correlated with the global gene expression of blood leukocytes. The androgen receptor (AR) signaling pathway was investigated by transcriptomics and tracheal aspirate was obtained from severe patients for SARS-COV-2 quantification in the respiratory tract. The interplay among clinical, endocrine and immunological data deciphered the sex differences in COVID-19. Importantly, statistical analyses, using 95% confidence interval, considered confounding factors such as age and comorbidities, to definitely parse the role of androgens in the disease outcome. RESULTS There were notable contrasting levels of testosterone and dihydrotestosterone (DHT) throughout the disease course in male but not female patients. Inflammatory mediators presented significant negative correlations with testosterone, which was partially dependent on age and diabetes in men. Male subjects with severe COVID-19 had a significant up regulation of the AR signaling pathway, including modulation of TMPRSS2 and SRD5A1 genes, which are related to the viral infection and DHT production. Indeed, men had a higher viral load in the tracheal aspirate and levels of DHT were associated with increased relative risk of death. In contrast, the testosterone hormone, which was notably reduced in severe disease, was significantly related with susceptibility to COVID-19 worsening in male patients. Secondary hypogonadism was ruled out in the male severe COVID-19 subjects, as FSH, LH, and SHBG levels were not significantly altered. Instead, these subjects tended to have increased gonadotropin levels. Most interestingly, in this study we identified, for the first time, combined sets of clinical and immunoendocrine parameters that together predicted progression from non-severe to severe COVID-19 in men. One of the limitations of our study was the low or undetectable levels of DHT in many patients. Then, the evaluation of enzymes related to biosynthesis and signaling by androgens was mandatory and reiterated our findings. CONCLUSIONS These original results unraveled the disease immunoendocrine regulation, despite vaccination or comorbidities and pointed to the fundamental divergent role of the androgens testosterone and DHT in the determination of COVID-19 outcomes in men. Therefore, sex-specific management of the dysregulated responses, treatments or public health measures should be considered for the control of COVID-19 pandemic.
Collapse
Affiliation(s)
- Murillo Duarte-Silva
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto - FCFRP/USP, Brazil; Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto - FMRP/USP, Brazil
| | - Camilla N S Oliveira
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto - FCFRP/USP, Brazil; Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto - FMRP/USP, Brazil
| | - Carlos Fuzo
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto - FCFRP/USP, Brazil
| | - Pedro V Silva-Neto
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto - FCFRP/USP, Brazil; Instituto de Ciências Biológicas Amazonas, Universidade Federal do Amazonas - UFAM, Brazil
| | - Diana M Toro
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto - FCFRP/USP, Brazil; Instituto de Ciências Biológicas Amazonas, Universidade Federal do Amazonas - UFAM, Brazil
| | - Vinícius E Pimentel
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto - FCFRP/USP, Brazil; Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto - FMRP/USP, Brazil
| | - Malena M Pérez
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto - FCFRP/USP, Brazil
| | - Thais F C Fraga-Silva
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto - FMRP/USP, Brazil
| | - Jonatan C S Carvalho
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto - FCFRP/USP, Brazil; Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto - FFCLRP/USP, Brazil
| | - Firmino M S Neto
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto - FCFRP/USP, Brazil
| | - Ronaldo B M Júnior
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto - FCFRP/USP, Brazil; Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto - FMRP/USP, Brazil
| | - Eurico Arruda
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto - FMRP/USP, Brazil
| | - Fernando C Vilar
- Departamento de Clínica Médica, Faculdade de Medicina de Ribeirão Preto - FMRP/USP, Brazil
| | | | - Fátima M Ostini
- Hospital Santa Casa de Misericórdia de Ribeirão Preto, Brazil
| | - Marley R Feitosa
- Departamento de Cirurgia e Anatomia, Faculdade de Medicina de Ribeirão Preto - FMRP/USP, Brazil
| | - Rogerio S Parra
- Departamento de Cirurgia e Anatomia, Faculdade de Medicina de Ribeirão Preto - FMRP/USP, Brazil
| | - Gilberto G Gaspar
- Departamento de Clínica Médica, Faculdade de Medicina de Ribeirão Preto - FMRP/USP, Brazil
| | - José J R Rocha
- Departamento de Cirurgia e Anatomia, Faculdade de Medicina de Ribeirão Preto - FMRP/USP, Brazil
| | - Omar Feres
- Departamento de Cirurgia e Anatomia, Faculdade de Medicina de Ribeirão Preto - FMRP/USP, Brazil
| | | | - Sandra R Maruyama
- Departamento de Genética e Evolução, Centro de Ciências Biológicas e da Saúde, Universidade Federal de São Carlos - UFSCar, Brazil
| | - Elisa M S Russo
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto - FCFRP/USP, Brazil
| | - Vânia L D Bonato
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto - FMRP/USP, Brazil
| | - Isabel K F M Santos
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto - FMRP/USP, Brazil
| | - Carlos A Sorgi
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto - FMRP/USP, Brazil; Instituto de Ciências Biológicas Amazonas, Universidade Federal do Amazonas - UFAM, Brazil; Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto - FFCLRP/USP, Brazil
| | - Marcelo Dias-Baruffi
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto - FCFRP/USP, Brazil
| | - Lúcia H Faccioli
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto - FCFRP/USP, Brazil
| | - Cristina R B Cardoso
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto - FCFRP/USP, Brazil.
| |
Collapse
|
3
|
Gu X, Li SY, Matsuyama S, DeFalco T. Immune Cells as Critical Regulators of Steroidogenesis in the Testis and Beyond. Front Endocrinol (Lausanne) 2022; 13:894437. [PMID: 35573990 PMCID: PMC9096076 DOI: 10.3389/fendo.2022.894437] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 03/31/2022] [Indexed: 12/01/2022] Open
Abstract
Steroidogenesis is an essential biological process for embryonic development, reproduction, and adult health. While specific glandular cells, such as Leydig cells in the testis, are traditionally known to be the principal players in steroid hormone production, there are other cell types that contribute to the process of steroidogenesis. In particular, immune cells are often an important component of the cellular niche that is required for the production of steroid hormones. For several decades, studies have reported that testicular macrophages and Leydig cells are intimately associated and exhibit a dependency on the other cell type for their proper development; however, the mechanisms that underlie the functional relationship between macrophages and Leydig cells are unclear. Beyond the testis, in certain instances immune cells themselves, such as certain types of lymphocytes, are capable of steroid hormone production, thus highlighting the complexity and diversity that underlie steroidogenesis. In this review we will describe how immune cells are critical regulators of steroidogenesis in the testis and in extra-glandular locations, as well as discuss how this area of research offers opportunities to uncover new insights into steroid hormone production.
Collapse
Affiliation(s)
- Xiaowei Gu
- Division of Reproductive Sciences, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Shu-Yun Li
- Division of Reproductive Sciences, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Satoko Matsuyama
- Division of Reproductive Sciences, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Tony DeFalco
- Division of Reproductive Sciences, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
4
|
Chakraborty S, Pramanik J, Mahata B. Revisiting steroidogenesis and its role in immune regulation with the advanced tools and technologies. Genes Immun 2021; 22:125-140. [PMID: 34127827 PMCID: PMC8277576 DOI: 10.1038/s41435-021-00139-3] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 05/03/2021] [Accepted: 05/21/2021] [Indexed: 12/19/2022]
Abstract
Historically tools and technologies facilitated scientific discoveries. Steroid hormone research is not an exception. Unfortunately, the dramatic advancement of the field faded this research area and flagged it as a solved topic. However, it should have been the opposite. The area should glitter with its strong foundation and attract next-generation scientists. Over the past century, a myriad of new facts on biochemistry, molecular biology, cell biology, physiology and pathology of the steroid hormones was discovered. Several innovations were made and translated into life-saving treatment strategies such as synthetic steroids, and inhibitors of steroidogenesis and steroid signaling. Steroid molecules exhibit their diverse effects on cell metabolism, salt and water balance, development and function of the reproductive system, pregnancy, and immune-cell function. Despite vigorous research, the molecular basis of the immunomodulatory effect of steroids is still mysterious. The recent excitement on local extra-glandular steroidogenesis in regulating inflammation and immunity is revitalizing the topic with a new perspective. Therefore, here we review the role of steroidogenesis in regulating inflammation and immunity, discuss the unresolved questions, and how this area can bring another golden age of steroid hormone research with the development of new tools and technologies and advancement of the scientific methods.
Collapse
Affiliation(s)
| | - Jhuma Pramanik
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Bidesh Mahata
- Department of Pathology, University of Cambridge, Cambridge, UK.
| |
Collapse
|
5
|
A Novel Method to Differentiate Tonsil-Derived Mesenchymal Stem Cells In Vitro into Estrogen-Secreting Cells. Tissue Eng Regen Med 2020; 18:253-264. [PMID: 33113109 DOI: 10.1007/s13770-020-00307-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 09/24/2020] [Accepted: 09/29/2020] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND The advantages of tonsil-derived mesenchymal stem cells (TMSCs) over other mesenchymal stem cells (MSCs) include higher proliferation rates, various differentiation potentials, efficient immune-modulating capacity, and ease of obtainment. Specifically, TMSCs have been shown to differentiate into the endodermal lineage. Estrogen deficiency is a major cause of postmenopausal osteoporosis and is associated with higher incidences of ischemic heart disease and cerebrovascular attacks during the postmenopausal period. Therefore, stem cell-derived, estrogen-secreting cells might be used for estrogen deficiency. METHODS Here, we developed a novel method that utilizes retinoic acid, insulin-like growth factor-1, basic fibroblast growth factor, and dexamethasone to evaluate the differentiating potential of TMSCs into estrogen-secreting cells. The efficacy of the novel differentiating method for generation of estrogen-secreting cells was also evaluated with bone marrow- and adipose tissue-derived MSCs. RESULTS Incubating TMSCs in differentiating media induced the gene expression of cytochrome P450 19A1 (CYP19A1), which plays a key role in estrogen biosynthesis, and increased 17β-estradiol secretion upon testosterone addition. Furthermore, CYP11A1, CYP17A1, and 3β-hydroxysteroid dehydrogenase type-1 gene expression levels were significantly increased in TMSCs. In bone marrow-derived and adipose tissue-derived MSCs, this differentiation method also induced the gene expression of CYP19A1, but not CYP17A1, suggesting TMSCs are a superior source for estrogen secretion. CONCLUSION These results imply that TMSCs can differentiate into functional estrogen-secreting cells, thus providing a novel, alternative cell therapy for estrogen deficiency.
Collapse
|
6
|
Tan SY, Lei X, Little HC, Rodriguez S, Sarver DC, Cao X, Wong GW. CTRP12 ablation differentially affects energy expenditure, body weight, and insulin sensitivity in male and female mice. Am J Physiol Endocrinol Metab 2020; 319:E146-E162. [PMID: 32421370 PMCID: PMC7468785 DOI: 10.1152/ajpendo.00533.2019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Secreted hormones facilitate tissue cross talk to maintain energy balance. We previously described C1q/TNF-related protein 12 (CTRP12) as a novel metabolic hormone. Gain-of-function and partial-deficiency mouse models have highlighted important roles for this fat-derived adipokine in modulating systemic metabolism. Whether CTRP12 is essential and required for metabolic homeostasis is unknown. We show here that homozygous deletion of Ctrp12 gene results in sexually dimorphic phenotypes. Under basal conditions, complete loss of CTRP12 had little impact on male mice, whereas it decreased body weight (driven by reduced lean mass and liver weight) and improved insulin sensitivity in female mice. When challenged with a high-fat diet, Ctrp12 knockout (KO) male mice had decreased energy expenditure, increased weight gain and adiposity, elevated serum TNFα level, and reduced insulin sensitivity. In contrast, female KO mice had reduced weight gain and liver weight. The expression of lipid synthesis and catabolism genes, as well as profibrotic, endoplasmic reticulum stress, and oxidative stress genes were largely unaffected in the adipose tissue of Ctrp12 KO male mice. Despite greater adiposity and insulin resistance, Ctrp12 KO male mice fed an obesogenic diet had lower circulating triglyceride and free fatty acid levels. In contrast, lipid profiles of the leaner female KO mice were not different from those of WT controls. These data suggest that CTRP12 contributes to whole body energy metabolism in genotype-, diet-, and sex-dependent manners, underscoring complex gene-environment interactions influencing metabolic outcomes.
Collapse
Affiliation(s)
- Stefanie Y Tan
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Xia Lei
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Hannah C Little
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Susana Rodriguez
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Dylan C Sarver
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Xi Cao
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - G William Wong
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
7
|
Expression and activation of the steroidogenic enzyme CYP11A1 is associated with IL-13 production in T cells from peanut allergic children. PLoS One 2020; 15:e0233563. [PMID: 32497050 PMCID: PMC7272076 DOI: 10.1371/journal.pone.0233563] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 05/07/2020] [Indexed: 11/19/2022] Open
Abstract
Activation of the steroidogenic enzyme CYP11A1 was shown to be necessary for the development of peanut-induced intestinal anaphylaxis and IL-13 production in allergic mice. We determined if levels of CYP11A1 in peripheral blood T cells from peanut-allergic (PA) children compared to non-allergic controls were increased and if levels correlated to IL-13 production and oral challenge outcomes to peanut. CYP11A1 mRNA and protein levels were significantly increased in activated CD4+ T cells from PA patients. In parallel, IL-13 production was significantly increased; IFNγ levels were not different between groups. There were significant correlations between expression levels of CYP11A1 mRNA and levels of IL13 mRNA and protein, levels of serum IgE anti-Ara h 2 and to outcomes of peanut challenge. The importance of CYP11A1 on cytokine production was tested using a CYP11A1 CRISPR/Cas9 KO plasmid or an inhibitor of enzymatic CYP11A1 activity. Inhibition of CYP11A1 activation in patient cells treated with the inhibitor, aminoglutethimide, or CD4+ T cell line transfected with the CYP11A1 KO plasmid resulted in reduced IL-13 production. These data suggest that the CYP11A1-CD4+Tcell-IL-13 axis in activated CD4+ T cells from PA children is associated with development of PA reactions. CYP11A1 may represent a novel target for therapeutic intervention in PA children.
Collapse
|
8
|
Aromatase deficiency in hematopoietic cells improves glucose tolerance in male mice through skeletal muscle-specific effects. PLoS One 2020; 15:e0227830. [PMID: 31971970 PMCID: PMC6977739 DOI: 10.1371/journal.pone.0227830] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 12/30/2019] [Indexed: 12/14/2022] Open
Abstract
Estrogens are important for maintaining metabolic health in males. However, the key sources of local estrogen production for regulating energy metabolism have not been fully defined. Immune cells exhibit aromatase activity and are resident in metabolic tissues. To determine the relative contribution of immune cell-derived estrogens for metabolic health in males, C57BL6/J mice underwent bone marrow transplant with marrow from either wild-type (WT(WT)) or aromatase-deficient (WT(ArKO)) donors. Body weight, body composition, and glucose and insulin tolerance were assessed over 24 weeks with mice maintained on a regular chow diet. No differences were found in insulin sensitivity between groups, but WT(ArKO) mice were more glucose tolerant than WT(WT) mice 20 weeks after transplant, suggestive of enhanced glucose disposal (AUCglucose 6061±3349 in WT(WT) mice versus 3406±1367 in WT(ArKO) mice, p = 0.01). Consistent with this, skeletal muscle from WT(ArKO) mice showed higher expression of the mitochondrial genes Ppargc1a (p = 0.03) and Nrf1 (p = 0.01), as well as glucose transporter type 4 (GLUT4, Scl2a4; p = 0.02). Skeletal muscle from WT(ArKO) mice had a lower concentration of 17β-estradiol (5489±2189 pg/gm in WT(WT) mice versus 3836±2160 pg/gm in WT(ArKO) mice, p = 0.08) but higher expression of estrogen receptor-α (ERα, Esr1), raising the possibility that aromatase deficiency in immune cells led to a compensatory increase in ERα signaling. No differences between groups were found with regard to body weight, adiposity, or gene expression within adipose tissue or liver. Immune cells are a key source of local 17β-estradiol production and contribute to metabolic regulation in males, particularly within skeletal muscle. The respective intracrine and paracrine roles of immune cell-derived estrogens require further delineation, as do the pathways that regulate aromatase activity in immune cells specifically within metabolic tissues.
Collapse
|
9
|
Vitamin D and Demyelinating Diseases: Neuromyelitis Optica (NMO) and Multiple Sclerosis (MS). Autoimmune Dis 2020; 2020:8718736. [PMID: 32373353 PMCID: PMC7187724 DOI: 10.1155/2020/8718736] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 11/30/2019] [Indexed: 12/18/2022] Open
Abstract
Vitamin D deficiency is prevalent in all ages regardless of climate or geographical location and evidence is emerging that the incidence of autoimmune diseases is increasing worldwide. Women make up a large proportion of autoimmune disease diagnoses, underscoring the importance of fully elucidating the complex synergistic relationships between estrogens and vitamin D. Vitamin D receptor-activating drugs appear to enhance remyelination in patients diagnosed with multiple sclerosis (MS) and other demyelinating diseases such as neuromyelitis optica (NMO). This review is intended to update health practitioners about the potential role of vitamin D deficiency demyelination and to motivate future research on dietary recommendations for vitamin D in preventing and treating demyel1nating diseases.
Collapse
|
10
|
Park CJ, Kim H, Jin J, Barakat R, Lin PC, Choi JM, Ko CJ. Porcine intestinal lymphoid tissues synthesize estradiol. J Vet Sci 2018; 19:477-482. [PMID: 29486537 PMCID: PMC6070586 DOI: 10.4142/jvs.2018.19.4.477] [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: 11/29/2017] [Revised: 02/05/2018] [Accepted: 02/10/2018] [Indexed: 12/27/2022] Open
Abstract
Estradiol (17β-estradiol) is synthesized primarily in the gonads of both sexes and regulates the development and function of reproductive organs. Recently, we reported that intestinal lymphocyte homeostasis is regulated by estradiol synthesized de novo in the endothelial cells of the high endothelial venules (HEVs) of mesenteric lymph nodes and Peyer's patches in mice. This observation prompted us to hypothesize that HEVs of intestinal lymphoid tissues are sites of estradiol synthesis across species. In this study, we examined whether estradiol is synthesized in the intestinal lymphoid tissues of adolescent piglets. Comparisons of estradiol levels in blood and tissue showed that estradiol concentrations in mesenteric lymph nodes and Peyer's patches were significantly higher than the level in serum. Reverse transcription polymerase chain reaction showed that porcine intestinal lymphoid tissues express mRNAs for steroidogenic enzymes (StAR, 17β-Hsd,3β-Hsd, Cyp17a1, and Cyp19a1), and immunohistochemical results in ilial tissue showed expression of aromatase (CYP19) in Peyer's patch-localized endothelial cells of HEVs. When mesenteric lymph node and Peyer's patch tissues were cultured in vitro, they produced estradiol. Taken together, the results indicate that mesenteric lymph nodes and Peyer's patches are sites of estradiol synthesis in adolescent piglets.
Collapse
Affiliation(s)
- Chan Jin Park
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois, Urbana-Campaign, IL 61802, USA
| | - Heehyen Kim
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois, Urbana-Campaign, IL 61802, USA.,Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Korea
| | - Jooyoung Jin
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois, Urbana-Campaign, IL 61802, USA.,Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Korea
| | - Radwa Barakat
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois, Urbana-Campaign, IL 61802, USA.,Department of Toxicology, Faculty of Veterinary Medicine, Benha University, Benha 13518, Egypt
| | - Po-Ching Lin
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois, Urbana-Campaign, IL 61802, USA
| | - Jeong Moon Choi
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois, Urbana-Campaign, IL 61802, USA
| | - CheMyong Jay Ko
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois, Urbana-Campaign, IL 61802, USA
| |
Collapse
|
11
|
Rubinow KB. An intracrine view of sex steroids, immunity, and metabolic regulation. Mol Metab 2018; 15:92-103. [PMID: 29551633 PMCID: PMC6066741 DOI: 10.1016/j.molmet.2018.03.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 02/26/2018] [Accepted: 03/01/2018] [Indexed: 12/21/2022] Open
Abstract
Background Over the past two decades, parallel recognition has grown of the importance of both sex steroids and immune activity in metabolic regulation. More recently, these discrete areas have been integrated in studies examining the metabolic effects of sex steroid immunomodulation. Implicit in these studies has been a traditional, endocrine model of sex steroid delivery from the gonads to target cells, including immune cells. Thus, research to date has focused on the metabolic effects of sex steroid receptor signaling in immune cells. This endocrine model, however, overlooks the extensive capacity of immune cells to generate and metabolize sex steroids, enabling the production of sex steroids for intracrine signaling – that is, sex steroid production for signaling within the cell of origin. Intracrine function allows highly cell-autonomous regulation of sex steroid exposure, and sex steroid secretion by immune cells could confer paracrine signaling effects in neighboring cells within metabolic tissues. In this review, immune cell intracrinology will denote sex steroid production within immune cells for either intracrine or paracrine signaling. This intracrine capacity of immune cells has been well established, and prior work has supported its importance in autoimmune disorders, trauma, and cancer. The potential relevance of immune cell intracrine function to the regulation of energy balance, body weight, body composition, and insulin sensitivity has yet to be explored. Scope of review The following review will detail findings to date regarding the steroidogenic and steroid metabolizing capacity of immune cells, the regulation of immune cell intracrine function, and the biological effects of immune-derived sex steroids, including the clinical relevance of immune cell intracrinology in fields other than metabolism. These findings will serve as the basis for a proposed model of immune cell intracrinology constituting a new frontier in metabolism research. Major conclusions The development of highly sensitive mass spectrometric methods for sex steroid measurement and quantitation of metabolic flux now allows unprecedented ability to interrogate sex steroid production, metabolism and secretion by immune cells. Immune cell intracrinology could reveal key mechanisms underlying immune cell-mediated metabolic regulation. Sex steroids exert immunomodulatory effects that may influence metabolic health. Immune cells can synthesize, modify, and metabolize sex steroids. Immune cell-derived sex steroids may play intracrine, autocrine, paracrine, and possibly even endocrine roles. Immune cell steroidogenesis is a largely unexplored area of metabolism research.
Collapse
Affiliation(s)
- Katya B Rubinow
- Diabetes Institute, Department of Medicine, University of Washington, School of Medicine, 850 Republican St., Box 358055, Seattle, WA 98109, USA.
| |
Collapse
|
12
|
Barakat R, Oakley O, Kim H, Jin J, Ko CJ. Extra-gonadal sites of estrogen biosynthesis and function. BMB Rep 2017; 49:488-96. [PMID: 27530684 PMCID: PMC5227141 DOI: 10.5483/bmbrep.2016.49.9.141] [Citation(s) in RCA: 131] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Indexed: 12/23/2022] Open
Abstract
Estrogens are the key hormones regulating the development and function of reproductive organs in all vertebrates. Recent evidence indicates that estrogens play important roles in the immune system, cancer development, and other critical biological processes related to human well-being. Obviously, the gonads (ovary and testis) are the primary sites of estrogen synthesis, but estrogens synthesized in extra- gonadal sites play an equally important role in controlling biological activities. Understanding non-gonadal sites of estrogen synthesis and function is crucial and will lead to therapeutic interventions targeting estrogen signaling in disease prevention and treatment. Developing a rationale targeting strategy remains challenging because knowledge of extra-gonadal biosynthesis of estrogens, and the mechanism by which estrogen activity is exerted, is very limited. In this review, we will summarize recent discoveries of extra-gonadal sites of estrogen biosynthesis and their local functions and discuss the significance of the most recent novel discovery of intestinal estrogen biosynthesis. [BMB Reports 2016; 49(9): 488-496]
Collapse
Affiliation(s)
- Radwa Barakat
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Illinois 61802, Unites States; Department of Toxicology, Faculty of Veterinary Medicine, Benha University, Benha 13518, Egypt
| | - Oliver Oakley
- Department of Biology, College of Arts and Sciences, Eastern Kentucky University, Kentucky 40475, United States
| | - Heehyen Kim
- Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Korea
| | - Jooyoung Jin
- Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Korea
| | - CheMyong Jay Ko
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Illinois 61802, Unites States
| |
Collapse
|
13
|
Spanier JA, Nashold FE, Mayne CG, Nelson CD, Hayes CE. Vitamin D and estrogen synergy in Vdr-expressing CD4+ T cells is essential to induce Helios+FoxP3+ T cells and prevent autoimmune demyelinating disease. J Neuroimmunol 2015; 286:48-58. [DOI: 10.1016/j.jneuroim.2015.06.015] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 06/16/2015] [Accepted: 06/19/2015] [Indexed: 01/08/2023]
|
14
|
Albertsmeier M, Pratschke S, Chaudry I, Angele MK. Gender-Specific Effects on Immune Response and Cardiac Function after Trauma Hemorrhage and Sepsis. VISZERALMEDIZIN 2015; 30:91-6. [PMID: 26288583 PMCID: PMC4513799 DOI: 10.1159/000360149] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Background Studies in human as well as animal models indicate a gender-specific responsiveness of the immune and organ systems with regard to shock, trauma, and sepsis. Methods A literature review was performed. Results Cell-mediated immune responses and cardiovascular functions are suppressed in males following trauma hemorrhage, whereas they are maintained or even enhanced in females in the proestrus state of the estrus cycle. Experimental studies have demonstrated that divergent immune responses in males and females following adverse circulatory conditions are mediated by the gender-specific hormones testosterone and estrogen. Several clinical trials, however, failed to demonstrate a significant association of gender and inflammatory response. This may be explained by the heterogeneity of the population in terms of their hormonal status at the time of injury. Conclusions With regard to the underlying mechanisms, receptors for sex hormones have been identified on various immune cells, suggesting direct effects of these hormones on immune function. Alternatively, indirect effects of sex steroids such as changes in cardiovascular responses or androgen- and estrogen-synthesizing enzymes might contribute to gender-specific immune responses. Clinical studies suggest that sex hormones, such as dehydroepiandrosterone, modulate the function of peripheral blood mononuclear cells also following abdominal surgery. Thus, sex hormones, receptor antagonists, and sex steroid-synthesizing enzymes might be useful in the future for modulating the complex immune responses after trauma hemorrhage and sepsis.
Collapse
Affiliation(s)
- Markus Albertsmeier
- Department of General, Visceral, Transplantation, Vascular and Thoracic Surgery, Ludwig Maximilian University, Munich, Germany
| | - Sebastian Pratschke
- Department of General, Visceral, Transplantation, Vascular and Thoracic Surgery, Ludwig Maximilian University, Munich, Germany
| | - Irshad Chaudry
- Center for Surgical Research and Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Martin K Angele
- Department of General, Visceral, Transplantation, Vascular and Thoracic Surgery, Ludwig Maximilian University, Munich, Germany
| |
Collapse
|
15
|
Weniger M, D'Haese JG, Angele MK, Chaudry IH. Potential therapeutic targets for sepsis in women. Expert Opin Ther Targets 2015; 19:1531-43. [PMID: 26083575 DOI: 10.1517/14728222.2015.1057570] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
INTRODUCTION Gender is increasingly recognized as a key factor in trauma and sepsis. Multiple clinical and experimental studies on sepsis have shown a distinct advantage of females in the proestrus cycle to survive sepsis compared with age-matched males. In addition, estrogen treatment is beneficial in non-proestrus cycles and also in ovarectomized females. In this manuscript, the effects of gender and sex hormones in sepsis are summarized and potential gender-specific therapeutic strategies in women are evaluated. AREAS COVERED This review comprises current clinical studies on the effect of gender in sepsis and gives an overview on gender and sex hormone-related effects on immune cells and organ function. Based on clinical and experimental data, potential therapeutic targets are presented. EXPERT OPINION Estrogens and estrogen-receptor agonists have been extensively shown to be beneficial in the setting of sepsis. Clinical data, however, do not clearly support their therapeutic use. This discrepancy appears to be mainly due to insufficient study design in clinical trials conducted up to now. Therefore, improved study protocols with exact analysis of the patients' hormonal status are needed to clarify the role of gender and sex hormones in trauma and sepsis.
Collapse
Affiliation(s)
- Maximilian Weniger
- a 1 Ludwig Maximilians-University, Department of General, Visceral, Transplantation, Vascular and Thoracic Surgery, Campus Grosshadern , Munich, Germany
| | - Jan G D'Haese
- b 2 Ludwig Maximilians-University, Department of General, Visceral, Transplantation, Vascular and Thoracic Surgery, Campus Grosshadern , Munich, Germany
| | - Martin K Angele
- c 3 Ludwig Maximilians-University, Department of General, Visceral, Transplantation, Vascular and Thoracic Surgery, Campus Grosshadern , Munich, Germany
| | - Irshad H Chaudry
- d 4 University of Alabama at Birmingham, Center for Surgical Research and Department of Surgery , G094 Volker Hall, 1670 University Boulevard, Birmingham, AL 35294, USA +1 205 975 2195 ; +1 205 975 9719 ;
| |
Collapse
|
16
|
Arsenović-Ranin N, Kosec D, Pilipović I, Bufan B, Stojić-Vukanić Z, Radojević K, Nacka-Aleksić M, Leposavić G. Androgens contribute to age-associated changes in peripheral T-cell homeostasis acting in a thymus-independent way. Neuroimmunomodulation 2014; 21:161-82. [PMID: 24504059 DOI: 10.1159/000355349] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Accepted: 08/26/2013] [Indexed: 01/09/2023] Open
Abstract
OBJECTIVE Considering a causal role of androgens in thymic involution, age-related remodeling of peripheral T-cell compartments in the absence of testicular hormones was evaluated. METHODS Rats were orchidectomized (ORX) at the age of 1 month, and T-peripheral blood lymphocytes (PBLs) and splenocytes from young (75-day-old) and aged (24-month-old) rats were examined for differentiation/activation and immunoregulatory marker expression. RESULTS In ORX rats, following the initial rise, the counts of CD4+ and CD8+ PBLs diminished with aging. This reflected the decline in thymic export as shown by recent thymic emigrant (RTE) enumeration. Orchidectomy increased the count of both of the major T-splenocyte subsets in young rats, and they (differently from controls) remained stable with aging. The CD4+:CD8+ T-splenocyte ratio in ORX rats shifted towards CD4+ cells compared to age-matched controls. Although in the major T-cell subsets in the blood and spleen from aged ORX rats the numbers of RTEs were comparable to the corresponding values in age-matched controls, the numbers of mature naïve and memory/activated cells substantially differed. Compared with age-matched controls, in aged ORX rats the numbers of CD4+ mature naïve PBLs and splenocytes were reduced, whereas those of CD4+ memory/activated cells (predictive of early mortality) were increased. Additionally, in spleens from aged ORX rats, despite unaltered thymic export, CD4+CD25+FoxP3+ and natural killer T cell counts were greater than in age-matched controls. CONCLUSION (i) Age-related decline in thymopoietic efficacy is not dependent on androgen presence, and (ii) androgens are involved in the maintenance of peripheral T-cell (particularly CD4+ cell) homeostasis during aging.
Collapse
Affiliation(s)
- Nevena Arsenović-Ranin
- Department of Microbiology and Immunology, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Steroidogenic enzyme Cyp11a1 regulates Type 2 CD8+ T cell skewing in allergic lung disease. Proc Natl Acad Sci U S A 2013; 110:8152-7. [PMID: 23630275 DOI: 10.1073/pnas.1216671110] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Allergic asthma is a heterogeneous inflammatory disorder of the airways characterized by chronic airway inflammation and airway hyperresponsiveness. Numbers of CD8(+)IL-13(+) T cells are increased in asthmatics and during the development of experimental asthma in mice. In an atopic environment rich in IL-4, these CD8(+) T cells mediate asthmatic responses, but the mechanisms regulating the conversion of CD8(+) effector T cells from IFN-γ- to pathogenic IL-13-producing effector cells that contribute to an asthma phenotype have not been defined. Here, we show that cholesterol side-chain cleavage P450 enzyme, Cyp11a1, is a key regulator of CD8(+) T-cell conversion. Expression of the gene, protein, and enzymatic activity of Cyp11a1 were markedly increased in CD8(+) T cells differentiated in the presence of IL-2 plus IL-4 compared with cells differentiated in IL-2 alone. Inhibition of Cyp11a1 enzymatic activity with aminoglutethimide or reduction in the expression of Cyp11a1 using short hairpin RNA prevented the IL-4-induced conversion of IFN-γ- to IL-13-producing cells without affecting expression of the lineage-specific transcription factors T-box expressed in T cells (T-bet) or GATA binding protein 3 (GATA3). Adoptive transfer of aminoglutethimide-treated CD8(+) T cells into sensitized and challenged CD8-deficient recipients failed to restore airway hyperresponsiveness and inflammation. We demonstrate that Cyp11a1 controls the phenotypic conversion of CD8(+) T cells from IFN-γ to IL-13 production, linking steroidogenesis in CD8(+) T cells, a nonclassical steroidogenic tissue, to a proallergic differentiation pathway.
Collapse
|
18
|
Effects of trauma-hemorrhage and IL-6 deficiency on splenic immune function in a murine trauma model. Mediators Inflamm 2012; 2012:186709. [PMID: 22496597 PMCID: PMC3306962 DOI: 10.1155/2012/186709] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2011] [Accepted: 11/10/2011] [Indexed: 01/08/2023] Open
Abstract
Splenic immune function is known to be depressed following hemorrhage. The present study investigates the effects of femoral shaft fracture, isolated or in combination with hemorrhage, on early stage cytokine production capacity of splenocytes and observes the role of IL-6 under these conditions. Male IL-6 knockout (IL-6−/−) and wild-type mice (WT) were randomly divided into three groups: sham (S), isolated femoral fracture (Fx), and femoral fracture + volume controlled hemorrhage (TH-Fx) (n = 6 per group). Animals were sacrificed four hours after induction of hemorrhage and fracture. Cytokine release (TNF-α, IL-6, and IL-10) of isolated and LPS-stimulated splenocytes was determined by cytometric bead array. Femoral fracture with or without hemorrhage caused a suppression of in vitro cytokine production capacity of splenocytes at an early posttraumatic stage in WT and IL-6−/−. In the absence of IL-6, the profile of splenic cytokine secretion is significantly altered, identifying this cytokine as a potential therapeutic target to modulate the posttraumatic immune response.
Collapse
|
19
|
Casagrande S, Groothuis TGG. The interplay between gonadal steroids and immune defence in affecting a carotenoid-dependent trait. Behav Ecol Sociobiol 2011; 65:2007-2019. [PMID: 21957328 PMCID: PMC3172405 DOI: 10.1007/s00265-011-1210-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2011] [Revised: 05/30/2011] [Accepted: 05/31/2011] [Indexed: 02/03/2023]
Abstract
The hypothesis that sexual ornaments are honest signals of quality because their expression is dependent on hormones with immune-depressive effects has received ambiguous support. The hypothesis might be correct for those signals that are carotenoid-dependent because the required carotenoid deposition in the signal, stimulated by testosterone, might lower the carotenoid-dependent immune defence of the organism. Two pathways underlying this androgen-dependent honest signaling have been suggested. Firstly, androgens that are needed for ornament expression may suppress immune defence, a cost that only high-quality animals can afford. Alternatively, immune activation may downregulate the production of androgens in low-quality individuals. Which of these alternatives is correct, and to what extent these effects are mediated by the different metabolites of androgens, remain open questions. To provide answers to these questions, we manipulated the levels of testosterone (T), 5α-dihydrotestosterone (DHT), and 17-β-estradiol (E2) in diamond doves Geopelia cuneata, a species in which both sexes exhibit a carotenoid-dependent, androgen-regulated red–orange periorbital ring of bare skin. On the first day of the experiment (day 0), we inserted steroid-releasing implants into groups of birds and on day 14, we subjected half of the birds to an immunological challenge by immunizing them with sheep red blood cells (SRBC). In females, but not in males, androgen but not estradiol treatments reduced antibody production to SRBC. In addition, the immunological challenge reduced redness and size of the trait as well as androgens levels in both sexes and in all treatments. This indicates that an immunological challenge can lower circulating T at the cost of the trait expression. These findings are in accordance with both pathways postulated in the immunocompetence-handicap hypothesis, but do not entirely support the idea that the immunosuppressive effect of androgens yields honest signaling since both T and DHT were not immunosuppressive in males, for which sexual signaling is supposed to be especially important.
Collapse
Affiliation(s)
- Stefania Casagrande
- Behavioural Biology, Centre for Behaviour and Neuroscience, University of Groningen, Nijenborgh 7, 9747 AG Groningen, The Netherlands
- Department of Evolutionary and Functional Biology, University of Parma, 43100 Parma, Italy
| | - Ton G. G. Groothuis
- Behavioural Biology, Centre for Behaviour and Neuroscience, University of Groningen, Nijenborgh 7, 9747 AG Groningen, The Netherlands
| |
Collapse
|
20
|
Misiti S, Stigliano A, Borro M, Gentile G, Michienzi S, Cerquetti L, Bucci B, Argese N, Brunetti E, Simmaco M, Toscano V. Proteomic profiles in hyperandrogenic syndromes. J Endocrinol Invest 2010; 33:156-64. [PMID: 19820292 DOI: 10.1007/bf03346575] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) and congenital adrenal hyperplasia (CAH) represent the most common causes of hyperandrogenism. Although the etiopathogeneses of these syndromes are different, they share many clinical and biochemical signs, such as hirsutism, acne, and chronic anovulation. Experimental data have shown that peripheral T-lymphocytes function as molecular sensors, being able to record molecular signals either at staminal and mature cell levels, or hormones at systemic levels. METHODS Twenty PCOS women and 10 CAH with 21-hydroxylase deficiency, aged between 18-35 yr, were studied. T-cells purified from all patients and 20 healthy donors have been analyzed by 2-dimensional gel electrophoresis. Silver-stained proteomic map of each patient was compared with a control map obtained by pooling protein samples of the 20 healthy subjects. RESULTS Spots of interest were identified by peptide mass fingerprint. Computer analysis evidenced several peptidic spots significantly modulated in all patients examined. Some proteins were modulated in both syndromes, others only in PCOS or in CAH. These proteins are involved in many physiological processes as the functional state of immune system, the regulation of the cytoskeleton structure, the oxidative stress, the coagulation process, and the insulin resistance. CONCLUSION Identification of the physiological function of these proteins could help to understand ethiopathogenetic mechanisms of hyperandrogenic syndromes and its complications.
Collapse
Affiliation(s)
- S Misiti
- II Faculty of Medicine, Sapienza University of Rome, Rome, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Mukhopadhyay R, Mishra MK, Basu A, Bishayi B. Effect of particulate antigenic stimulation or in vivo administration of interleukin-6 on the level of steroidogenic enzymes in adrenal glands and lymphoid tissues of mice with parallel alteration in endogenous inflammatory cytokine level. Cell Immunol 2010; 261:23-8. [DOI: 10.1016/j.cellimm.2009.10.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2009] [Revised: 07/21/2009] [Accepted: 10/13/2009] [Indexed: 10/20/2022]
|
22
|
Abstract
Sepsis is a major cause of mortality and morbidity in the trauma patient. Sepsis following traumatic injury is related to the type of injury, together with the extent of injury and the anatomical location. Burn injuries are associated with the highest risk of sepsis. The diagnosis of sepsis in the trauma patient remains difficult. Interpretation of abnormal results is key to successful diagnosis, particularly in conjunction with clinical findings. This review will consider the specific features of sepsis in the context of trauma relating to epidemiology, risk factors, diagnosis and management.
Collapse
Affiliation(s)
- Robert Thornhill
- Department of Military Anaesthesia and Critical Care, Royal Centre for Defence Medicine, Raddlebarn Road, Selly Oak, Birmingham, B29 6JD, UK, , Department of Critical Care Medicine, Queen Elizabeth Medical Centre, University Hospital Birmingham, Edgbaston, Birmingham B15 2TH, UK
| | - Dan Strong
- Department of Critical Care Medicine, Queen Elizabeth Medical Centre, University Hospital Birmingham, Edgbaston, Birmingham B15 2TH, UK
| | - Suresh Vasanth
- Department of Critical Care Medicine, Queen Elizabeth Medical Centre, University Hospital Birmingham, Edgbaston, Birmingham B15 2TH, UK
| | - Iain Mackenzie
- Department of Critical Care Medicine, Queen Elizabeth Medical Centre, University Hospital Birmingham, Edgbaston, Birmingham B15 2TH, UK, School of Clinical and Experimental Medicine, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| |
Collapse
|
23
|
Mukhopadhyay R, Mishra MK, Basu A, Bishayi B. Modulation of steroidogenic enzymes in murine lymphoid organs after immune activation. Immunol Invest 2009; 38:14-30. [PMID: 19172483 DOI: 10.1080/08820130802480570] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
To study the effects of immune cell activation by a protein antigen or lymphoid tissue derived cytokines on peripheral steroidogenesis activities of 3beta HSD and 17beta HSD was measured in lymphoid organs of control and BSA immunized mice after 3 weeks treatment. We demonstrated the presence of 3betaHSD and 17betaHSD in the lymphoid organs after active immunization. We found elevated serum corticosterone after 3 weeks of antigen administration in presence of CFA and a higher serum IL-6 level that also alter lymphoid tissue cytokine responses like TNF-alpha, IL-12p70, and IL-6, among which IL-12p70 and TNF-alpha down-regulate the activity of steroidogenic enzymes in the thymus during an immune response.
Collapse
Affiliation(s)
- Rupanjan Mukhopadhyay
- Department of Physiology, University Colleges of Science and Technology, University of Calcutta, West Bengal, India
| | | | | | | |
Collapse
|
24
|
Raju R, Chaudry IH. Sex steroids/receptor antagonist: their use as adjuncts after trauma-hemorrhage for improving immune/cardiovascular responses and for decreasing mortality from subsequent sepsis. Anesth Analg 2008; 107:159-66. [PMID: 18635483 DOI: 10.1213/ane.0b013e318163213d] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Studies in human as well as animal models demonstrate that females in the proestrus cycle (i.e., with high estrogen) tolerate trauma-hemorrhage and sepsis far better than males. The female sex steroid, estrogen, is the significant factor contributing to this observed gender difference in outcome. One reason for the lack of significant gender association in some clinical studies is the possibility of heterogeneity of the population in terms of their hormonal status at the time of injury. Several experimental investigations have revealed that androgens produce immune and cardiovascular depression after trauma-hemorrhage. However, the use of an androgen receptor antagonist after trauma-hemorrhage has salutary effects of immune and cardiovascular function. Likewise, estrogen produces beneficial effects on immune and cardiovascular function after trauma-hemorrhage and significantly decreases mortality rates from subsequent sepsis. The salutary effects of estrogen after trauma-hemorrhage have been shown to be due to both genomic and nongenomic effects. Thus, the use of an estrogen or androgen receptor antagonist as an adjunct after trauma-hemorrhage is a safe and novel approach for restoring immune and cardiovascular function after trauma-hemorrhage and for decreasing the mortality from subsequent sepsis.
Collapse
Affiliation(s)
- Raghavan Raju
- Center for Surgical Research and Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama 35294-0019, USA
| | | |
Collapse
|
25
|
Raju R, Bland KI, Chaudry IH. Estrogen: a novel therapeutic adjunct for the treatment of trauma-hemorrhage-induced immunological alterations. Mol Med 2008; 14:213-21. [PMID: 18235843 DOI: 10.2119/2008-00001.raju] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2008] [Accepted: 01/17/2008] [Indexed: 12/16/2022] Open
Abstract
Trauma-hemorrhage leads to prolonged immune suppression, sepsis, and multiple organ failure. The condition affects all compartments of the immune system, and extensive studies have been carried out elucidating the immunological events following trauma-hemorrhage. The immune alteration observed following trauma-hemorrhage is gender dependent in both animal models and humans, though some studies in humans are contradictory. Within 30 min after trauma-hemorrhage, splenic and peritoneal macrophages, as well as T-cell function, are depressed in male animals, but not in proestrus females. Studies have also shown that the mortality [corrected] rate and the induction of subsequent sepsis following trauma-hemorrhage are significantly higher in males and ovariectomized females compared with proestrus females. These and other investigations show that sex hormones form the basis of this gender dichotomy, and administration of estrogen can ameliorate the immune depression and increase the survival rate after trauma-hemorrhage. This review specifically elaborates the studies carried out thus far demonstrating immunological alteration after trauma-hemorrhage and its modulation by estrogen. Also, estrogen was shown to produce its salutary effects through nuclear as well as extranuclear receptors. Estrogen rapidly activates several protein kinases and phosphatases, as well as the release of calcium in different cell types. The results of the studies exemplify the promise of estrogen as a therapeutic adjunct in treating adverse pathophysiological conditions following trauma-hemorrhage.
Collapse
Affiliation(s)
- Raghavan Raju
- Center for Surgical Research and Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama 35294-0019, USA
| | | | | |
Collapse
|
26
|
Dossett LA, Swenson BR, Heffernan D, Bonatti H, Metzger R, Sawyer RG, May AK. High levels of endogenous estrogens are associated with death in the critically injured adult. THE JOURNAL OF TRAUMA 2008; 64:580-5. [PMID: 18332796 PMCID: PMC2654263 DOI: 10.1097/ta.0b013e31816543dd] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Sex hormones exhibit predictable changes in their physiologic patterns during critical illness. Endogenous estrogens are elevated in both genders as a result of the peripheral conversion of androgens to estrogens by the aromatase enzyme. Elevated endogenous estrogens have been associated with death in medical and mixed surgical intensive care unit (ICU) patients. Our objective was to determine the relationship between endogenous estrogens and outcomes in critically injured patients. METHODS A prospective cohort of injured patients remaining in the ICU for at least 48 hours at two trauma centers was enrolled. Sex hormones (estradiol, progesterone, testosterone, prolactin, and dehydroepiandrosterone-sulfate) were assayed and mortality was assessed. A logistic regression model was used to determine the association between estradiol and death. The area under the receiver operating characteristic (AUROC) curve was used to estimate the accuracy of estradiol in predicting death. RESULTS Nine hundred ninety-one patients were enrolled with a 13.4% mortality rate. Despite no detectable difference in mortality among genders, estradiol was significantly elevated in nonsurvivors (16 pg/mL vs. 35 pg/mL, p < 0.001). Estradiol was a marker for injury severity with the most severely injured patients exhibiting the highest levels. The ability of estradiol to predict death (AUROC = 0.65) was comparable with Trauma and Injury Severity Score (AUROC = 0.65) and superior to Injury Severity Score (AUROC = 0.54) in this cohort. CONCLUSIONS Serum estradiol is a marker of injury severity and a predictor of death in the critically injured patient, regardless of gender. Whether or not estradiol plays a causal role in outcomes is unclear, but estrogen modulation represents a potential therapy for improving outcomes in critically ill trauma patients.
Collapse
Affiliation(s)
- Lesly A Dossett
- Division of Trauma and Surgical Critical Care, Department of Surgery, Vanderbilt University Medical Center; Nashville, Tennessee, USA.
| | | | | | | | | | | | | |
Collapse
|
27
|
Influence of surgical trauma on the mRNA expression of sex hormone receptors in PBMCs in male and female patients. Langenbecks Arch Surg 2008; 393:871-6. [PMID: 18297304 DOI: 10.1007/s00423-008-0304-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2007] [Accepted: 01/31/2008] [Indexed: 11/27/2022]
Abstract
INTRODUCTION Gender-specific immune responses have been found after trauma-hemorrhage. Male and female sex hormones seem to be responsible for this gender dimorphism. Alterations in sex hormone receptor expression in mice appear to contribute to the immunomodulatory effect of sex hormones after blood loss. The effect of surgical trauma on the expression of sex hormone receptors in peripheral blood mononuclear cells (PBMCs) from patients, however, remains unknown. MATERIALS AND METHODS PBMCs were obtained from 14 patients (7 men and 7 women) undergoing major abdominal surgery preoperatively and 2 h postoperatively. The expression of the androgen and the estrogen alpha- and beta- receptors were determined by reverse transcriptase polymerase chain reaction (RT-PCR). beta-Actin was used as housekeeping gene. RESULTS The results indicate that surgical trauma has no influence on the expression of the androgen receptor and the estrogen receptors alpha and beta in male and female patients. DISCUSSION The data demonstrate that, in contrast to mice, no alterations in the expression of androgen and estrogen hormone receptors were evident after surgery in patients. Thus, differences in the expression of sex hormone receptors do not appear to be responsible for the gender-specific immune response after surgery.
Collapse
|
28
|
Dossett LA, Swenson BR, Evans HL, Bonatti H, Sawyer RG, May AK. Serum estradiol concentration as a predictor of death in critically ill and injured adults. Surg Infect (Larchmt) 2008; 9:41-8. [PMID: 18363467 PMCID: PMC2654258 DOI: 10.1089/sur.2007.037] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Whereas animal models of sepsis demonstrate survival benefits for the pro-estrus state, human observational studies have failed to demonstrate a consistent survival advantage among female patients. Estrogen biosynthesis differs substantially in primate and non-primate animals, and estrogens have diverse immunologic actions. Estrogen concentrations are elevated in response to critical illness and injury (regardless of sex), and elevated concentrations of serum estradiol are associated with a higher mortality rate. Our objective was to determine the predictive ability and test characteristics of the serum estradiol concentration at 48 h in critically ill patients. METHODS A prospective cohort study of surgical and trauma adult intensive care unit patients at two academic tertiary-care centers. Sex hormones (estradiol, progesterone, testosterone, prolactin, and dehydroepiandrosterone) and cytokines were assayed at 48 h, and the 28-day all-cause mortality rate was assessed. RESULTS There was no difference in mortality rates between the sexes (survivors being male in 75.2% of cases vs. 76.0% in non-survivors; p = 0.43). The serum estradiol concentration was significantly elevated in non-survivors regardless of sex (median 18.7 pg/mL [interquartile range {IRQ} 9.99-43.6] in survivors and 40.7 pg/mL [IQR 9.99-94.8] in non-survivors; p < 0.001). The area under the receiver-operating characteristic (ROC) curve for serum estradiol was 0.64 (95% confidence interval [CI] 0.55, 0.72). The parameter with the largest ROC curve was the Acute Physiology and Chronic Health Evaluation (APACHE) II score (0.75; 95% CI 0.68, 0.82). A serum estradiol cut-point of 50 pg/mL was 48% sensitive and 80% specific in predicting death and classified the outcome of 76% of patients correctly. CONCLUSIONS Serum estradiol concentration is a valuable prognostic tool and potential contributor to adverse outcomes of critically ill or injured surgical patients.
Collapse
Affiliation(s)
- Lesly A Dossett
- Division of Trauma & Surgical Critical Care, Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee 37212, USA
| | | | | | | | | | | |
Collapse
|
29
|
Abstract
A major consequence of traumatic injury is immunosuppression. Findings from previous studies suggest that the depression of immune functions is severe in young males, ovariectomised and aged females. In contrast, the immune functions in proestrus females following trauma-haemorrhage are maintained. Studies have also shown that the survival rate in proestrus females following trauma-haemorrhage and the induction of subsequent sepsis is significantly higher than in age-matched males and ovariectomised females. Furthermore, administration of female sex hormone 17beta-oestradiol in males and ovariectomised females after trauma-haemorrhage prevents the suppression of immune response. Thus, these findings suggest that sex hormones play a significant role in shaping the host response following trauma. This article reviews studies delineating the mechanism by which sex hormones regulate immune cell functions in the experimental model of trauma-haemorrhage. The findings from the studies reviewed in this article suggest that sex steroids can be synthesised by the immune cell. The findings further indicate that T cell and macrophages express receptors for androgen and oestrogen. Since these cells are also the cells that produce cytokines, local synthesis of active steroids in these cells may become the significant factor in modulating their cytokine production.
Collapse
Affiliation(s)
- Mashkoor A Choudhry
- Center for Surgical Research and Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | |
Collapse
|
30
|
Borro M, Gentile G, Stigliano A, Misiti S, Toscano V, Simmaco M. Proteomic analysis of peripheral T lymphocytes, suitable circulating biosensors of strictly related diseases. Clin Exp Immunol 2007; 150:494-501. [PMID: 17900306 PMCID: PMC2219372 DOI: 10.1111/j.1365-2249.2007.03498.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
T lymphocytes and/or their subpopulations from peripheral blood may represent molecular sensors to be used for the evaluation of gene expression modification in physiological and pathological conditions, providing a unique and easily available biological model for integrated studies of gene expression in humans. In this study, a proteomic approach was applied to evaluate the association between changes in T cell protein expression patterns and specific diseased conditions. In particular, two hyperandrogenic syndromes were studied, sharing many clinical and biochemical signs: polycystic ovary syndrome (PCOS) and congenital adrenal hyperplasia (CAH). Comparison of proteomic maps of T lymphocytes derived from patients affected by PCOS or CAH with those derived from healthy subjects showed that 14 proteins are expressed differentially in both PCOS and CAH, 15 exclusively in PCOS and 35 exclusively in CAH. Seventeen of these proteins have been identified by mass spectrometry analysis. Furthermore, proteomic data mining by hierarchical clustering was performed, highlighting T lymphocytes competence as a living biosensor system.
Collapse
Affiliation(s)
- M Borro
- Department of Biochemical Sciences and Laboratory of Advanced Molecular Diagnostics, II Faculty of Medicine, University La Sapienza, S. Andrea Hospital, Rome, Italy
| | | | | | | | | | | |
Collapse
|
31
|
Angele MK, Frantz MC, Chaudry IH. Gender and sex hormones influence the response to trauma and sepsis: potential therapeutic approaches. Clinics (Sao Paulo) 2006; 61:479-88. [PMID: 17072448 DOI: 10.1590/s1807-59322006000500017] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2006] [Accepted: 08/07/2006] [Indexed: 11/22/2022] Open
Abstract
Several clinical and experimental studies have demonstrated gender dimorphism in immune and organ responsiveness and in the susceptibility to and morbidity from shock, trauma, and sepsis. In this respect, cell-mediated immune responses have been shown to be depressed in males following trauma-hemorrhage, whereas they were aintained/enhanced in proestrus females. Furthermore, sex hormones have been shown to be responsible for this gender-specific immune response following adverse circulatory conditions. More specifically, studies indicate that androgens produce immunodepression following trauma-hemorrhage in males. In contrast, female sex steroids appear to exhibit immunoprotective properties following trauma and severe blood loss. With regard to the underlying mechanisms, receptors for sex hormones have been identified on various immune cells suggesting direct effects of these hormones on the immune cells. Alternatively, indirect effects of sex hormones, ie, modulation of cardiovascular responses or androgen- and estrogen-synthesizing enzymes, might contribute to gender-specific immune responses. Recent studies indicate that sex hormones, eg, dehydroepiandrosterone (DHEA), also modulate the function of peripheral blood mononuclear cells in surgical patients. Thus, the immunomodulatory properties of sex hormones/receptor antagonists/sex steroid synthesizing enzymes following trauma-hemorrhage suggests novel therapeutic strategies for the treatment of immunodepression in surgical patients.
Collapse
Affiliation(s)
- Martin K Angele
- Department of Surgery, Klinikum Grosshadern, Munich, Germany
| | | | | |
Collapse
|
32
|
Mayr S, Walz CR, Angele P, Hernandez-Richter T, Chaudry IH, Loehe F, Jauch KW, Angele MK. Castration prevents suppression of MHC class II (Ia) expression on macrophages after trauma-hemorrhage. J Appl Physiol (1985) 2006; 101:448-53. [PMID: 16614359 DOI: 10.1152/japplphysiol.00166.2006] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Several studies indicate that cell-mediated immune responses, i.e., macrophage (MΦ) cytokine release capacities, myosin heavy chain (MHC) class II (Ia) expression, etc., are suppressed after trauma-hemorrhage in male mice. Testosterone has been shown to be responsible for the depression of MΦ cytokine responses in males after trauma-hemorrhage. Antigen presentation via MHC class II plays a key role in initiating and maintaining cell-mediated and humoral immune responses. It remains unknown, however, whether testosterone has any effect on MHC class II after trauma-hemorrhage. To study this, male C3H/HeN mice were castrated or sham castrated 2 wk before trauma (midline laparotomy) and hemorrhage (Hem; blood pressure 35 ± 5 mmHg for 90 min and resuscitation) or sham operation. Four hours thereafter, MHC class II (Ia) expression was measured using flow cytometry. The results indicate that MHC class II (Ia) expression on peritoneal and splenic MΦ was significantly suppressed in male mice after trauma-hemorrhage. Prior castration, however, prevented the depression in MHC class II (Ia) expression on peritoneal and splenic MΦ after trauma-hemorrhage. Castration did not affect MHC class II (Ia) expression in MΦ from sham-castrated mice. Thus testosterone depresses MHC class II (Ia) expression on peritoneal and splenic MΦ after trauma-hemorrhage in males. Because MHC class II is necessary for an adequate immune response, our results suggest that depletion of male sex steroids or blockade of androgen receptors using agents such as flutamide might prevent immunosuppression via maintaining MHC class II (Ia) expression after trauma and severe blood loss.
Collapse
Affiliation(s)
- S Mayr
- Dept. of Surgery, Klinikum Grosshadern, Ludwig-Maximilians Univ., Marchioninistr. 15, 81377 Munich, Germany
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Salazar KD, Miller MR, Barnett JB, Schafer R. Evidence for a Novel Endocrine Disruptor: The Pesticide Propanil Requires the Ovaries and Steroid Synthesis to Enhance Humoral Immunity. Toxicol Sci 2006; 93:62-74. [PMID: 16788000 DOI: 10.1093/toxsci/kfl038] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Steroid hormones are known to affect the humoral immune response to a variety of antigens. However, the mechanisms regulating these effects are poorly understood. The immunotoxic chemical propanil and estrogen have similar effects on the immune system including augmentation of humoral immune responses. Propanil enhances the number of phosphorylcholine (PC)-specific IgG2b, IgG3, and IgM antibody-secreting cells (ASCs) in the spleen four- to sixfold 7 days after vaccination of female C57BL/6 mice with heat-killed Streptococcus pneumoniae. Several experiments were performed to test the hypothesis that propanil increases the response via an estrogenic pathway. Ovariectomy abrogated the effect of propanil on the PC-specific ASC response. Both in vitro and in vivo assays indicate that propanil does not bind either estrogen receptor (ER) alpha or beta. Exogenous estradiol administration in ovariectomized mice failed to restore the effect of propanil on the PC response. Treatment of female mice with a pure ER antagonist, ICI 182,780, or the progesterone antagonist RU486 did not inhibit the increase in ASCs. These data suggest that estrogen and progesterone do not regulate the effect of propanil. However, complete inhibition of steroid synthesis with the gonadotropin-releasing hormone (GnRH) antagonist antide abrogated the increased response in propanil-treated mice, indicating a necessary role for steroid synthesis. Experiments in male mice demonstrated that propanil increased the number of ASCs comparable to female mice. However, orchiectomy did not inhibit this effect, suggesting that androgens do not regulate the amplification of the humoral response. These data suggest a novel role for the ovarian hormones in the regulation of the PC-specific antibody response.
Collapse
Affiliation(s)
- Keith D Salazar
- Department of Microbiology, Immunology and Cell Biology, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, West Virginia 26506, USA
| | | | | | | |
Collapse
|
34
|
Hsieh YC, Yang S, Choudhry MA, Yu HP, Bland KI, Schwacha MG, Chaudry IH. Flutamide restores cardiac function after trauma-hemorrhage via an estrogen-dependent pathway through upregulation of PGC-1. Am J Physiol Heart Circ Physiol 2006; 290:H416-23. [PMID: 16155096 DOI: 10.1152/ajpheart.00865.2005] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Although previous studies have shown that flutamide improves cardiovascular function after trauma-hemorrhage, the mechanisms responsible for the salutary effect remain unknown. We hypothesized that flutamide mediates its beneficial effects via an estrogen-dependent pathway through upregulation of peroxisome proliferator-activated receptor-γ coactivator 1 (PGC-1). PGC-1, a key regulator of cardiac mitochondrial ATP production, induces mitochondrial DNA (mtDNA)-encoded genes such as cytochrome- c oxidase (COX) subunit I, II, and III (COX I, COX II, and COX III), which regulates mitochondrial oxidative phosphorylation. To test this hypothesis, male rats underwent trauma-hemorrhage (mean arterial pressure of 35–40 mmHg for ∼90 min) followed by resuscitation. At the onset of resuscitation, rats received vehicle, flutamide (25 mg/kg body wt), flutamide in combination with estrogen receptor (ER) antagonist ICI-182,780 (3 mg/kg body wt), or ICI-182,780 alone. Flutamide administration after trauma-hemorrhage restored the depressed cardiac function and increased cardiac testosterone, estrogen levels, and aromatase activity. These increases were accompanied by normalized cardiac ER-α and ER-β protein levels, PGC-1, and COX I mRNA expression, mitochondrial COX activity, and ATP contents. However, cardiac dihydrotestosterone, 5α-reductase II, androgen receptor protein levels, and mtDNA-encoded genes COX II and COX III were unaffected by flutamide treatment. The flutamide-mediated restoration of cardiac function, the increases in aromatase activity and estrogen levels, ER-α, ER-β, PGC-1, COX I, COX activity, and ATP contents were, however, abolished when ER antagonist ICI-182,780 was administrated along with flutamide. These findings suggest that the salutary effect of flutamide on cardiac function after trauma-hemorrhage is mediated via an estrogen-dependent pathway through upregulation of PGC-1.
Collapse
Affiliation(s)
- Ya-Ching Hsieh
- Center for Surgical Research, Univ. of Alabama at Birmingham, 1670 Univ. Boulevard, Birmingham Alabama 35294-0019, USA
| | | | | | | | | | | | | |
Collapse
|
35
|
Choudhry MA, Schwacha MG, Hubbard WJ, Kerby JD, Rue LW, Bland KI, Chaudry IH. GENDER DIFFERENCES IN ACUTE RESPONSE TO TRAUMA-HEMORRHAGE. Shock 2005; 24 Suppl 1:101-6. [PMID: 16374381 DOI: 10.1097/01.shk.0000191341.31530.5e] [Citation(s) in RCA: 119] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
To understand the pathogenesis of a disease, experimental models are needed. A good experimental model is the one that simulates responses observed in the clinical setting. In recent years, clinical studies have indicated that gender might be a factor that plays a significant role in the outcome of patients with shock, trauma, and sepsis. These observations are now being evaluated in experimental setting. Studies performed in a rodent model of trauma-hemorrhage have concluded that alterations in immune and cardiac functions after trauma-hemorrhage are more markedly depressed in adult males, and ovariectomized and aged females. However, both are maintained in castrated males and in proestrus females. Moreover, the survival rate of proestrus females subjected to sepsis after trauma-hemorrhage is significantly higher than age-matched males or ovariectomized females. Although these observations suggest gender-specific response after trauma-hemorrhage, the mechanisms responsible for gender specificity remain largely unknown. Furthermore, in other injuries such as burn, experimental studies dealing with sexual dimorphism are limited. Therefore, more studies in clinical and experimental settings are required to determine whether gender-specific responses are global across the injuries or are observed in specific injury situations. Studies are also needed to delineate underlying mechanisms responsible for differences between males and females after trauma-hemorrhage. The information gained from the experimental studies will help in designing innovative therapeutic approaches for the treatment of trauma patients.
Collapse
Affiliation(s)
- Mashkoor A Choudhry
- Department of Surgery, University of Alabama, Birmingham, AL 35294-0019, USA.
| | | | | | | | | | | | | |
Collapse
|
36
|
Frantz MC, Prix NJ, Wichmann MW, van den Engel NK, Hernandez-Richter T, Faist E, Chaudry IH, Jauch KW, Angele MK. Dehydroepiandrosterone restores depressed peripheral blood mononuclear cell function following major abdominal surgery via the estrogen receptors. Crit Care Med 2005; 33:1779-86. [PMID: 16096456 DOI: 10.1097/01.ccm.0000172278.91959.38] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Peripheral blood mononuclear cell (PBMC) dysfunction occurs following major abdominal surgery and correlates with an increased rate of septic complications. Studies have shown that dehydroepiandrosterone (DHEA) restores cell-mediated immune responses after trauma-hemorrhage in mice. Nonetheless, it remains unknown whether DHEA has any salutary effects on depressed PBMC function in surgical patients. DESIGN Laboratory experiment. SETTING University laboratory. PATIENTS Fifteen patients undergoing major abdominal surgery. INTERVENTIONS Blood samples were obtained preoperatively and 2 hrs postoperatively. MEASUREMENTS AND MAIN RESULTS PBMCs were cultured with 33% plasma in the presence or absence of DHEA (10(-10) M, 10(-8) M physiologic concentration, 10(-6) M, 10(-5) M). In an additional set of samples, the estrogen receptor antagonist tamoxifen (10(-6) M) was added. The release of proinflammatory cytokines (interleukin-1beta, interleukin-6, and tumor necrosis factor-alpha) was measured in the supernatants by enzyme-linked immunosorbent assay. Abdominal surgery resulted in depressed interleukin-1beta and tumor necrosis factor-alpha release by PBMC. Addition of DHEA to the culture medium, however, significantly improved the release of interleukin-1beta and tumor necrosis factor-alpha and stimulated the interleukin-6 release capacity of PBMC. This effect was most pronounced for a concentration of 10(-5)M DHEA. The immunomodulatory effect of DHEA on PBMC cytokine release was completely blocked by tamoxifen. In contrast, the modulatory effect of DHEA was enhanced by the addition of postoperative plasma. CONCLUSIONS DHEA stimulates proinflammatory cytokine release capacities of human PBMCs following major abdominal surgery. The estrogen receptor appears to be involved in mediating the immunomodulatory effect of DHEA. Thus, DHEA might be a useful adjunct for preventing immunosuppression in surgical patients.
Collapse
Affiliation(s)
- Markus C Frantz
- Department of Surgery, Klinikum Grosshadern, Ludwig-Maximilians University, Munich, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
Aging in men is associated with a progressive decline in the production of several hormones, including androgens. The extent to which an age-dependent decline in androgen levels lead to health problems or can affect quality of life remains under debate. Clinical results on replacement therapy do not yet provide a definitive clue on the benefit/risk balance. A sexual dimorphism of the immune system is well established, and the differences between female and male immune responses under normal, as well as pathological, conditions are generally attributed to the influence of estrogens, progestins, and androgens. The suppressive effects of male sex hormones on immune functions have been observed in a wide variety of disease processes and appear to be testosterone-mediated. Endogenous testosterone inhibits skin wound healing response in males and is associated with an enhanced inflammatory response. Although there are no known gender-related differences in permeability barrier function in adults, estrogens accelerates--whereas testosterone retards--barrier development in fetal skin, and male fetuses demonstrate slower barrier development than female littermates.
Collapse
Affiliation(s)
- S Fimmel
- Department of Dermatology, Charité Universitaetsmedizin Berlin, Berlin, Germany
| | | |
Collapse
|
38
|
Matsutani T, Samy TSA, Rue LW, Bland KI, Chaudry IH. Transgenic prolactin−/− mice: effect of trauma-hemorrhage on splenocyte functions. Am J Physiol Cell Physiol 2005; 288:C1109-16. [PMID: 15601751 DOI: 10.1152/ajpcell.00478.2004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Prolactin (PRL) is involved in the regulation of immune functions under normal and pathological conditions. Trauma-hemorrhage (T-H) produces profound immunosuppression in male mice but not in proestrus female mice. Administration of PRL in males after T-H, however, restores immune functions. In this study, PRL+/+ and transgenic (PRL−/−) male and female mice were used to assess immune suppression after T-H and to determine the reasons for the hormone's beneficial effect. In vitro lymphoproliferation assay with Nb2 cells showed complete absence of PRL in the circulation of the transgenic PRL−/− mice of both sexes, whereas very high levels of the hormone were detected in the wild-type PRL+/+ mice of both sexes. Moreover, T-H resulted in the appearance of significant levels of the hormone in circulation, but only in PRL+/+ mice. Splenocyte proliferation in male PRL−/− mice was significantly lower than in PRL+/+ mice after T-H. Marginal differences between PRL+/+ and PRL−/− mice were observed in the release of IL-2 and IFN-γ by splenocytes, while the release of IL-10 was significantly higher in PRL−/− than in PRL+/+ mice. A significant observation of our study is the release of a ∼25-kDa protein in the concanavalin A-stimulated splenocytes of male PRL+/+ and PRL−/− mice that was active in the in vitro lymphoproliferation assay with Nb2 cells. It is unlikely that this protein is PRL because it is also present in the splenocyte extracts of PRL−/− transgenic mice. Nonetheless, because control of lymphoid cell proliferation is considered one of the characteristics of the immune system, the local release of this protein may be significant in the differences observed in splenocyte cytokine release after T-H in wild-type as well as transgenic mice.
Collapse
Affiliation(s)
- Takeshi Matsutani
- Center for Surgical Research and Dept. of Surgery, Univ. of Alabama at Birmingham, Volker Hall G094, 1670 University Blvd., Birmingham, AL 35294-0019, USA
| | | | | | | | | |
Collapse
|
39
|
Molloy EJ, O'Neill AJ, Grantham JJ, Sheridan-Pereira M, Fitzpatrick JM, Webb DW, Watson RWG. Sex-specific alterations in neutrophil apoptosis: the role of estradiol and progesterone. Blood 2003; 102:2653-9. [PMID: 12791649 DOI: 10.1182/blood-2003-02-0649] [Citation(s) in RCA: 152] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Women are conferred with greater immunologic and survival benefits compared to men. Female sex steroids contribute to this sexual dimorphism. Furthermore, during human pregnancy when female sex hormones are elevated, neutrophil apoptosis is delayed. This study examines the specific effects of estradiol and progesterone on neutrophil apoptosis and function in healthy adult men and women. We also examined the contribution of these hormones to the persistence and resolution of an inflammatory response. Spontaneous apoptosis was significantly decreased in women compared with men. Physiologic doses of estradiol and progesterone caused a further delay in spontaneous apoptosis in both men and women but did not diminish Fas antibody-induced apoptosis. The delay in apoptosis was mediated at the level of the mitochondria with decreased release of cytochrome c, which may alter caspase cleavage and activity. There were no associated alterations in neutrophil CD11b, but production of reactive oxygen intermediates (ROIs) in women was increased. Thus, female sex hormones mediate delayed neutrophil apoptosis in both sexes and enhance female intracellular production of ROIs. Modulating hormonal responses may be an effective therapeutic tool in combating inflammatory diseases.
Collapse
Affiliation(s)
- Eleanor J Molloy
- Department of Surgery, Mater Misericordiae University Hospital, Dublin, Ireland
| | | | | | | | | | | | | |
Collapse
|
40
|
Schneider CP, Schwacha MG, Samy TSA, Bland KI, Chaudry IH. Androgen-mediated modulation of macrophage function after trauma-hemorrhage: central role of 5alpha-dihydrotestosterone. J Appl Physiol (1985) 2003; 95:104-12. [PMID: 12665535 DOI: 10.1152/japplphysiol.00182.2003] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Androgens have been implicated as the causative factor for the postinjury immune dysfunction in males; however, it remains unknown whether androgens directly affect macrophages. To study this, male mice were sham operated or subjected to trauma (i.e., midline laparotomy) and hemorrhagic shock (mean arterial pressure, 30 +/- 5 mmHg for 90 min and then resuscitated). The mice received the 5alpha-reductase inhibitor 4-hydroxyandrostenedione (4-OHA) before resuscitation. Plasma TNF-alpha, IL-6, and IL-10 levels were elevated after trauma-hemorrhage and normalized by 4-OHA. TNF-alpha and IL-6 production by splenic macrophages was decreased after injury, whereas Kupffer cell production of these mediators was enhanced. 4-OHA normalized cytokine production. Androgens suppressed cytokine production by splenic macrophages from hemorrhaged mice, whereas it enhanced TNF-alpha and IL-6 production by Kupffer cells. The addition of 4-OHA in vitro normalized cytokine production by cells treated with testosterone, but it had no effect on dihydrotestosterone-treated cells. These results indicate that androgens directly affect macrophage function in males after trauma and hemorrhagic shock and that the intracellular conversion of testosterone to dihydrotestosterone is of particular importance in mediating the androgen-induced effects.
Collapse
Affiliation(s)
- Christian P Schneider
- Department of Research, University of Alabama at Birmingham, Birmingham, AL 35294-0019, USA
| | | | | | | | | |
Collapse
|
41
|
Samy TSA, Zheng R, Matsutani T, Rue LW, Bland KI, Chaudry IH. Mechanism for normal splenic T lymphocyte functions in proestrus females after trauma: enhanced local synthesis of 17beta-estradiol. Am J Physiol Cell Physiol 2003; 285:C139-49. [PMID: 12660147 DOI: 10.1152/ajpcell.00058.2003] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Trauma-hemorrhage and resuscitation (TH) produces profound immunodepression and enhances susceptibility to sepsis in males but not in proestrus females, suggesting gender dimorphism in the immune responses. However, the mechanism responsible for the maintenance of immune functions in proestrus females after TH is unclear. Splenic T lymphocytes express receptors for estrogen (ER), contain enzymes involved in estrogen metabolism, and are the major source of cytokine production; the metabolism of 17beta-estradiol was assessed in the splenic T lymphocytes of proestrus and ovariectomized mice by using appropriate substrates after TH. Analysis for aromatase and 17beta-hydroxysteroid dehydrogenases indicated increased 17beta-estradiol synthesis and low conversion into estrone in T lymphocytes of proestrus but not of ovariectomized mice. The effect of 17beta-estradiol on T lymphocyte cytokine release was reliant on ER expressions. This was apparent in the differences of ER expression, especially that of ER-beta, and an association between increased 17beta-estradiol synthesis and sustained release of IL-2 and IL-6 in T lymphocytes of proestrus females after TH. Because 17beta-estradiol is able to regulate cytokine genes, and the splenic T lymphocyte cytokine releases is altered after TH, continued synthesis of 17beta-estradiol in proestrus females appears to be responsible for the maintenance of T lymphocyte cytokine release associated with the protection of immune functions after TH.
Collapse
Affiliation(s)
- T S Anantha Samy
- Department of Surgery, University of Alabama School of Medicine, Birmingham, AL 35294, USA
| | | | | | | | | | | |
Collapse
|
42
|
Angele MK, Nitsch S, Knoferl MW, Ayala A, Angele P, Schildberg FW, Jauch KW, Chaudry IH. Sex-specific p38 MAP kinase activation following trauma-hemorrhage: involvement of testosterone and estradiol. Am J Physiol Endocrinol Metab 2003; 285:E189-96. [PMID: 12791604 DOI: 10.1152/ajpendo.00035.2003] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Although immune functions are markedly depressed in males and not in proestrous females following trauma-hemorrhage (T-H), the mechanisms responsible for the divergent responses remain unknown. Because sex steroids modulate the activation of p38, our aim was to determine whether differences in the activation of p38 by phosphorylation (p38-P) might contribute to the sex-dimorphic immune response following T-H. The effects of testosterone and estradiol on the activation of p38 were also examined. Intact male mice (C3H/HeN), castrated males treated with vehicle, 5alpha-dihydrotestosterone (DHT), or 17beta-estradiol, and proestrous females were subjected to trauma (i.e., midline laparotomy) and hemorrhagic shock (35 +/- 5 mmHg for 90 min and resuscitation) or sham operation. At 2 h thereafter, splenic (SMphi) and peritoneal macrophages (PMphi) were harvested and cultured (with 10 microg/ml LPS), and Western blot analysis was carried out for quantification of p38 and p38-P. Sex, testosterone and estradiol plasma levels, and T-H did not alter the constitutive expression of p38 in SMphi and PMphi. In contrast, the activated form of p38 (p38-P) was markedly increased in SMphi and PMphi from female shams compared with male shams. Moreover, the phosphorylation of p38-P increased in males after T-H, whereas it decreased in females under those conditions. Castration before T-H prevented the increase in p38-P in males. Castrated animals treated with DHT displayed increased p38-P following T-H, whereas 17beta-estradiol had no effect on p38-P in castrated mice. Thus 1) sex influences the activation of p38 MAP kinase, 2) DHT is responsible for the increased activation of p38 in male mice, and 3) this sex-specific activation of p38 might be responsible for the sexually dimorphic immune response following T-H.
Collapse
Affiliation(s)
- Martin K Angele
- Center for Surgical Research and Department of Surgery, University of Alabama at Birmingham, 35294, USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Chaudry IH, Samy TSA, Schwacha MG, Wang P, Rue LW, Bland KI. Endocrine targets in experimental shock. THE JOURNAL OF TRAUMA 2003; 54:S118-25. [PMID: 12768113 DOI: 10.1097/01.ta.0000064511.14322.f1] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Modified resuscitation regimens and cytokine blockade/receptor antagonism after trauma have not been successful in decreasing the mortality rates from sepsis in trauma patients; therefore, an alternative approach using endocrine targets as modulators or inhibitors may be useful. Information regarding the influence of gender and hormones on immune and cardiovascular responses after nonthermal trauma-hemorrhagic shock is, on the one hand, considerable but, on the other hand, disappointingly incomplete. Trauma-hemorrhagic shock produces gender dimorphic immune and cardiovascular responses; men exhibit cardiovascular depression and are immunosuppressed, whereas proestrus women do not show cardiovascular or immunologic depression under those conditions. Furthermore, experimental studies have demonstrated the use of hormones, hormone antagonists, sex steroids, and receptor antagonists as salutary adjuncts, without any adverse effects on gastrointestinal, hepatic, and renal functions, for restoring the depressed immune and cardiovascular responses after trauma-hemorrhage. Thus, flutamide, dehydroepiandrosterone, metoclopramide, and 17beta-estradiol, which are readily availably clinically and do not produce any adverse hemodynamic effects, appear to be safe and novel agents/hormones for the treatment of immune and cardiovascular depression after severe blood loss in male and female trauma victims.
Collapse
Affiliation(s)
- Irshad H Chaudry
- Center for Surgical Research, Department of Surgery, University of Alabama at Birmingham School of Medicine, 35294-0019, USA.
| | | | | | | | | | | |
Collapse
|
44
|
Ueyama T, Shirasawa N, Numazawa M, Yamada K, Shelangouski M, Ito T, Tsuruo Y. Gastric parietal cells: potent endocrine role in secreting estrogen as a possible regulator of gastro-hepatic axis. Endocrinology 2002; 143:3162-70. [PMID: 12130582 DOI: 10.1210/endo.143.8.8974] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Estrogen, if it is produced in the gastrointestinal tract, may overflow into the systemic circulation in the case of increased portal-systemic shunting. This idea is in accord with a significant step-up in serum estradiol (E2) concentration in the portal vein of rats, compared with that in the artery. Gene expression of aromatase, estrogen synthetase, was demonstrated by RT-PCR in the gastric mucosa of male and female adult rats, equivalent to that in the ovary. Aromatase activity and production of E2 in the gastric mucosa were demonstrated by (3)H(2)O assay and gas chromatography-mass spectrometry, and they were inhibited by aromatase inhibitor, 4-hydroxyandrostenedione. Conversion of (14)C-androstenedione to (14)C-E2 through (14)C-testosterone in cultured gastric mucosa was also demonstrated. Parietal cells exhibited strong signals for aromatase mRNA and immunoreactive protein by in situ hybridization histochemistry and immunohistochemistry. Estrogen receptor alpha mRNA and immunoreactive protein were demonstrated in hepatocytes by RT-PCR, in situ hybridization histochemistry, and immunohistochemistry. Total gastrectomy reduced portal venous E2 concentration, without changing systemic E2 concentration, together with down-regulation of estrogen receptor alpha mRNA level in the liver. These findings indicate that gastric parietal cells play a potent endocrine role in secreting estrogen that may function as a regulator of the gastro-hepatic axis.
Collapse
Affiliation(s)
- Takashi Ueyama
- Department of Anatomy and Cell Biology, Wakayama Medical University, Kimiidera 811-1, Wakayama 641-8509, Japan.
| | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
There is a naturally occurring gender difference in immune responses which persists after traumatic injury. Physiological levels of 17beta-estradiol (E(2)) are immunostimulatory, whereas high pregnancy and superphysiological levels are immunosuppressive. In contrast, at all concentrations, testosterone suppresses immune responses. Evidence from this laboratory and others suggest that the gender difference in immune responses after injury is mediated in part by alterations in the circulating levels of gonadal steroid hormones through modulation of production of inflammatory and immunoregulatory cytokines, including interleukin-6 (IL-6). Aberrant production of IL-6 is known to be an important mediator of immunity after injury. Since E(2) is a critical regulator of IL-6 production and overall immune function, it suggests gender specific therapies should be considered for the treatment of patients.
Collapse
Affiliation(s)
- Elizabeth J Kovacs
- Immunology and Aging Program, Loyola University Chicago, Maywood, IL, USA.
| | | | | |
Collapse
|
46
|
Zheng R, Samy TSA, Schneider CP, Rue LW, Bland KI, Chaudry IH. Decreased 5alpha-dihydrotestosterone catabolism suppresses T lymphocyte functions in males after trauma-hemorrhage. Am J Physiol Cell Physiol 2002; 282:C1332-8. [PMID: 11997248 DOI: 10.1152/ajpcell.00560.2001] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Trauma-hemorrhage produces profound immunosuppression in males but not in proestrus females. Prior castration or flutamide treatment of males following trauma-hemorrhage prevents immunosuppression, implicating 5alpha-dihydrotestosterone for the immunosuppressive effects. 5alpha-dihydrotestosterone, a high-affinity androgen receptor-binding steroid, is synthesized in tissues as needed and seldom accumulates. The presence of steroidogenic enzymes in T lymphocytes suggests both synthesis and catabolism of 5alpha-dihydrotestosterone. We hypothesized, therefore, that the basis for high 5alpha-dihydrotestosterone activity in T lymphocytes of males following trauma-hemorrhage is due to decreased catabolism. Accordingly, catabolism of 5alpha-dihydrotestosterone was assessed in splenic T lymphocytes by examining the activity and expression of enzymes involved. Analysis showed increased synthesis and decreased catabolism of 5alpha-dihydrotestosterone in intact male T lymphocytes following trauma-hemorrhage. In contrast, reduced 5alpha-reductase activity and increased expression of 17beta-hydroxysteroid dehydrogenase oxidative isomers suggest inactivation of 5alpha-dihydrotestosterone in precastrated males. Thus our study suggests increased synthesis and decreased catabolism of 5alpha-dihydrotestosterone as a reason for loss of T lymphocyte functions in intact males following trauma-hemorrhage, as evidenced by decreased release of interleukin-2 and -6.
Collapse
Affiliation(s)
- Rui Zheng
- Center for Surgical Research and Department of Surgery, University of Alabama School of Medicine, Birmingham, Alabama 35294-0018, USA
| | | | | | | | | | | |
Collapse
|