1
|
Zhu Y, Liu X, Shi Y, Liu X, Li H, Ru S, Tian H. Prenatal exposure to bisphenol AF causes toxicities in liver, spleen, and kidney tissues of SD rats. Food Chem Toxicol 2024; 192:114939. [PMID: 39151878 DOI: 10.1016/j.fct.2024.114939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/04/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
As a replacement for bisphenol A (BPA), bisphenol AF (BPAF) showed stronger maternal transfer and higher fetal accumulation than BPA. Therefore, concerns should be raised about the health risks of maternal exposure to BPAF during gestation on the offspring. In this study, SD rats were exposed to BPAF (0, 50, and 100 mg/kg/day) during gestation to investigate the bioaccumulation and adverse effects in liver, spleen, and kidney tissues of the offspring at weaning period. Bioaccumulation of BPAF in these tissues with concentrations ranging from 1.56 ng/mg (in spleen of males) to 55.44 ng/mg (in liver of females) led to adverse effects at different biological levels, including increased relative weights of spleen and kidneys, histopathological damage in liver, spleen, and kidney, organ functional damage in liver, spleen, and kidney, upregulated expression of genes related to lipid metabolism (in liver), oxidative stress response (in kidney), immunity and inflammatory (in spleen). Furthermore, dysregulated metabolomics was identified in spleen, with 217 differential metabolites screened and 9 KEGG pathways significantly enriched. This study provides a comprehensive insight into the systemic toxicities of prenatal exposure to BPAF in SD rats. Given the broad applications and widespread occurrence of BPAF, its safety should be re-considered.
Collapse
Affiliation(s)
- Yaxuan Zhu
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Xiuxiang Liu
- Qingdao Women and Children's Hospital, Qingdao, 266034, China
| | - Yijiao Shi
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Xiuying Liu
- Wudi County Hospital of Traditional Chinese Medicine, Binzhou, 251900, China
| | - Huaxin Li
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Shaoguo Ru
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Hua Tian
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China.
| |
Collapse
|
2
|
Lee J, Yurkovetskiy LA, Reiman D, Frommer L, Strong Z, Chang A, Kahaly GJ, Khan AA, Chervonsky AV. Androgens contribute to sex bias of autoimmunity in mice by T cell-intrinsic regulation of Ptpn22 phosphatase expression. Nat Commun 2024; 15:7688. [PMID: 39227386 PMCID: PMC11372096 DOI: 10.1038/s41467-024-51869-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 08/21/2024] [Indexed: 09/05/2024] Open
Abstract
Autoimmune diseases such as systemic lupus erythematosus (SLE) display a strong female bias. Although sex hormones have been associated with protecting males from autoimmunity, the molecular mechanisms are incompletely understood. Here we report that androgen receptor (AR) expressed in T cells regulates genes involved in T cell activation directly, or indirectly via controlling other transcription factors. T cell-specific deletion of AR in mice leads to T cell activation and enhanced autoimmunity in male mice. Mechanistically, Ptpn22, a phosphatase and negative regulator of T cell receptor signaling, is downregulated in AR-deficient T cells. Moreover, a conserved androgen-response element is found in the regulatory region of Ptpn22 gene, and the mutation of this transcription element in non-obese diabetic mice increases the incidence of spontaneous and inducible diabetes in male mice. Lastly, Ptpn22 deficiency increases the disease severity of male mice in a mouse model of SLE. Our results thus implicate AR-regulated genes such as PTPN22 as potential therapeutic targets for autoimmune diseases.
Collapse
MESH Headings
- Animals
- Protein Tyrosine Phosphatase, Non-Receptor Type 22/genetics
- Protein Tyrosine Phosphatase, Non-Receptor Type 22/metabolism
- Male
- Female
- Autoimmunity
- Receptors, Androgen/metabolism
- Receptors, Androgen/genetics
- Mice
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Lupus Erythematosus, Systemic/immunology
- Lupus Erythematosus, Systemic/genetics
- Androgens/metabolism
- Mice, Knockout
- Lymphocyte Activation
- Mice, Inbred NOD
- Mice, Inbred C57BL
- Disease Models, Animal
- Signal Transduction
Collapse
Affiliation(s)
- Jean Lee
- Committee on Cancer Biology, The University of Chicago, Chicago, IL, 60637, USA
- Department of Pathology, The University of Chicago, Chicago, IL, 60637, USA
| | - Leonid A Yurkovetskiy
- Department of Pathology, The University of Chicago, Chicago, IL, 60637, USA
- Committee on Microbiology, The University of Chicago, Chicago, IL, 60637, USA
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01655, USA
| | - Derek Reiman
- Toyota Technological Institute at Chicago, Chicago, IL, 60637, USA
| | - Lara Frommer
- Department of Medicine I, Johannes Gutenberg University (JGU) Medical Center, Mainz, 55101, Germany
| | - Zoe Strong
- Department of Pathology, The University of Chicago, Chicago, IL, 60637, USA
| | - Anthony Chang
- Department of Pathology, The University of Chicago, Chicago, IL, 60637, USA
| | - George J Kahaly
- Department of Medicine I, Johannes Gutenberg University (JGU) Medical Center, Mainz, 55101, Germany
| | - Aly A Khan
- Department of Pathology, The University of Chicago, Chicago, IL, 60637, USA.
- Toyota Technological Institute at Chicago, Chicago, IL, 60637, USA.
- Department of Family Medicine, The University of Chicago, Chicago, IL, 60637, USA.
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01655, USA.
| | - Alexander V Chervonsky
- Department of Pathology, The University of Chicago, Chicago, IL, 60637, USA.
- Committee on Microbiology, The University of Chicago, Chicago, IL, 60637, USA.
- Committee on Immunology, The University of Chicago, Chicago, IL, 60637, USA.
| |
Collapse
|
3
|
Lombardo G, Mondelli V, Worrell C, Sforzini L, Mariani N, Nikkheslat N, Nettis MA, Kose M, Zajkowska Z, Cattaneo A, Pointon L, Turner L, Cowen PJ, Drevets WC, Cavanagh J, Harrison NA, Bullmore ET, Dazzan P, Pariante CM. Disturbed sex hormone milieu in males and females with major depressive disorder and low-grade inflammation. J Affect Disord 2024; 356:167-176. [PMID: 38494137 DOI: 10.1016/j.jad.2024.03.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 02/06/2024] [Accepted: 03/07/2024] [Indexed: 03/19/2024]
Abstract
Sex hormones have biological effects on inflammation, and these might contribute to the sex-specific features of depression. C-reactive protein (CRP) is the most widely used inflammatory biomarker and consistent evidence shows a significant proportion (20-30 %) of patients with major depressive disorder (MDD) have CRP levels above 3 mg/L, a threshold indicating at least low-grade inflammation. Here, we investigate the interplay between sex hormones and CRP in the cross-sectional, observational Biomarkers in Depression Study. We measured serum high-sensitivity (hs-)CRP, in 64 healthy controls and 178 MDD patients, subdivided into those with hs-CRP below 3 mg/L (low-CRP; 53 males, 72 females) and with hs-CRP above 3 mg/L (high-CRP; 19 males, 34 females). We also measured interleukin-6, testosterone, 17-β-estradiol (E2), progesterone, sex-hormone binding globulin (SHBG), follicle-stimulating and luteinising hormones, and calculated testosterone-to-E2 ratio (T/E2), free androgen and estradiol indexes (FAI, FEI), and testosterone secretion index. In males, high-CRP patients had lower testosterone than controls (p = 0.001), and lower testosterone (p = 0.013), T/E2 (p < 0.001), and higher FEI (p = 0.015) than low-CRP patients. In females, high-CRP patients showed lower SHGB levels than controls (p = 0.033) and low-CRP patients (p = 0.034). The differences in testosterone, T/E2 ratio, and FEI levels in males survived the Benjamini-Hochberg FDR correction. In linear regression analyses, testosterone (β = -1.069 p = 0.033) predicted CRP concentrations (R2 = 0.252 p = 0.002) in male patients, and SHBG predicted CRP levels (β = -0.628 p = 0.009, R2 = 0.172 p = 0.003) in female patients. These findings may guide future research investigating interactions between gonadal and immune systems in depression, and the potential of hormonal therapies in MDD with inflammation.
Collapse
Affiliation(s)
- Giulia Lombardo
- Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, Maurice Wohl Clinical Neuroscience Institute, King's College London, SE5 9RT, UK.
| | - Valeria Mondelli
- Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, Maurice Wohl Clinical Neuroscience Institute, King's College London, SE5 9RT, UK; National Institute for Health Research (NIHR) Maudsley Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King's College London
| | - Courtney Worrell
- Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, Maurice Wohl Clinical Neuroscience Institute, King's College London, SE5 9RT, UK
| | - Luca Sforzini
- Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, Maurice Wohl Clinical Neuroscience Institute, King's College London, SE5 9RT, UK; National Institute for Health Research (NIHR) Maudsley Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King's College London
| | - Nicole Mariani
- Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, Maurice Wohl Clinical Neuroscience Institute, King's College London, SE5 9RT, UK
| | - Naghmeh Nikkheslat
- Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, Maurice Wohl Clinical Neuroscience Institute, King's College London, SE5 9RT, UK
| | - Maria A Nettis
- Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, Maurice Wohl Clinical Neuroscience Institute, King's College London, SE5 9RT, UK; South London and Maudsley NHS Foundation Trust, UK
| | - Melisa Kose
- Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, Maurice Wohl Clinical Neuroscience Institute, King's College London, SE5 9RT, UK
| | - Zuzanna Zajkowska
- Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, Maurice Wohl Clinical Neuroscience Institute, King's College London, SE5 9RT, UK
| | - Annamaria Cattaneo
- Biological Psychiatric Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy; Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Linda Pointon
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge CB2 0SZ, UK
| | - Lorinda Turner
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge CB2 0SZ, UK
| | - Philip J Cowen
- University of Oxford Department of Psychiatry, Warneford Hospital, Oxford OX3 7JX, UK
| | - Wayne C Drevets
- Janssen Research & Development, Neuroscience Therapeutic Area, 3210 Merryfield Row, San Diego, CA 92121, USA
| | - Jonathan Cavanagh
- Centre for Immunobiology, University of Glasgow and Sackler Institute of Psychobiological Research, Queen Elizabeth University Hospital, Glasgow G51 4TF, UK
| | - Neil A Harrison
- School of Medicine, School of Psychology, Cardiff University Brain Research Imaging Centre, Maindy Road, Cardiff CF24 4HQ, UK
| | - Edward T Bullmore
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge CB2 0SZ, UK
| | - Paola Dazzan
- Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, Maurice Wohl Clinical Neuroscience Institute, King's College London, SE5 9RT, UK; National Institute for Health Research (NIHR) Maudsley Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King's College London
| | - Carmine M Pariante
- Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, Maurice Wohl Clinical Neuroscience Institute, King's College London, SE5 9RT, UK; National Institute for Health Research (NIHR) Maudsley Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King's College London
| |
Collapse
|
4
|
Ma TC, Gao F, Liu XL, Wang CX, Liu Q, Zhou J. Association between dietary inflammatory index and NT-proBNP levels in US adults: A cross-sectional analysis. PLoS One 2024; 19:e0304289. [PMID: 38837950 PMCID: PMC11152272 DOI: 10.1371/journal.pone.0304289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 05/09/2024] [Indexed: 06/07/2024] Open
Abstract
BACKGROUND With cardiovascular diseases standing as a leading cause of mortality worldwide, the interplay between diet-induced inflammation, as quantified by the Dietary Inflammatory Index (DII), and heart failure biomarker NT-proBNP has not been investigated in the general population. METHODS This study analyzed data from the National Health and Nutrition Examination Survey (NHANES) 1999-2004, encompassing 10,766 individuals. The relationship between the DII and NT-proBNP levels was evaluated through multivariable-adjusted regression models. To pinpoint crucial dietary components influencing NT-proBNP levels, the LASSO regression model was utilized. Stratified analyses were then conducted to examine the associations within specific subgroups to identify differential effects of the DII on NT-proBNP levels across diverse populations. RESULTS In individuals without heart failure, a unit increase in the DII was significantly associated with an increase in NT-proBNP levels. Specifically, NT-proBNP levels rose by 9.69 pg/mL (95% CI: 6.47, 12.91; p < 0.001) without adjustments, 8.57 pg/mL (95% CI: 4.97, 12.17; p < 0.001) after adjusting for demographic factors, and 5.54 pg/mL (95% CI: 1.75, 9.32; p = 0.001) with further adjustments for health variables. In participants with a history of heart failure, those in the second and third DII quartile showed a trend towards higher NT-proBNP levels compared to those in the lowest quartile, with increases of 717.06 pg/mL (95% CI: 76.49-1357.63, p = 0.030) and 855.49 pg/mL (95% CI: 156.57-1554.41, p = 0.018). Significant interactions were observed in subgroup analyses by age (<50: β = 3.63, p = 0.141; 50-75: β = 18.4, p<0.001; >75: β = 56.09, p<0.001), gender (men: β = 17.82, p<0.001; women: β = 7.43, p = 0.061),hypertension (β = 25.73, p<0.001) and diabetes (β = 38.94, p<0.001). CONCLUSION This study identified a positive correlation between the DII and NT-proBNP levels, suggesting a robust link between pro-inflammatory diets and increased heart failure biomarkers, with implications for dietary modifications in cardiovascular risk management.
Collapse
Affiliation(s)
- Teng-Chi Ma
- The First Affiliated Hospital of Xi’an Jiaotong University, Yulin Hospital, Yulin, Shaanxi, China
| | - Feng Gao
- Department of Cardiology, The Affiliated Hospital of Yan’an University, Yan’an, Shaanxi, China
| | - Xin-Lu Liu
- Department of Cardiology, The Affiliated Hospital of Yan’an University, Yan’an, Shaanxi, China
| | - Chen-Xi Wang
- Department of Cardiology, The Affiliated Hospital of Yan’an University, Yan’an, Shaanxi, China
| | - Qiang Liu
- Department of Cardiology, The Affiliated Hospital of Yan’an University, Yan’an, Shaanxi, China
| | - Jing Zhou
- Department of Cardiology, The Affiliated Hospital of Yan’an University, Yan’an, Shaanxi, China
| |
Collapse
|
5
|
Mengelkoch S, Slavich GM. Sex Differences in Stress Susceptibility as a Key Mechanism Underlying Depression Risk. Curr Psychiatry Rep 2024; 26:157-165. [PMID: 38470558 PMCID: PMC10978685 DOI: 10.1007/s11920-024-01490-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/26/2024] [Indexed: 03/14/2024]
Abstract
PURPOSE OF REVIEW Although females are at relatively greater risk for a variety of disorders, including depression, the biological mechanisms underlying this striking health disparity remain unclear. To address this issue, we highlight sex differences in stress susceptibility as a key mechanism potentially driving this effect and describe the interacting inflammatory, hormonal, epigenomic, and social-environmental mechanisms involved. RECENT FINDINGS Using the Social Signal Transduction Theory of Depression as a theoretical framework, women's elevated risk for depression may stem from a tight link between life stress, inflammation, and depression in women. Further, research finds hormonal contraceptive use alters cortisol and inflammatory reactivity to acute stress in ways that may increase depression risk in females. Finally, beyond established epigenetic mechanisms, mothers may transfer risk for depression to their female offspring through stressful family environments, which influence stress generation and stress-related gene expression. Together, these findings provide initial, biologically plausible clues that may help explain the relatively greater risk for depression in females vs. males. Looking forward, much more research is needed to address the longstanding underrepresentation of females in biomedical research on the biology of stress and depression.
Collapse
Affiliation(s)
- Summer Mengelkoch
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, USA.
| | - George M Slavich
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, USA
| |
Collapse
|
6
|
Mengelkoch S, Gassen J, Slavich GM, Hill SE. Hormonal contraceptive use is associated with differences in women's inflammatory and psychological reactivity to an acute social stressor. Brain Behav Immun 2024; 115:747-757. [PMID: 37914104 PMCID: PMC11216059 DOI: 10.1016/j.bbi.2023.10.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 10/06/2023] [Accepted: 10/29/2023] [Indexed: 11/03/2023] Open
Abstract
Women using hormonal contraceptives (HCs) exhibit numerous signs of chronic inflammation, including elevated C-reactive protein levels and greater risk of developing mood and autoimmune disorders. However, users and non-users of HCs often have similar circulating proinflammatory cytokine levels, making the mechanism of association unclear. One possible explanation for this paradox is that HC users exhibit differences in their inflammatory responses to psychosocial stress that, over time, could contribute to chronic inflammation and its pathologies. Here, we tested this possibility by examining women's glucocorticoid, inflammatory, and psychological responses to the Trier Social Stress Test (TSST) in 67 naturally cycling (NC) and 60 oral HC-using women (Mage = 19.31, SDage = 1.95). As hypothesized, HC users and NC women exhibited different glucocorticoid and proinflammatory cytokine responses to the TSST. For NC women, TSST-induced increases in glucocorticoids were uncommon, and increases in glucocorticoids were accompanied by elevations in IL-6. In contrast, for women using HCs, increases in glucocorticoids in response to the TSST were common, and increases in glucocorticoids were accompanied by increases in TNF-α. HC users and NC women also differed in their psychological responses to the TSST, with HC users reporting elevated stress levels compared to NC women. Together, these results suggest that HC use impacts women's glucocorticoid, inflammatory, and psychological responses to psychosocial stress, potentially contributing to observed differences in these women's mental and physical health.
Collapse
Affiliation(s)
- Summer Mengelkoch
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, 760 Westwood Plaza, Los Angeles, CA 90095, United States; Department of Psychology, Texas Christian University, 2955 South University Drive, Fort Worth TX 76129, United States.
| | - Jeffrey Gassen
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, 760 Westwood Plaza, Los Angeles, CA 90095, United States
| | - George M Slavich
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, 760 Westwood Plaza, Los Angeles, CA 90095, United States
| | - Sarah E Hill
- Department of Psychology, Texas Christian University, 2955 South University Drive, Fort Worth TX 76129, United States
| |
Collapse
|
7
|
Alarcon PC, Damen MSMA, Ulanowicz CJ, Sawada K, Oates JR, Toth A, Wayland JL, Chung H, Stankiewicz TE, Moreno-Fernandez ME, Szabo S, Zacharias WJ, Divanovic S. Obesity amplifies influenza virus-driven disease severity in male and female mice. Mucosal Immunol 2023; 16:843-858. [PMID: 37730122 PMCID: PMC10842771 DOI: 10.1016/j.mucimm.2023.09.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 08/18/2023] [Accepted: 09/13/2023] [Indexed: 09/22/2023]
Abstract
Influenza virus-induced respiratory pneumonia remains a major public health concern. Obesity, metabolic diseases, and female sex are viewed as independent risk factors for worsened influenza virus-induced lung disease severity. However, lack of experimental models of severe obesity in female mice limits discovery-based studies. Here, via utility of thermoneutral housing (30 °C) and high-fat diet (HFD) feeding, we induced severe obesity and metabolic disease in female C57BL/6 mice and compared their responses to severely obese male C57BL/6 counterparts during influenza virus infection. We show that lean male and female mice have similar lung edema, inflammation, and immune cell infiltration during influenza virus infection. At standard housing conditions, HFD-fed male, but not female, mice exhibit severe obesity, metabolic disease, and exacerbated influenza disease severity. However, combining thermoneutral housing and HFD feeding in female mice induces severe obesity and metabolic disease, which is sufficient to amplify influenza virus-driven disease severity to a level comparable to severely obese male counterparts. Lastly, increased total body weights of male and female mice at time of infection correlated with worsened influenza virus-driven disease severity metrics. Together, our findings confirm the impact of obesity and metabolic disease as key risk factors to influenza disease severity and present a novel mouse experimental model suitable for future mechanistic interrogation of sex, obesity, and metabolic disease traits in influenza virus-driven disease severity.
Collapse
Affiliation(s)
- Pablo C Alarcon
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Michelle S M A Damen
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Cassidy J Ulanowicz
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Keisuke Sawada
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Jarren R Oates
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; Molecular and Developmental Biology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Andrea Toth
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; Molecular and Developmental Biology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Jennifer L Wayland
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Hak Chung
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Traci E Stankiewicz
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Maria E Moreno-Fernandez
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Division of Gastroenterology, Hepatology and Nutrition Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Sara Szabo
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Division of Pathology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - William J Zacharias
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; Molecular and Developmental Biology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Senad Divanovic
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.
| |
Collapse
|
8
|
Awonuga AO, Camp OG, Abu-Soud HM. A review of nitric oxide and oxidative stress in typical ovulatory women and in the pathogenesis of ovulatory dysfunction in PCOS. Reprod Biol Endocrinol 2023; 21:111. [PMID: 37996893 PMCID: PMC10666387 DOI: 10.1186/s12958-023-01159-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 11/05/2023] [Indexed: 11/25/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is a heterogeneous functional endocrine disorder associated with a low-grade, chronic inflammatory state. Patients with PCOS present an increased risk of metabolic comorbidities and often menstrual dysregulation and infertility due to anovulation and/or poor oocyte quality. Multiple mechanisms including oxidative stress and low-grade inflammation are believed to be responsible for oocyte deterioration; however, the influence of nitric oxide (NO) insufficiency in oocyte quality and ovulatory dysfunction in PCOS is still a matter for debate. Higher production of superoxide (O2•-) mediated DNA damage and impaired antioxidant defense have been implicated as contributory factors for the development of PCOS, with reported alteration in superoxide dismutase (SOD) function, an imbalanced zinc/copper ratio, and increased catalase activity. These events may result in decreased hydrogen peroxide (H2O2) accumulation with increased lipid peroxidation events. A decrease in NO, potentially due to increased activity of NO synthase (NOS) inhibitors such as asymmetric dimethylarginine (ADMA), and imbalance in the distribution of reactive oxygen species (ROS), such as decreased H2O2 and increased O2•-, may offset the physiological processes surrounding follicular development, oocyte maturation, and ovulation contributing to the reproductive dysfunction in patients with PCOS. Thus, this proposal aims to evaluate the specific roles of NO, oxidative stress, ROS, and enzymatic and nonenzymatic elements in the pathogenesis of PCOS ovarian dysfunction, including oligo- anovulation and oocyte quality, with the intent to inspire better application of therapeutic options. The authors believe more consideration into the specific roles of oxidative stress, ROS, and enzymatic and nonenzymatic elements may allow for a more thorough understanding of PCOS. Future efforts elaborating on the role of NO in the preoptic nucleus to determine its influence on GnRH firing and follicle-stimulating hormone/Luteinizing hormone (FSH/LH) production with ovulation would be of benefit in PCOS. Consequently, treatment with an ADMA inhibitor or NO donor may prove beneficial to PCOS patients experiencing reproductive dysfunction and infertility.
Collapse
Affiliation(s)
- Awoniyi O Awonuga
- Departments of Obstetrics and Gynecology and Biochemistry and Molecular Biology, The C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, 275 E. Hancock Detroit, Detroit, MI, 48201, USA.
| | - Olivia G Camp
- Departments of Obstetrics and Gynecology and Biochemistry and Molecular Biology, The C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, 275 E. Hancock Detroit, Detroit, MI, 48201, USA
| | - Husam M Abu-Soud
- Departments of Obstetrics and Gynecology and Biochemistry and Molecular Biology, The C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, 275 E. Hancock Detroit, Detroit, MI, 48201, USA
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, 48201, USA
- Department of Microbiology, Immunology and Biochemistry, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| |
Collapse
|
9
|
Gu J, Zhang J, Liu Q, Xu S. Neurological risks of COVID-19 in women: the complex immunology underpinning sex differences. Front Immunol 2023; 14:1281310. [PMID: 38035090 PMCID: PMC10685449 DOI: 10.3389/fimmu.2023.1281310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 10/31/2023] [Indexed: 12/02/2023] Open
Abstract
The COVID-19 pandemic has uncovered many mysteries about SARS-CoV-2, including its potential to trigger abnormal autoimmune responses. Emerging evidence suggests women may face higher risks from COVID-induced autoimmunity manifesting as persistent neurological symptoms. Elucidating the mechanisms underlying this female susceptibility is now imperative. We synthesize key insights from existing studies on how COVID-19 infection can lead to immune tolerance loss, enabling autoreactive antibodies and lymphocyte production. These antibodies and lymphocytes infiltrate the central nervous system. Female sex hormones like estrogen and X-chromosome mediated effects likely contribute to dysregulated humoral immunity and cytokine profiles among women, increasing their predisposition. COVID-19 may also disrupt the delicate immunological balance of the female microbiome. These perturbations precipitate damage to neural damage through mechanisms like demyelination, neuroinflammation, and neurodegeneration - consistent with the observed neurological sequelae in women. An intentional focus on elucidating sex differences in COVID-19 pathogenesis is now needed to inform prognosis assessments and tailored interventions for female patients. From clinical monitoring to evaluating emerging immunomodulatory therapies, a nuanced women-centered approach considering the hormonal status and immunobiology will be vital to ensure equitable outcomes. Overall, deeper insights into the apparent female specificity of COVID-induced autoimmunity will accelerate the development of solutions mitigating associated neurological harm.
Collapse
Affiliation(s)
- Jienan Gu
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jiale Zhang
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qianhui Liu
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shijie Xu
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
10
|
Joshi NP, Madiwale SD, Sundrani DP, Joshi SR. Fatty acids, inflammation and angiogenesis in women with gestational diabetes mellitus. Biochimie 2023; 212:31-40. [PMID: 37059350 DOI: 10.1016/j.biochi.2023.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 03/01/2023] [Accepted: 04/11/2023] [Indexed: 04/16/2023]
Abstract
Gestational diabetes mellitus (GDM) is a metabolic disorder in pregnancy whose prevalence is on the rise. Reports suggest a likely association between inflammation and maternal GDM. A balance between pro and anti-inflammatory cytokines is necessary for the regulation of maternal inflammation system throughout pregnancy. Along with various inflammatory markers, fatty acids also act as pro-inflammatory molecules. However, studies reporting the role of inflammatory markers in GDM are contradictory, suggesting the need of more studies to better understand the role of inflammation in pregnancies complicated by GDM. Inflammatory response can be regulated by angiopoietins suggesting a link between inflammation and angiogenesis. Placental angiogenesis is a normal physiological process which is tightly regulated during pregnancy. Various pro and anti-angiogenic factors influence the regulation of the feto-placental vascular development. Studies evaluating the levels of angiogenic markers in women with GDM are limited and the findings are inconsistent. This review summarizes the available literature on fatty acids, inflammatory markers and angiogenesis in women with GDM. We also discuss the possible link between them and their influence on placental development in GDM.
Collapse
Affiliation(s)
- Nikita P Joshi
- Mother and Child Health, Interactive Research School for Health Affairs, Bharati Vidyapeeth University, Pune, India
| | - Shweta D Madiwale
- Mother and Child Health, Interactive Research School for Health Affairs, Bharati Vidyapeeth University, Pune, India
| | - Deepali P Sundrani
- Mother and Child Health, Interactive Research School for Health Affairs, Bharati Vidyapeeth University, Pune, India
| | - Sadhana R Joshi
- Mother and Child Health, Interactive Research School for Health Affairs, Bharati Vidyapeeth University, Pune, India.
| |
Collapse
|
11
|
Xu S, Fan Y, Tan Y, Zhang L, Li X. Association between blood lipid levels and risk of gastric cancer: A systematic review and meta-analysis. PLoS One 2023; 18:e0288111. [PMID: 37418353 DOI: 10.1371/journal.pone.0288111] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 06/20/2023] [Indexed: 07/09/2023] Open
Abstract
OBJECTIVE The association between blood lipid levels and the risk of gastric cancer (GC) is well known. Therefore, to clarify this association, all relevant prospective cohort studies were included in this meta-analysis. METHODS Our study was registered in PROSPERO (CRD42022354899) prior to its commencement. A systematic review and meta-analysis were conducted in accordance with the PRISMA recommendations. Chinese databases (CNKI, CBM, Wanfang, and VIP) and English databases (PubMed, Embase, Web of Science, and the Cochrane Library) were systematically searched up to October 2022. This study included all relevant cohort studies that reported hazard ratios (HRs) or relative risks (RRs) and their corresponding 95% confidence intervals (95% CIs) to examine the association between various lipid profiles (e.g., total cholesterol, triglycerides, high-density lipoprotein cholesterol, and low-density lipoprotein cholesterol) and the risk of developing gastric cancer (GC). Fixed effects or random effects models were used based on the level of heterogeneity among the studies, and these models were employed to obtain pooled hazard ratios. Additionally, sensitivity analysis and publication bias analysis were conducted to ensure the robustness and reliability of the findings. RESULTS After conducting a systematic search, a total of 10 studies were selected out of 10,525 papers involving a total of 5,564,520 individuals. Among these individuals, there were 41,408 GC cases. The analysis revealed that the highest versus lowest serum total cholesterol (TC) concentration was associated with a pooled hazard ratio of 0.89 (95% CI = 0.87-0.92, I2 = 15%). For triglycerides (TGs), the hazard ratio was 1.00 (95% CI = 0.96-1.04, I2 = 37%), while for high-density lipoprotein cholesterol (HDL-C), the hazard ratio was 0.90 (95% CI = 0.86-0.93, I2 = 0%). The hazard ratio for low-density lipoprotein cholesterol (LDL-C) was 0.96 (95% CI = 0.91-1.00, I2 = 0%). CONCLUSIONS Based on the results of this meta-analysis, it was found that serum TC and HDL-C levels were inversely correlated with the risk of GC. No association was observed between serum TG levels and the risk of GC. Similarly, no association was found between serum LDL-C levels and the risk of GC.
Collapse
Affiliation(s)
- Shicong Xu
- Department of Gastrointestinal surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Ying Fan
- Department of Gastrointestinal surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yuyue Tan
- Department of Gastrointestinal surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Ling Zhang
- Department of Gastrointestinal surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xianrong Li
- Department of Gastrointestinal surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
12
|
Miller RAJ, Williams AP, Kovats S. Sex chromosome complement and sex steroid signaling underlie sex differences in immunity to respiratory virus infection. Front Pharmacol 2023; 14:1150282. [PMID: 37063266 PMCID: PMC10097973 DOI: 10.3389/fphar.2023.1150282] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 03/08/2023] [Indexed: 04/18/2023] Open
Abstract
Epidemiological studies have revealed sex differences in the incidence and morbidity of respiratory virus infection in the human population, and often these observations are correlated with sex differences in the quality or magnitude of the immune response. Sex differences in immunity and morbidity also are observed in animal models of respiratory virus infection, suggesting differential dominance of specific immune mechanisms. Emerging research shows intrinsic sex differences in immune cell transcriptomes, epigenomes, and proteomes that may regulate human immunity when challenged by viral infection. Here, we highlight recent research into the role(s) of sex steroids and X chromosome complement in immune cells and describe how these findings provide insight into immunity during respiratory virus infection. We focus on the regulation of innate and adaptive immune cells by receptors for androgen and estrogens, as well as genes with a propensity to escape X chromosome inactivation. A deeper mechanistic knowledge of these pathways will help us to understand the often significant sex differences in immunity to endemic or pandemic respiratory pathogens such as influenza viruses, respiratory syncytial viruses and pathogenic coronaviruses.
Collapse
Affiliation(s)
- Reegan A. J. Miller
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Abigael P. Williams
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Susan Kovats
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| |
Collapse
|
13
|
Su X, Jin K, Zhou X, Zhang Z, Zhang C, Li Y, Yang M, Huang X, Xu S, Wei Q, Cheng X, Yang L, Qiu S. The association between sex hormones and periodontitis among American adults: A cross-sectional study. Front Endocrinol (Lausanne) 2023; 14:1125819. [PMID: 36864844 PMCID: PMC9971556 DOI: 10.3389/fendo.2023.1125819] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 01/30/2023] [Indexed: 02/16/2023] Open
Abstract
Introduction After adulthood, as a person grows older, the secretion of sex hormones in the body gradually decreases, and the risk of periodontitis increases. But the relationship between sex hormones and periodontitis is still controversial. Methods We investigated the association between sex hormones and periodontitis among Americans over 30 years old. 4,877 participants containing 3,222 males and 1,655 postmenopausal females who had had periodontal examination and detailed available sex hormone levels, were included in our analysis from the 2009-2014 National Health and Nutrition Examination Surveys cycles. We applied multivariate linear regression models to estimate the connection between sex hormones and periodontitis after converting sex hormones into categorical variables through tertile. Additionally, to ensure the stability of the analysis results, we carried out a trend test, subgroup analysis, and interaction test. Results After fully adjusting the covariates, estradiol levels were not associated with periodontitis in both males and females with a P for trend = 0.064 and 0.064, respectively. For males, we found that sex hormone-binding globulin was positively associated with periodontitis (tertile3 vs tertile1: OR=1.63, 95% CI=1.17-2.28, p = 0.004, P for trend = 0.005). Congruously, free testosterone (tertile3 vs tertile1: OR=0.60, 95% CI=0.43-0.84, p = 0.003), bioavailable testosterone (tertile3 vs tertile1: OR=0.51, 95% CI=0.36-0.71, p < 0.001), and free androgen index (tertile3 vs tertile1: OR=0.53, 95% CI=0.37-0.75, p < 0.001) was found to be negatively associated with periodontitis. Moreover, subgroup analysis of age found a closer relationship between sex hormones and periodontitis in those younger than 50 years. Conclusion Our research suggested that males with lower bioavailable testosterone levels affected by sex hormone-binding globulin were at a higher risk of periodontitis. Meanwhile, estradiol levels were not associated with periodontitis in postmenopausal women.
Collapse
Affiliation(s)
- Xingyang Su
- Department of Urology, Institute of Urology and National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Kun Jin
- Department of Urology, the First Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Xianghong Zhou
- Department of Urology, Institute of Urology and National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Zilong Zhang
- Department of Urology, Institute of Urology and National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Chichen Zhang
- Department of Urology, Institute of Urology and National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Yifan Li
- Department of Urology, Institute of Urology and National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Mi Yang
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Xinyi Huang
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Shishi Xu
- Division of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
| | - Qiang Wei
- Department of Urology, Institute of Urology and National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Xu Cheng
- State Key Laboratory of Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Lu Yang
- Department of Urology, Institute of Urology and National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Shi Qiu
- Department of Urology, Institute of Urology and National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
- Center of Biomedical big data, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
14
|
Martin BE, Taylor EB, Attipoe EM, Wu W, Stec DE, Showmaker KC, Garrett MR. Sex and molecular differences in cardiovascular parameters at peak influenza disease in mice. Physiol Genomics 2023; 55:79-89. [PMID: 36645670 PMCID: PMC9925171 DOI: 10.1152/physiolgenomics.00146.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 12/29/2022] [Accepted: 01/09/2023] [Indexed: 01/17/2023] Open
Abstract
There is a growing interest in the detection of subtle changes in cardiovascular physiology in response to viral infection to develop better disease surveillance strategies. This is not only important for earlier diagnosis and better prognosis of symptomatic carriers but also useful to diagnose asymptomatic carriers of the virus. Previous studies provide strong evidence of an association between inflammatory biomarker levels and both blood pressure (BP) and heart rate (HR) during infection. The identification of novel biomarkers during an inflammatory event could significantly improve predictions for cardiovascular events. Thus, we evaluated changes in cardiovascular physiology induced in A/Puerto Rico/8/34 (PR8) influenza infections in female and male C57BL/6J mice and compared them with the traditional method of influenza disease detection using body weight (BW). Using radiotelemetry, changes in BP, HR, and activity were studied. Change in BW of infected females was significantly decreased from 5 to 13 days postinfection (dpi), yet alterations in normal physiology including loss of diurnal rhythm and reduced activity was observed starting at about 3 dpi for HR and 4 dpi for activity and BP; continuing until about 13 dpi. In contrast, males had significantly decreased BW 8 to 12 dpi and demonstrated altered physiological measurements for a shorter period compared with females with a reduction starting at 5 dpi for activity, 6 dpi for BP, and 7 dpi for HR until about 12 dpi, 10 dpi, and 9 dpi, respectively. Finally, females and males exhibited different patterns of inflammatory maker expression in lungs at peak disease by analyzing bulk RNA-sequencing data for lungs and Bio-plex cytokine assay for blood collected from influenza-infected and naïve C57BL/6J female and male mice at 7 dpi. In total, this study provides insight into cardiovascular changes and molecular markers to distinguish sex differences in peak disease caused by influenza virus infection.NEW & NOTEWORTHY This study performed longitudinal cardiovascular measurements of influenza viral infection and identified sex difference in both physiological and molecular markers at peak disease.
Collapse
Affiliation(s)
- Brigitte E Martin
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Erin B Taylor
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| | - Esinam M Attipoe
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Wenjie Wu
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - David E Stec
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| | | | - Michael R Garrett
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
- Division of Nephrology, Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi
- Division of Genetics, Department of Pediatrics, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
15
|
Shields CA, Wang X, Cornelius DC. Sex differences in cardiovascular response to sepsis. Am J Physiol Cell Physiol 2023; 324:C458-C466. [PMID: 36571442 PMCID: PMC9902216 DOI: 10.1152/ajpcell.00134.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 12/20/2022] [Accepted: 12/20/2022] [Indexed: 12/27/2022]
Abstract
Recently, there has been increased recognition of the importance of sex as a biological factor affecting disease and health. Many preclinical studies have suggested that males may experience a less favorable outcome in response to sepsis than females. The underlying mechanisms for these differences are still largely unknown but are thought to be related to the beneficial effects of estrogen. Furthermore, the immunosuppressive role of testosterone is also thought to contribute to the sex-dependent differences that are present in clinical sepsis. There are still significant knowledge gaps in this field. This mini-review will provide a brief overview of sex-dependent variables in relation to sepsis and the cardiovascular system. Preclinical animal models for sepsis research will also be discussed. The intent of this mini-review is to inspire interest for future considerations of sex-related variables in sepsis that should be addressed to increase our understanding of the underlying mechanisms in sepsis-induced cardiovascular dysfunction for the identification of therapeutic targets and improved sepsis management and treatment.
Collapse
Affiliation(s)
- Corbin A Shields
- Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Xi Wang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Denise C Cornelius
- Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
16
|
Etchevers L, Stassi A, Belotti E, Diaz P, Durante L, Notaro U, Chiaraviglio J, Rey F, Salvetti N, Ortega H, Amweg A. Exogenous ACTH stimulus during the preovulatory period alters patterns of leukocyte recruitment in the ovary of dairy cows. Theriogenology 2023; 195:176-186. [DOI: 10.1016/j.theriogenology.2022.10.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 09/02/2022] [Accepted: 10/21/2022] [Indexed: 11/06/2022]
|
17
|
Giotis ES, Cil E, Brooke GN. Use of Antiandrogens as Therapeutic Agents in COVID-19 Patients. Viruses 2022; 14:2728. [PMID: 36560732 PMCID: PMC9788624 DOI: 10.3390/v14122728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/23/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
COVID-19, caused by the severe acute respiratory syndrome coronavirus 2 (SARS CoV-2), is estimated to have caused over 6.5 million deaths worldwide. The emergence of fast-evolving SARS-CoV-2 variants of concern alongside increased transmissibility and/or virulence, as well as immune and vaccine escape capabilities, highlight the urgent need for more effective antivirals to combat the disease in the long run along with regularly updated vaccine boosters. One of the early risk factors identified during the COVID-19 pandemic was that men are more likely to become infected by the virus, more likely to develop severe disease and exhibit a higher likelihood of hospitalisation and mortality rates compared to women. An association exists between SARS-CoV-2 infectiveness and disease severity with sex steroid hormones and, in particular, androgens. Several studies underlined the importance of the androgen-mediated regulation of the host protease TMPRSS2 and the cell entry protein ACE2, as well as the key role of these factors in the entry of the virus into target cells. In this context, modulating androgen signalling is a promising strategy to block viral infection, and antiandrogens could be used as a preventative measure at the pre- or early hospitalisation stage of COVID-19 disease. Different antiandrogens, including commercial drugs used to treat metastatic castration-sensitive prostate cancer and other conditions, have been tested as antivirals with varying success. In this review, we summarise the most recent updates concerning the use of antiandrogens as prophylactic and therapeutic options for COVID-19.
Collapse
Affiliation(s)
- Efstathios S. Giotis
- Department of Infectious Diseases, Imperial College London, London W2 1PG, UK
- School of Life Sciences, University of Essex, Colchester CO4 3SQ, UK
| | - Emine Cil
- School of Life Sciences, University of Essex, Colchester CO4 3SQ, UK
| | - Greg N. Brooke
- School of Life Sciences, University of Essex, Colchester CO4 3SQ, UK
| |
Collapse
|
18
|
Lombardo G, Nettis MA, Hastings C, Zajkowska Z, Mariani N, Nikkheslat N, Worrell C, Enache D, McLaughlin A, Kose M, Bogdanova A, Sforzini L, Cleare AJ, Young AH, Dazzan P, Mondelli V, Pariante CM. Sex differences in a double-blind randomized clinical trial with minocycline in treatment-resistant depressed patients: CRP and IL-6 as sex-specific predictors of treatment response. Brain Behav Immun Health 2022; 26:100561. [PMID: 36467125 PMCID: PMC9712814 DOI: 10.1016/j.bbih.2022.100561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 11/13/2022] [Indexed: 11/18/2022] Open
Abstract
Background Inflammation is a well-known risk factor for depression. Specifically, patients who do not respond to antidepressant treatment show higher levels of inflammatory biomarkers compared with responders. Thus, several studies have investigated the efficacy of anti-inflammatory add-on treatment in this population. However, major depressive disorder is more prevalent in females than in males, with sex differences present in antidepressant treatment response and in immune system regulation. To explore sex differences in inflammatory profiles and treatment responses, we investigated a cohort of patients with treatment resistant depression (TRD), for which they received an adjunctive, anti-inflammatory treatment with minocycline - the Minocycline in Depression (MINDEP) study. Methods The MINDEP study is a 4-week double-blind, randomised, placebo-controlled clinical trial (stratified by sex) with 39 TRD participants, which demonstrated the efficacy of minocycline, an antibiotic with anti-inflammatory properties, in TRD patients with major depressive disorder (MDD) and evidence of low-grade inflammation measured with C-reactive protein (CRP) ≥ 3 mg/L. In these secondary analyses, we investigated the differential effects of minocycline in females (N = 22, 10 randomised to minocycline and 12 randomised to placebo) and in males (N = 17, 8 randomised to minocycline and 9 randomised to placebo) on changes in depressive symptoms (Δ- Hamilton Rating Scale for Depression (HAMD)-17), taking also into consideration CRP levels (CRP ≥3 mg/L vs. CRP <3 mg/L). Additionally, we investigated the role of serum IL-6 in predicting treatment response to minocycline, using sex-specific medians of IL-6, in novel exploratory analyses. Results Sex differences in Δ-HAMD-17 indicate that only females (F = 10.49, p = 0.005), but not males (F = 1.64, p = 0.22), presented an effect of CRP levels on the response to minocycline. Also, we detected sex differences in the relationship between serum CRP and IL-6 levels: CRP was strongly correlated with IL-6 in females (Spearman's ρ = 0.658, P < 0.001) but not in males (ρ = 0.007, p = 0.979). Exploratory analyses found that IL-6 was indeed a better predictor of response than minocycline than CRP, as we found an interaction between study arms and IL-6 groups (above and below the IL-6 sex-specific median) in females (F = 4.435 p = 0.050) and, at trend statistical level, in males (F = 4.258 p = 0.060). Moreover, Δ-HAMD-17 was numerically comparable in the two high-IL-6 group taking minocycline (females, mean 9.20 ± SD 7.80; males, mean 8.80 ± SD 5.97), confirming that high IL-6, differently from high CRP, identified responders to minocycline both in males and females. Conclusion Our findings highlight the need of sex-specific inflammatory biomarkers in predicting antidepressant response to anti-inflammatories in TRD patients, with the possibility of CRP being a relevant predictor of treatment response only for females, and IL-6 being relevant for both sexes.
Collapse
Affiliation(s)
- Giulia Lombardo
- King's College London, Institute of Psychiatry Psychology and Neuroscience, Department of Psychological Medicine, London, UK
| | - Maria Antonietta Nettis
- King's College London, Institute of Psychiatry Psychology and Neuroscience, Department of Psychological Medicine, London, UK.,National Institute for Health Research Mental Health Biomedical Research Centre, South London and Maudsley NHS Foundation Trust and King's College London, London, UK
| | - Caitlin Hastings
- King's College London, Institute of Psychiatry Psychology and Neuroscience, Department of Psychological Medicine, London, UK
| | - Zuzanna Zajkowska
- King's College London, Institute of Psychiatry Psychology and Neuroscience, Department of Psychological Medicine, London, UK
| | - Nicole Mariani
- King's College London, Institute of Psychiatry Psychology and Neuroscience, Department of Psychological Medicine, London, UK
| | - Naghmeh Nikkheslat
- King's College London, Institute of Psychiatry Psychology and Neuroscience, Department of Psychological Medicine, London, UK
| | - Courtney Worrell
- King's College London, Institute of Psychiatry Psychology and Neuroscience, Department of Psychological Medicine, London, UK
| | - Daniela Enache
- King's College London, Institute of Psychiatry Psychology and Neuroscience, Department of Psychological Medicine, London, UK.,Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm, Sweden
| | - Anna McLaughlin
- King's College London, Institute of Psychiatry Psychology and Neuroscience, Department of Psychological Medicine, London, UK
| | - Melisa Kose
- King's College London, Institute of Psychiatry Psychology and Neuroscience, Department of Psychological Medicine, London, UK
| | - Anna Bogdanova
- King's College London, Institute of Psychiatry Psychology and Neuroscience, Department of Psychological Medicine, London, UK
| | - Luca Sforzini
- King's College London, Institute of Psychiatry Psychology and Neuroscience, Department of Psychological Medicine, London, UK
| | - Anthony J Cleare
- King's College London, Institute of Psychiatry Psychology and Neuroscience, Department of Psychological Medicine, London, UK.,National Institute for Health Research Mental Health Biomedical Research Centre, South London and Maudsley NHS Foundation Trust and King's College London, London, UK
| | - Allan H Young
- King's College London, Institute of Psychiatry Psychology and Neuroscience, Department of Psychological Medicine, London, UK.,National Institute for Health Research Mental Health Biomedical Research Centre, South London and Maudsley NHS Foundation Trust and King's College London, London, UK
| | - Paola Dazzan
- King's College London, Institute of Psychiatry Psychology and Neuroscience, Department of Psychological Medicine, London, UK.,National Institute for Health Research Mental Health Biomedical Research Centre, South London and Maudsley NHS Foundation Trust and King's College London, London, UK
| | - Valeria Mondelli
- King's College London, Institute of Psychiatry Psychology and Neuroscience, Department of Psychological Medicine, London, UK.,National Institute for Health Research Mental Health Biomedical Research Centre, South London and Maudsley NHS Foundation Trust and King's College London, London, UK
| | - Carmine M Pariante
- King's College London, Institute of Psychiatry Psychology and Neuroscience, Department of Psychological Medicine, London, UK.,National Institute for Health Research Mental Health Biomedical Research Centre, South London and Maudsley NHS Foundation Trust and King's College London, London, UK
| |
Collapse
|
19
|
Kumar N, Vyas A, Agnihotri SK, Chattopadhyay N, Sachdev M. Small secretory proteins of immune cells can modulate gynecological cancers. Semin Cancer Biol 2022; 86:513-531. [PMID: 35150864 DOI: 10.1016/j.semcancer.2022.02.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 02/04/2022] [Accepted: 02/05/2022] [Indexed: 01/27/2023]
Abstract
Small secretory proteins of immune cells are mostly Cytokines, which include chemokines, interleukins, interferons, lymphokines and tumor necrosis factors but not hormones or growth factors. These secretory proteins are the molecular messengers and primarily involved in autocrine, paracrine and endocrine signaling as immunomodulating agents. Hence, these proteins actually regulate the cells of immune system to communicate with one another to produce a synchronized, robust, still self-regulated response to a specific antigen. Chemokines are smaller secreted proteins that control overall immune cell movement and location; these chemokines are divided into 4 subgroups, namely, CXC, CC, CX3C and C according to the position of 4 conserved cysteine residues. Complete characterization of cytokines and chemokines can exploit their vast signaling networks to develop cancer treatments. These secretory proteins like IL-6, IL-10, IL-12, TNFα, CCL2, CXCL4 & CXCL8 are predominantly expressed in most of the gynecological cancers, which directly stimulate immune effector cells and stromal cells at the tumor site and augment tumor cell recognition by cytotoxic T-cells. Hence; these secretory proteins are the major regulators, which can actually modulate all kinds of gynecological cancers. Furthermore, advancements in adoptive T-cell treatment have relied on the use of multiple cytokines/chemokines to establish a highly regulated environment for anti-tumor T cell growth. A number of in vitro studies as well as animal models and clinical subjects have also shown that cytokines/chemokines have broad antitumor activity, which has been translated into a number of cancer therapy approaches. This review will focus on the foremost cytokines & chemokines involved in the majority of the gynecological malignancies and discuss their basic biology as well as clinical applications.
Collapse
Affiliation(s)
- Niranjan Kumar
- Division of Endocrinology, CSIR- Central Drug Research Institute, Lucknow, 226 031, India; Academy of Scientific and Innovative Research (AcSIR), Sector 19, Kamla Nehru Nagar, Ghaziabad, 201 002, India
| | - Akanksha Vyas
- Division of Endocrinology, CSIR- Central Drug Research Institute, Lucknow, 226 031, India; Academy of Scientific and Innovative Research (AcSIR), Sector 19, Kamla Nehru Nagar, Ghaziabad, 201 002, India
| | | | - Naibedya Chattopadhyay
- Division of Endocrinology, CSIR- Central Drug Research Institute, Lucknow, 226 031, India; Academy of Scientific and Innovative Research (AcSIR), Sector 19, Kamla Nehru Nagar, Ghaziabad, 201 002, India.
| | - Monika Sachdev
- Division of Endocrinology, CSIR- Central Drug Research Institute, Lucknow, 226 031, India; Academy of Scientific and Innovative Research (AcSIR), Sector 19, Kamla Nehru Nagar, Ghaziabad, 201 002, India.
| |
Collapse
|
20
|
Lee SA, Kwon SO, Park H, Shu XO, Lee JK, Kang D. Association of serum high-sensitivity C reactive protein with risk of mortality in an Asian population: the Health Examinees cohort. BMJ Open 2022; 12:e052630. [PMID: 35788076 PMCID: PMC9255402 DOI: 10.1136/bmjopen-2021-052630] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVES This study aimed to examine the association of high-sensitivity C reactive protein (hsCRP) with mortality risk and the attenuated effect of non-communicable disease history (NCD history ) on the association. DESIGN Prospective cohort study. SETTING Health Examinees cohort. PARTICIPANTS A total of 41 070 men and 81 011 women aged ≥40 years were involved (follow-up: 6.8 years). OUTCOME MEASURES Data and cause of death occurring until 31 December 2015 were confirmed by death statistics from the National Statistical Office. We conducted advanced analysis after stratification by NCD history and sensitivity analysis after excluding death before 1 or 2 years from recruitment. Cox proportional hazard and restricted cubic spline models were used to assess the association. RESULTS The association between serum hsCRP and risk of all-cause mortality was observed with strong linearity in both genders and was not influenced by NCD history . The association of serum hsCRP with risk of cancer mortality was not observed in women with NCD history , but the association with risk of cardiovascular disease (CVD) mortality was predominantly observed in men with NCD history . CONCLUSIONS This study suggests a dose-response association of hsCRP with mortality risk, including cancer and CVD mortality, in Koreans with low serum hsCRP, although the association with cancer and CVD mortality risk could be influenced by gender and NCD history .
Collapse
Affiliation(s)
- Sang-Ah Lee
- Preventive Medicine, Kangwon National University School of Medicine, Chuncheon, Republic of Korea
- Division of Epidemiology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Sung Ok Kwon
- Preventive Medicine, Kangwon National University School of Medicine, Chuncheon, Republic of Korea
| | - Hyerim Park
- Preventive Medicine, Kangwon National University School of Medicine, Chuncheon, Republic of Korea
| | - Xiao-Ou Shu
- Division of Epidemiology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jong-Koo Lee
- JW LEE Center for Global Medicine, Seoul, Republic of Korea
| | - Daehee Kang
- Preventive Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
21
|
Hart DA, Nakamura N. Creating an Optimal In Vivo Environment to Enhance Outcomes Using Cell Therapy to Repair/Regenerate Injured Tissues of the Musculoskeletal System. Biomedicines 2022; 10:1570. [PMID: 35884875 PMCID: PMC9313221 DOI: 10.3390/biomedicines10071570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 06/20/2022] [Accepted: 06/27/2022] [Indexed: 11/16/2022] Open
Abstract
Following most injuries to a musculoskeletal tissue which function in unique mechanical environments, an inflammatory response occurs to facilitate endogenous repair. This is a process that usually yields functionally inferior scar tissue. In the case of such injuries occurring in adults, the injury environment no longer expresses the anabolic processes that contributed to growth and maturation. An injury can also contribute to the development of a degenerative process, such as osteoarthritis. Over the past several years, researchers have attempted to use cellular therapies to enhance the repair and regeneration of injured tissues, including Platelet-rich Plasma and mesenchymal stem/medicinal signaling cells (MSC) from a variety of tissue sources, either as free MSC or incorporated into tissue engineered constructs, to facilitate regeneration of such damaged tissues. The use of free MSC can sometimes affect pain symptoms associated with conditions such as OA, but regeneration of damaged tissues has been challenging, particularly as some of these tissues have very complex structures. Therefore, implanting MSC or engineered constructs into an inflammatory environment in an adult may compromise the potential of the cells to facilitate regeneration, and neutralizing the inflammatory environment and enhancing the anabolic environment may be required for MSC-based interventions to fulfill their potential. Thus, success may depend on first eliminating negative influences (e.g., inflammation) in an environment, and secondly, implanting optimally cultured MSC or tissue engineered constructs into an anabolic environment to achieve the best outcomes. Furthermore, such interventions should be considered early rather than later on in a disease process, at a time when sufficient endogenous cells remain to serve as a template for repair and regeneration. This review discusses how the interface between inflammation and cell-based regeneration of damaged tissues may be at odds, and outlines approaches to improve outcomes. In addition, other variables that could contribute to the success of cell therapies are discussed. Thus, there may be a need to adopt a Precision Medicine approach to optimize tissue repair and regeneration following injury to these important tissues.
Collapse
Affiliation(s)
- David A. Hart
- Department of Surgery, Faculty of Kinesiology, McCaig Institute for Bone & Joint Health, University of Calgary, Calgary, AB T2N 4N1, Canada
- Bone & Joint Health Strategic Clinical Network, Alberta Health Services, Edmonton, AB T5J 3E4, Canada
| | - Norimasa Nakamura
- Institute of Medical Science in Sport, Osaka Health Science University, 1-9-27 Tenma, Kita-ku, Osaka 530-0043, Japan;
| |
Collapse
|
22
|
Singh LK, Pandey M, Baithalu RK, Fernandes A, Ali SA, Jaiswal L, Pannu S, Neeraj, Mohanty TK, Kumaresan A, Datta TK, Kumar S, Mohanty AK. Comparative Proteome Profiling of Saliva Between Estrus and Non-Estrus Stages by Employing Label-Free Quantitation (LFQ) and Tandem Mass Tag (TMT)-LC-MS/MS Analysis: An Approach for Estrus Biomarker Identification in Bubalus bubalis. Front Genet 2022; 13:867909. [PMID: 35754844 PMCID: PMC9217162 DOI: 10.3389/fgene.2022.867909] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 04/05/2022] [Indexed: 02/05/2023] Open
Abstract
Accurate determination of estrus is essentially required for efficient reproduction management of farm animals. Buffalo is a shy breeder and does not manifest overt signs of estrus that make estrus detection difficult resulting in a poor conception rate. Therefore, identifying estrus biomarkers in easily accessible biofluid such as saliva is of utmost interest. In the current study, we generated saliva proteome profiles during proestrus (PE), estrus (E), metestrus (ME), and diestrus (DE) stages of the buffalo estrous cycle using both label-free quantitation (LFQ) and labeled (TMT) quantitation and mass spectrometry analysis. A total of 520 proteins were identified as DEPs in LFQ; among these, 59 and four proteins were upregulated (FC ≥ 1.5) and downregulated (FC ≤ 0.5) during E vs. PE, ME, and DE comparisons, respectively. Similarly, TMT-LC-MS/MS analysis identified 369 DEPs; among these, 74 and 73 proteins were upregulated and downregulated during E vs. PE, ME, and DE stages, respectively. Functional annotations of GO terms showed enrichment of glycolysis, pyruvate metabolism, endopeptidase inhibitor activity, salivary secretion, innate immune response, calcium ion binding, oocyte meiosis, and estrogen signaling. Over-expression of SERPINB1, HSPA1A, VMO1, SDF4, LCN1, OBP, and ENO3 proteins during estrus was further confirmed by Western blotting. This is the first comprehensive report on differential proteome analysis of buffalo saliva between estrus and non-estrus stages. This study generated an important panel of candidate proteins that may be considered buffalo estrus biomarkers which can be applied in the development of a diagnostic kit for estrus detection in buffalo.
Collapse
|
23
|
Infrared thermography as valuable tool for gynoid lipodystrophy (cellulite) diagnosis. Lasers Med Sci 2022; 37:2639-2644. [PMID: 35237888 DOI: 10.1007/s10103-022-03530-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 02/18/2022] [Indexed: 10/19/2022]
Abstract
Cellulite is a morphological alteration of the tegument tissue, directly interfering in self-esteem with etiology and pathophysiology far from being a consensus. Although the visual diagnosis of cellulitis is well known, it does not represent the real pathological condition of the subcutaneous tissue. The aim of the study was to investigate the hypothesis that the more heterogeneous tissue pattern analyzed by infrared thermography, the more severe is the cellulite grade. Forty female participants were selected and 60 thighs were analyzed by clinical anamnesis and infrared thermography. Classical visual analysis was correlated to the tissue heterogeneity measured by thermography. R Spearman's correlation between visual evaluation and thermography was 0.92. Phototype presented a negative significant correlation of 0.67 with classical visual analysis. In the present study, we presented a simple method based on infrared thermography that can be adopted in any esthetics office with a correlation of 0.92 with the visual classic evaluation, but, besides, may be very helpful to the clinician to decide which treatment will be adopted, i.e., an aggressive and inflammatory approach such as the radiofrequency of shockwave therapy or an anti-inflammatory approach such as photobiomodulation, depending on the inflammatory status of cellulite.
Collapse
|
24
|
Gonen MS, De Bellis A, Durcan E, Bellastella G, Cirillo P, Scappaticcio L, Longo M, Bircan BE, Sahin S, Sulu C, Ozkaya HM, Konukoglu D, Kartufan FF, Kelestimur F. Assessment of Neuroendocrine Changes and Hypothalamo-Pituitary Autoimmunity in Patients with COVID-19. Horm Metab Res 2022; 54:153-161. [PMID: 35276740 DOI: 10.1055/a-1764-1260] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
SARS-CoV-2 may affect the hypothalamic-pituitary axis and pituitary dysfunction may occur. Therefore, we investigated neuroendocrine changes, in particular, secondary adrenal insufficiency, using a dynamic test and the role of autoimmunity in pituitary dysfunction in patients with COVID-19. The single-center, prospective, case-control study included patients with polymerase chain reaction (PCR)-confirmed COVID-19 and healthy controls. Basal hormone levels were measured, and the adrenocorticotropic hormone (ACTH) stimulation test was performed. Antipituitary (APA) and antihypothalamic antibodies (AHA) were also determined. We examined a total of 49 patients with COVID-19 and 28 healthy controls. The frequency of adrenal insufficiency in patients with COVID-19 was found as 8.2%. Patients with COVID-19 had lower free T3, IGF-1, and total testosterone levels, and higher cortisol and prolactin levels when compared with controls. We also demonstrated the presence of APA in three and AHA in one of four patients with adrenal insufficiency. In conclusion, COVID-19 may result in adrenal insufficiency, thus routine screening of adrenal functions in these patients is needed. Endocrine disturbances in COVID-19 are similar to those seen in acute stressful conditions or infections. Pituitary or hypothalamic autoimmunity may play a role in neuroendocrine abnormalities in COVID-19.
Collapse
Affiliation(s)
- Mustafa Sait Gonen
- Division of Endocrinology and Metabolism, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Annamaria De Bellis
- Unit of Endocrinology and Metabolic Diseases, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Emre Durcan
- Division of Endocrinology and Metabolism, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Giuseppe Bellastella
- Unit of Endocrinology and Metabolic Diseases, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Paolo Cirillo
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Lorenzo Scappaticcio
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Miriam Longo
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Basak Ecem Bircan
- Division of Endocrinology and Metabolism, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Serdar Sahin
- Division of Endocrinology and Metabolism, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Cem Sulu
- Division of Endocrinology and Metabolism, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Hande Mefkure Ozkaya
- Division of Endocrinology and Metabolism, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Dildar Konukoglu
- Department of Medical Biochemistry, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Fatma Ferda Kartufan
- Department of Anaesthesiology and Reanimation, Yeditepe University, Istanbul, Turkey
| | | |
Collapse
|
25
|
Singh A, Singh R, Tripathi MK. Evaluation of the sex steroids mediated modulation of leucocyte immune responses in an ophidian Natrix piscator. Curr Res Physiol 2022; 5:355-360. [PMID: 36185818 PMCID: PMC9519393 DOI: 10.1016/j.crphys.2022.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 08/23/2022] [Accepted: 09/20/2022] [Indexed: 11/28/2022] Open
Abstract
The immune-suppressive role of sex steroids in mammals is well documented, but information on other vertebrates is limited. The present study was planned to analyze the effect of testosterone and progesterone in the modulation of immune functions of leucocytes in a reptile, Natrix piscator. Reptiles are unique organisms and this study is novel in that it provides an insight into immune-reproductive cross-talk in a reptile. Leucocytes were isolated from peripheral blood, cultured with different concentrations of testosterone and progesterone and different immune parameters like phagocytosis, superoxide production, and nitrite release were assessed. Lymphocytes were isolated and cell-mediated immunity was assessed through proliferation responses utilizing tetrazolium salt. Concentration-dependent suppressive effects of both the steroids on immune responses were observed. A differential suppressive effect of testosterone was also observed when a lymphocyte proliferation assay was studied. Using receptor antagonists such as cyproterone acetate and mifepristone restored the immune responses of cultured cells. It was summarized that gonadal steroids mediate a direct suppressive effect on innate and cell-mediated immune responses of blood immune cells. It was concluded that when gonadal steroids are high in reproductive seasons, the immune functions are suppressed to gain optimum reproductive success. Reptilian immune responses are differentially affected by sex steroids. Testosterone interacts with nuclear receptors and suppress proliferative responses of lymphocytes. Superoxide anion production by blood immune cells are inhibited by gonadal steroids. Use of receptor antagonists resulted in amelioration of immune responses indicating the direct action of steroids.
Collapse
|
26
|
Acharya TK, Sahu RP, Kumar S, Kumar S, Rokade TP, Chakraborty R, Dubey NK, Shikha D, Chawla S, Goswami C. Function and regulation of thermosensitive ion channel TRPV4 in the immune system. CURRENT TOPICS IN MEMBRANES 2022; 89:155-188. [DOI: 10.1016/bs.ctm.2022.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
|
27
|
Coco G, Hamill KJ, Troughton LD, Kaye SB, Romano V. Risk factors for corneal epithelial wound healing: Can sex play a role? Eur J Ophthalmol 2021; 32:2676-2682. [PMID: 34889141 DOI: 10.1177/11206721211066705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE To determine if sex is associated with corneal epithelial wound healing time in patients with persistent corneal epithelial defects (PCEDs). METHODS Retrospective case series on patients with PCED from November 2014 to January 2019. Records of 127 patients with diagnosis of PCED were reviewed. Patients with an epithelial defect that lasted more than two weeks in the absence of an active corneal infection were included. Main outcome was corneal epithelial wound healing time. RESULTS 55 patients (29 males) with a mean age of 65.3 ± 16.5 years were included. No difference was found between female and male patients in terms of risk factors, age, treatment strategies or intervals between visits (median of 15 days in females and 12 days in males; p = 0.24). Median duration of the PCED was 51 days (IQR 32-130), with a median number of 5 clinical visits (IQR 4-8). Female patients had significantly longer healing times (p = 0.004) and a corresponding increase in the number of clinical visits (median of 7 visits vs. 5 clinical visits in males, p = 0.012). CONCLUSION Results from this study suggest female patients with PCED might have a longer corneal epithelial wound healing duration and may therefore require earlier intervention.
Collapse
Affiliation(s)
- Giulia Coco
- 159020The Royal Liverpool University Hospital, Liverpool, UK.,Department of Clinical Science and Translational Medicine, 60259University of Rome Tor Vergata, Rome, Italy
| | - Kevin J Hamill
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, 4591University of Liverpool, Liverpool, UK
| | - Lee D Troughton
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, 4591University of Liverpool, Liverpool, UK
| | - Stephen B Kaye
- 159020The Royal Liverpool University Hospital, Liverpool, UK.,Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, 4591University of Liverpool, Liverpool, UK
| | - Vito Romano
- 159020The Royal Liverpool University Hospital, Liverpool, UK.,Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, 4591University of Liverpool, Liverpool, UK
| |
Collapse
|
28
|
Montanaro M, Scimeca M, Toschi N, Bonanno E, Giacobbi E, Servadei F, Ippoliti A, Santeusanio G, Mauriello A, Anemona L. Effects of Risk Factors on In Situ Expression of Proinflammatory Markers Correlated to Carotid Plaque Instability. Appl Immunohistochem Mol Morphol 2021; 29:741-749. [PMID: 34039839 DOI: 10.1097/pai.0000000000000947] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 04/13/2021] [Indexed: 11/25/2022]
Abstract
BACKGROUND AND AIMS Several studies demonstrated a role of active chronic inflammatory infiltrate in carotid plaques progression suggesting a possible link between cardiovascular risk factors and inflammation-related plaque instability. The aim of this study is therefore to evaluate the possible effects of cardiovascular risk factors on in situ expression of proinflammatory markers associated with carotid plaque instability. METHODS AND RESULTS A tissue microarray containing carotid plaques from 36 symptomatic (major stroke or transient ischemic attack) and 37 asymptomatic patients was built. Serial sections were employed to evaluate the expression of some inflammatory markers by immunohistochemistry [CD3, CD4a, CD8, CD20, CD86, CD163, interleukin (IL)-2, IL-6, IL-17]. Immunohistochemical data were analyzed to study the possible associations between in situ expression of inflammatory biomarker and the main cardiovascular risk factors. Our data demonstrated that plaque instability is associated with the high in situ expression of some cytokines, such as IL-2, IL-6, IL-17. Besides the female sex, none of the risk factors analyzed showed a significant association between the in situ expression of these markers and unstable plaques. A significant increase of IL-6-positive and IL-17-positive cells was observed in unstable atheromatous plaques of female patients, as compared with unstable plaques of male patients. CONCLUSIONS Plaque destabilization is certainly correlated with the presence of the major cardiovascular risk factors, however, our results showed that, with the exception of sex, their action in the evolutive process of plaque instability seems rather nonspecific, favoring a general release of proinflammatory cytokines.
Collapse
Affiliation(s)
| | - Manuel Scimeca
- Departments of Experimental Medicine
- University of San Raffaele
- Saint Camillus International University of Health Sciences, Rome, Italy
| | - Nicola Toschi
- Biomedicine and Prevention, University of Rome "Tor Vergata"
- Imaging Martinos Center for Biomedical Imaging
- Harvard Medical School, Massachusetts General Hospital, Boston, MA
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Walker CJ, Schroeder ME, Aguado BA, Anseth KS, Leinwand LA. Matters of the heart: Cellular sex differences. J Mol Cell Cardiol 2021; 160:42-55. [PMID: 34166708 PMCID: PMC8571046 DOI: 10.1016/j.yjmcc.2021.04.010] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 04/12/2021] [Accepted: 04/24/2021] [Indexed: 02/06/2023]
Abstract
Nearly all cardiovascular diseases show sexual dimorphisms in prevalence, presentation, and outcomes. Until recently, most clinical trials were carried out in males, and many animal studies either failed to identify the sex of the animals or combined data obtained from males and females. Cellular sex in the heart is relatively understudied and many studies fail to report the sex of the cells used for in vitro experiments. Moreover, in the small number of studies in which sex is reported, most of those studies use male cells. The observation that cells from males and females are inherently different is becoming increasingly clear - either due to acquired differences from hormones and other factors or due to intrinsic differences in genotype (XX or XY). Because of the likely contribution of cellular sex differences in cardiac health and disease, here, we explore differences in mammalian male and female cells in the heart, including the less-studied non-myocyte cell populations. We discuss how the heart's microenvironment impacts male and female cellular phenotypes and vice versa, including how secretory profiles are dependent on cellular sex, and how hormones contribute to sexually dimorphic phenotypes and cellular functions. Intracellular mechanisms that contribute to sex differences, including gene expression and epigenetic remodeling, are also described. Recent single-cell sequencing studies have revealed unexpected sex differences in the composition of cell types in the heart which we discuss. Finally, future recommendations for considering cellular sex differences in the design of bioengineered in vitro disease models of the heart are provided.
Collapse
Affiliation(s)
- Cierra J Walker
- Materials Science and Engineering Program, University of Colorado, Boulder, CO 80303, United States of America; Interdisciplinary Quantitative Biology, University of Colorado, Boulder, CO 80303, United States of America; BioFrontiers Institute, University of Colorado, Boulder, CO 80303, United States of America
| | - Megan E Schroeder
- Chemical and Biological Engineering Department, University of Colorado, Boulder, CO 80303, United States of America; BioFrontiers Institute, University of Colorado, Boulder, CO 80303, United States of America
| | - Brian A Aguado
- Chemical and Biological Engineering Department, University of Colorado, Boulder, CO 80303, United States of America; BioFrontiers Institute, University of Colorado, Boulder, CO 80303, United States of America
| | - Kristi S Anseth
- Chemical and Biological Engineering Department, University of Colorado, Boulder, CO 80303, United States of America; BioFrontiers Institute, University of Colorado, Boulder, CO 80303, United States of America
| | - Leslie A Leinwand
- BioFrontiers Institute, University of Colorado, Boulder, CO 80303, United States of America; Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, CO 80309, United States of America.
| |
Collapse
|
30
|
Barros MPD, Bachi ALL, Santos JDMBD, Lambertucci RH, Ishihara R, Polotow TG, Caldo-Silva A, Valente PA, Hogervorst E, Furtado GE. The poorly conducted orchestra of steroid hormones, oxidative stress and inflammation in frailty needs a maestro: Regular physical exercise. Exp Gerontol 2021; 155:111562. [PMID: 34560197 DOI: 10.1016/j.exger.2021.111562] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 09/14/2021] [Accepted: 09/17/2021] [Indexed: 12/25/2022]
Abstract
This review outlines the various factors associated with unhealthy aging which includes becoming frail and dependent. With many people not engaging in recommended exercise, facilitators and barriers to engage with exercise must be investigated to promote exercise uptake and adherence over the lifespan for different demographics, including the old, less affluent, women, and those with different cultural-ethnic backgrounds. Governmental and locally funded public health messages and environmental facilitation (gyms, parks etc.) can play an important role. Studies have shown that exercise can act as a conductor to balance oxidative stress, immune and endocrine functions together to promote healthy aging and reduce the risk for age-related morbidities, such as cardiovascular disease and atherosclerosis, and promote cognition and mood over the lifespan. Like a classic symphony orchestra, consisting of four groups of related musical instruments - the woodwinds, brass, percussion, and strings - the aging process should also perform in harmony, with compassion, avoiding the aggrandizement of any of its individual parts during the presentation. This review discusses the wide variety of molecular, cellular and endocrine mechanisms (focusing on the steroid balance) underlying this process and their interrelationships.
Collapse
Affiliation(s)
- Marcelo Paes de Barros
- Institute of Physical Activity Sciences and Sports (ICAFE), MSc/PhD Interdisciplinary Program in Health Sciences, Cruzeiro do Sul University, 01506-000 São Paulo, Brazil.
| | - André Luís Lacerda Bachi
- Department of Otorhinolaryngology, ENT Lab, Federal University of São Paulo (UNIFESP), São Paulo 04025-002, Brazil; Post-Graduation Program in Health Sciences, Santo Amaro University (UNISA), São Paulo 04829-300, Brazil
| | | | | | - Rafael Ishihara
- Department of Biosciences, Federal University of São Paulo (UNIFESP), Santos 11015-020, SP, Brazil
| | - Tatiana Geraldo Polotow
- Institute of Physical Activity Sciences and Sports (ICAFE), MSc/PhD Interdisciplinary Program in Health Sciences, Cruzeiro do Sul University, 01506-000 São Paulo, Brazil
| | - Adriana Caldo-Silva
- University of Coimbra, Research Unit for Sport and Physical Activity (CIDAF, UID/PTD/04213/2019) at Faculty of Sport Science and Physical Education, (FCDEF-UC), Portugal
| | - Pedro Afonso Valente
- University of Coimbra, Research Unit for Sport and Physical Activity (CIDAF, UID/PTD/04213/2019) at Faculty of Sport Science and Physical Education, (FCDEF-UC), Portugal
| | - Eef Hogervorst
- Applied Cognitive Research National Centre for Sports and Exercise Medicine, Loughborough University, Loughborough, UK
| | - Guilherme Eustáquio Furtado
- Health Sciences Research Unit: Nursing (UICISA: E), Nursing School of Coimbra (ESEnfC), Coimbra, Portugal; Institute Polytechnic of Maia, Porto, Portugal; University of Coimbra, Research Unit for Sport and Physical Activity (CIDAF, UID/PTD/04213/2019) at Faculty of Sport Science and Physical Education, (FCDEF-UC), Portugal.
| |
Collapse
|
31
|
Elzokm SS, Fouda MA, Abdel Moneim RA, El-Mas MM. Distinct effects of calcineurin dependent and independent immunosuppressants on endotoxaemia-induced nephrotoxicity in rats: Role of androgens. Clin Exp Pharmacol Physiol 2021; 48:1261-1270. [PMID: 34042216 DOI: 10.1111/1440-1681.13526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 05/05/2021] [Accepted: 05/24/2021] [Indexed: 11/26/2022]
Abstract
Evidence suggests that immunosuppressant therapies protect against harmful effects of endotoxaemia. In this study, we tested whether calcineurin-dependent (cyclosporine/tacrolimus) and -independent (sirolimus) immunosuppressants variably influence nephrotoxicity induced by endotoxaemia and whether this interaction is modulated by testosterone. We investigated the effects of immunosuppressants on renal histopathological, biochemical and inflammatory profiles in endotoxic male rats and the role of androgenic state in the interaction. Six-hour treatment of rats with lipopolysaccharide (LPS, 3 mg/kg) increased (i) serum urea/creatinine, (ii) width of proximal/distal tubules, (iii) tubular degeneration and vacuolation, (iv) Western protein expressions of renal toll-like receptor 4, monocyte chemoattractant protein-1, and NADPH oxidase-2, and (v) serum tumour necrosis factor-α and myeloperoxidase. These endotoxic manifestations were intensified and eliminated upon concurrent exposure to cyclosporine and sirolimus, respectively. The cyclosporine actions appear to be a class rather than a drug effect because similar exacerbation of LPS nephrotoxicity was observed in rats treated with tacrolimus, another calcineurin inhibitor (CNI). Moreover, the deteriorated renal outcomes in LPS/tacrolimus-treated rats were reduced after castration or androgen receptor blockade by flutamide. The data suggest opposite effects for calcineurin-dependent (exaggeration) and -independent immunosuppressants (amelioration) on renal defects of endotoxaemia and implicate androgenic pathways in the worsened endotoxic renal profile induced by CNIs.
Collapse
Affiliation(s)
- Shrouk S Elzokm
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Pharos University in Alexandria, Alexandria, Egypt
| | - Mohamed A Fouda
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Rahab A Abdel Moneim
- Department of Histology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Mahmoud M El-Mas
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
- Department of Pharmacology and Toxicology, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| |
Collapse
|
32
|
Nasser SA, Afify EA, Kobeissy F, Hamam B, Eid AH, El-Mas MM. Inflammatory Basis of Atherosclerosis: Modulation by Sex Hormones. Curr Pharm Des 2021; 27:2099-2111. [PMID: 33480335 DOI: 10.2174/1381612827666210122142811] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 12/17/2020] [Indexed: 11/22/2022]
Abstract
Atherosclerosis-related cardiovascular diseases (CVDs) are the leading cause of death globally. Several lines of evidence are supportive of the contributory role of vascular inflammation in atherosclerosis. Diverse immune cell types, including monocytes/macrophages, T-cells and neutrophils, as well as specialized proresolving lipid mediators, have been successfully characterized as key players in vascular inflammation. The increased prevalence of atherosclerotic CVD in men in comparison to age-matched premenopausal women and the abolition of sex differences in prevalence during menopause strongly suggest a pivotal role of sex hormones in the development of CVD. Indeed, many animal and human studies conclusively implicate sex hormones as a crucial component in driving the immune response. This is further corroborated by the effective identification of sex hormone receptors in vascular endothelial cells, vascular smooth muscle cells and immune cells. Collectively, these findings suggest a cellular communication between sex hormones and vascular or immune cells underlying the vascular inflammation in atherosclerosis. The aim of this review is to provide an overview of vascular inflammation as a causal cue underlying atherosclerotic CVDs within the context of the modulatory effects of sex hormones. Moreover, the cellular and molecular signaling pathways underlying the sex hormones- immune system interactions as potential culprits for vascular inflammation are highlighted with detailed and critical discussion. Finally, the review concludes by speculations on the potential sex-related efficacy of currently available immunotherapies in mitigating vascular inflammation. Conceivably, a deeper understanding of the immunoregulatory influence of sex hormones on vascular inflammation-mediated atherosclerosis permits sex-based management of atherosclerosis-related CVDs.
Collapse
Affiliation(s)
- Suzanne A Nasser
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Beirut Arab University, P.O. Box 11-5020, Beirut, Lebanon
| | - Elham A Afify
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, American University of Beirut, P.O. Box 11-0236, Beirut, Lebanon
| | - Bassam Hamam
- Department of Biological and Chemical Sciences, School of Arts and Sciences, Lebanese International University, P.O. Box 146404, Beirut, Lebanon
| | - Ali H Eid
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Mahmoud M El-Mas
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| |
Collapse
|
33
|
Lombardo G, Mondelli V, Dazzan P, Pariante CM. Sex hormones and immune system: A possible interplay in affective disorders? A systematic review. J Affect Disord 2021; 290:1-14. [PMID: 33989924 DOI: 10.1016/j.jad.2021.04.035] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 03/15/2021] [Accepted: 04/20/2021] [Indexed: 12/18/2022]
Abstract
BACKGROUND Sex hormones and the immune system may play a key role in sex differences in affective disorders. The understanding of their interplay may lead to the detection of new sex-specific tailored therapeutic approaches. The aim of this systematic review is to summarise the evidence supporting a possible association between sex hormones and inflammatory biomarkers in people with affective disorders. METHODS A systematic search of the literature published until January 2021 was conducted on PubMed database. The initial search identified a total of 1259 studies; 20 studies investigating inflammatory biomarkers and sex hormones in patients exhibiting depressive symptoms were included: 10 studies focused on patients with affective disorders, and 10 studies focused on women in menopause or in the post-partum period exhibiting depressive symptoms. RESULTS Testosterone and exogenous female sex hormones may play protective roles through their modulation of the immune system, respectively, in male patients with bipolar disorder and in peri-/post-menopausal women with depression. LIMITATIONS The main limitations are the paucity of studies investigating both sex hormones and immune biomarkers, the lack of statistical analyses exploring specifically the association between these two classes of biomarkers, and the great heterogeneity between the participants' samples in the studies. CONCLUSION This review highlights the need to investigate the interplay between sex hormones and immune system in affective disorders. The inconsistent or incomplete evidence may be improved by studies in patients with moderate-high inflammatory levels that specifically evaluate the relationship between sex hormones and the immune system.
Collapse
Affiliation(s)
- Giulia Lombardo
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, UK.
| | - Valeria Mondelli
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, UK; National Institute for Health Research Mental Health Biomedical Research Centre, South London and Maudsley NHS Foundation Trust and King's College London, London, UK
| | - Paola Dazzan
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, UK
| | - Carmine Maria Pariante
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, UK; National Institute for Health Research Mental Health Biomedical Research Centre, South London and Maudsley NHS Foundation Trust and King's College London, London, UK
| |
Collapse
|
34
|
Darzianiazizi M, Allison KE, Kulkarni RR, Sharif S, Karimi K, Bridle BW. Disruption of type I interferon signaling causes sexually dimorphic dysregulation of anti-viral cytokines. Cytokine X 2021; 3:100053. [PMID: 34189454 PMCID: PMC8215187 DOI: 10.1016/j.cytox.2021.100053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 04/09/2021] [Accepted: 06/01/2021] [Indexed: 12/01/2022] Open
Abstract
Type I interferons (IFNs) play a crucial role in the establishment of an antiviral state via signaling through their cognate type I IFN receptor (IFNAR). In this study, a replication-competent but highly attenuated strain of VSV (rVSVΔm51) carrying a deletion at position 51 of the matrix protein to remove suppression of anti-viral type I IFN responses was used to explore the effect of disrupted IFNAR signaling on inflammatory cytokine responses in mice. The kinetic responses of interleukin-6, tumor necrosis factor-α and interleukin-12 were evaluated in virus-infected male and female mice with or without concomitant antibody-mediated IFNAR-blockade. Unlike controls, both male and female IFNAR-blocked mice showed signs of sickness by 24-hours post-infection. Female IFNAR-blocked mice experienced greater morbidity as demonstrated by a significant decrease in body temperature. This was not the case for males. In addition, females with IFNAR-blockade mounted prolonged and exaggerated systemic inflammatory cytokine responses to rVSVΔm51. This was in stark contrast to controls with intact IFNAR signaling and males with IFNAR-blockade; they were able to down-regulate virus-induced inflammatory cytokine responses by 24-hours post-infection. Exaggerated cytokine responses in females with impaired IFNAR signaling was associated with more effective control of viremia than their male counterparts. However, the trade-off was greater immune-mediated morbidity. The results of this study demonstrated a role for IFNAR signaling in the down-regulation of antiviral cytokine responses, which was strongly influenced by sex. Our findings suggested that the potential to mount toxic cytokine responses to a virus with concomitant disruption of IFNAR signaling was heavily biased towards females.
Collapse
Affiliation(s)
- Maedeh Darzianiazizi
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, 50 Stone Rd. E., Guelph, Ontario N1G 2W1, Canada
| | - Katrina E Allison
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, 50 Stone Rd. E., Guelph, Ontario N1G 2W1, Canada
| | - Raveendra R Kulkarni
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27607, USA
| | - Shayan Sharif
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, 50 Stone Rd. E., Guelph, Ontario N1G 2W1, Canada
| | - Khalil Karimi
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, 50 Stone Rd. E., Guelph, Ontario N1G 2W1, Canada
| | - Byram W Bridle
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, 50 Stone Rd. E., Guelph, Ontario N1G 2W1, Canada
| |
Collapse
|
35
|
Reyes-García J, Montaño LM, Carbajal-García A, Wang YX. Sex Hormones and Lung Inflammation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1304:259-321. [PMID: 34019274 DOI: 10.1007/978-3-030-68748-9_15] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Inflammation is a characteristic marker in numerous lung disorders. Several immune cells, such as macrophages, dendritic cells, eosinophils, as well as T and B lymphocytes, synthetize and release cytokines involved in the inflammatory process. Gender differences in the incidence and severity of inflammatory lung ailments including asthma, chronic obstructive pulmonary disease (COPD), pulmonary fibrosis (PF), lung cancer (LC), and infectious related illnesses have been reported. Moreover, the effects of sex hormones on both androgens and estrogens, such as testosterone (TES) and 17β-estradiol (E2), driving characteristic inflammatory patterns in those lung inflammatory diseases have been investigated. In general, androgens seem to display anti-inflammatory actions, whereas estrogens produce pro-inflammatory effects. For instance, androgens regulate negatively inflammation in asthma by targeting type 2 innate lymphoid cells (ILC2s) and T-helper (Th)-2 cells to attenuate interleukin (IL)-17A-mediated responses and leukotriene (LT) biosynthesis pathway. Estrogens may promote neutrophilic inflammation in subjects with asthma and COPD. Moreover, the activation of estrogen receptors might induce tumorigenesis. In this chapter, we summarize the most recent advances in the functional roles and associated signaling pathways of inflammatory cellular responses in asthma, COPD, PF, LC, and newly occurring COVID-19 disease. We also meticulously deliberate the influence of sex steroids on the development and progress of these common and severe lung diseases.
Collapse
Affiliation(s)
- Jorge Reyes-García
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico.,Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Luis M Montaño
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico
| | - Abril Carbajal-García
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico
| | - Yong-Xiao Wang
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA.
| |
Collapse
|
36
|
Subramanian A, Anand A, Adderley NJ, Okoth K, Toulis KA, Gokhale K, Sainsbury C, O'Reilly MW, Arlt W, Nirantharakumar K. Increased COVID-19 infections in women with polycystic ovary syndrome: a population-based study. Eur J Endocrinol 2021; 184:637-645. [PMID: 33635829 PMCID: PMC8052516 DOI: 10.1530/eje-20-1163] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 02/25/2021] [Indexed: 01/03/2023]
Abstract
OBJECTIVE Several recent observational studies have linked metabolic comorbidities to an increased risk from COVID-19. Here we investigated whether women with PCOS are at an increased risk of COVID-19 infection. DESIGN Population-based closed cohort study between 31 January 2020 and 22 July 2020 in the setting of a UK primary care database (The Health Improvement Network, THIN). METHODS The main outcome was the incidence of COVID-19 coded as suspected or confirmed by the primary care provider. We used Cox proportional hazards regression model with stepwise inclusion of explanatory variables (age, BMI, impaired glucose regulation, androgen excess, anovulation, vitamin D deficiency, hypertension, and cardiovascular disease) to provide unadjusted and adjusted hazard risks (HR) of COVID-19 infection among women with PCOS compared to women without PCOS. RESULTS We identified 21 292 women with a coded diagnosis of PCO/PCOS and randomly selected 78 310 aged and general practice matched control women. The crude COVID-19 incidence was 18.1 and 11.9 per 1000 person-years among women with and without PCOS, respectively. Age-adjusted Cox regression analysis suggested a 51% higher risk of COVID-19 among women with PCOS compared to women without PCOS (HR: 1.51 (95% CI: 1.27-1.80), P < 0.001). After adjusting for age and BMI, HR reduced to 1.36 (1.14-1.63)], P = 0.001. In the fully adjusted model, women with PCOS had a 28% increased risk of COVID-19 (aHR: 1.28 (1.05-1.56), P = 0.015). CONCLUSION Women with PCOS are at an increased risk of COVID-19 infection and should be specifically encouraged to adhere to infection control measures during the COVID-19 pandemic. SIGNIFICANCE STATEMENT Women with polycystic ovary syndrome (PCOS) have an increased risk of cardio-metabolic disease, which have been identified as a risk factor for COVID-19. To investigate whether the increased metabolic risk in PCOS translates into an increased risk of COVID-19 infection, we carried out a population-based closed cohort study in the UK during its first wave of the SARS-CoV-2 pandemic (January to July 2020), including 21 292 women with PCOS and 78 310 controls matched for sex, age and general practice location. Results revealed a 52% increased risk of COVID-19 infection in women with PCOS, which remained increased at 28% above controls after adjustment for age, BMI, impaired glucose regulation and other explanatory variables.
Collapse
Affiliation(s)
| | - Astha Anand
- Institute of Applied Health Research, University of Birmingham, Birmingham, UK
| | - Nicola J Adderley
- Institute of Applied Health Research, University of Birmingham, Birmingham, UK
| | - Kelvin Okoth
- Institute of Applied Health Research, University of Birmingham, Birmingham, UK
| | | | - Krishna Gokhale
- Institute of Applied Health Research, University of Birmingham, Birmingham, UK
| | | | - Michael W O'Reilly
- Royal College of Surgeons in Ireland (RCSI), University of Medicine and Health Sciences, Dublin, Republic of Ireland
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Wiebke Arlt
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
- National Institute for Health Research (NIHR), Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Birmingham, UK
- Correspondence should be addressed to W Arlt;
| | - Krishnarajah Nirantharakumar
- Institute of Applied Health Research, University of Birmingham, Birmingham, UK
- Midlands Health Data Research UK, Birmingham, UK
- Correspondence should be addressed to K Nirantharakumar;
| |
Collapse
|
37
|
Association between metabolic and hormonal profile, proinflammatory cytokines in saliva and gingival health in adolescent females with polycystic ovary syndrome. BMC Oral Health 2021; 21:193. [PMID: 33849511 PMCID: PMC8045362 DOI: 10.1186/s12903-021-01553-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 04/07/2021] [Indexed: 02/07/2023] Open
Abstract
Background Research studies indicate that polycystic ovary syndrome (PCOS) may increase susceptibility to periodontal disease. The mechanisms that link both conditions are not entirely understood. Thus, the study aimed to investigate the impact of hormonal and metabolic disturbances on the gingival health and salivary levels of tumor necrosis factor (TNF-α), interleukin 1β (IL1-β), and interleukin 6 (IL-6) in adolescent girls with PCOS. Methods Thirty-one patients with PCOS and twenty-eight healthy age-mates (as the control group) were enrolled in the study. Individuals with PCOS underwent blood tests for the determination of hormonal and metabolic parameters. Saliva samples were collected to measure salivary testosterone and proinflammatory cytokines in both studied groups. Calibrated dentist assessed oral hygiene and gingival health of all subjects. Results Salivary testosterone was significantly higher in the study group (p = 0.0007). The groups did not differ significantly concerning periodontal parameters. Patients with PCOS revealed higher levels of salivary cytokines (p < 0.0001). Gingival index (GI) and the percentage of sites bleeding upon probing (BOP%) were positively correlated with the plaque index (PI) in both groups (rs ≥ 0.60, p < 0.001), and negatively correlated with salivary testosterone level in the PCOS group (rs = − 0.44, p = 0.0138 and rs = − 0.37, p = 0.0424, respectively). BOP% was also positively correlated with body mass index (BMI) in the control group (rs = 0.40, p = 0.0368) and index of insulin resistance (HOMA-IR) in the study group (rs = 0.48, p = 0.0068). Salivary testosterone was positively correlated with TNF-α in the control group (rs = 0.41, p = 0.0321), while in the study group, total testosterone (TT) was positively correlated with IL-6 (rs = 0.37, p = 0.0400) and free androgen index (FAI) with TNF-α (rs = 0.36, p = 0.0491). Conclusions Gingival health of the examined population was associated primarily with oral hygiene and, to a lesser extent, with the hormonal and metabolic profile. Despite similar periodontal parameters in the both studied groups, patients with PCOS revealed significantly higher levels of proinflammatory cytokines in saliva, which might be the manifestation of the systemic low-grade inflammation associated with PCOS.
Collapse
|
38
|
Ambhore NS, Kalidhindi RSR, Sathish V. Sex-Steroid Signaling in Lung Diseases and Inflammation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1303:243-273. [PMID: 33788197 DOI: 10.1007/978-3-030-63046-1_14] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Sex/gender difference exists in the physiology of multiple organs. Recent epidemiological reports suggest the influence of sex-steroids in modulating a wide variety of disease conditions. Sex-based discrepancies have been reported in pulmonary physiology and various chronic inflammatory responses associated with lung diseases like asthma, chronic obstructive pulmonary disease (COPD), pulmonary fibrosis, and rare lung diseases. Notably, emerging clinical evidence suggests that several respiratory diseases affect women to a greater degree, with increased severity and prevalence than men. Although sex-specific differences in various lung diseases are evident, such differences are inherent to sex-steroids, which are major biological variables in men and women who play a central role to control these differences. The focus of this chapter is to comprehend the sex-steroid biology in inflammatory lung diseases and to understand the mechanistic role of sex-steroids signaling in regulating these diseases. Exploring the roles of sex-steroid signaling in the regulation of lung diseases and inflammation is crucial for the development of novel and effective therapy. Overall, we will illustrate the importance of differential sex-steroid signaling in lung diseases and their possible clinical implications for the development of complementary and alternative medicine to treat lung diseases.
Collapse
Affiliation(s)
- Nilesh Sudhakar Ambhore
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, ND, USA
| | | | - Venkatachalem Sathish
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, ND, USA.
| |
Collapse
|
39
|
Kalidhindi RSR, Ambhore NS, Balraj P, Schmidt T, Khan MN, Sathish V. Androgen receptor activation alleviates airway hyperresponsiveness, inflammation, and remodeling in a murine model of asthma. Am J Physiol Lung Cell Mol Physiol 2021; 320:L803-L818. [PMID: 33719566 DOI: 10.1152/ajplung.00441.2020] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Epidemiological studies demonstrate an apparent sex-based difference in the prevalence of asthma, with a higher risk in boys than girls, which is reversed postpuberty, where women become more prone to asthma than men, suggesting a plausible beneficial role for male hormones, especially androgens as a regulator of pathophysiology in asthmatic lungs. Using a murine model of asthma developed with mixed allergen (MA) challenge, we report a significant change in airway hyperresponsiveness (AHR), as demonstrated by increased thickness of epithelial and airway smooth muscle layers and collagen deposition, as well as Th2/Th17-biased inflammation in the airways of non-gonadectomized (non-GDX) and gonadectomized (GDX) male mice. Here, compared with non-GDX mice, MA-induced AHR and inflammatory changes were more prominent in GDX mice. Activation of androgen receptor (AR) using 5α-dihydrotestosterone (5α-DHT, AR agonist) resulted in decreased Th2/Th17 inflammation and remodeling-associated changes, resulting in improved lung function compared with MA alone challenged mice, especially in GDX mice. These changes were not observed with Flutamide (Flut, AR antagonist). Overall, we show that AR exerts a significant and beneficial role in asthma by regulating AHR and inflammation.
Collapse
Affiliation(s)
- Rama Satyanarayana Raju Kalidhindi
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, North Dakota
| | - Nilesh Sudhakar Ambhore
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, North Dakota
| | - Premanand Balraj
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, North Dakota
| | - Taylor Schmidt
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota
| | - M Nadeem Khan
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota
| | - Venkatachalem Sathish
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, North Dakota
| |
Collapse
|
40
|
Schiffer L, Bossey A, Kempegowda P, Taylor AE, Akerman I, Scheel-Toellner D, Storbeck KH, Arlt W. Peripheral blood mononuclear cells preferentially activate 11-oxygenated androgens. Eur J Endocrinol 2021; 184:353-363. [PMID: 33444228 PMCID: PMC7923147 DOI: 10.1530/eje-20-1077] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 01/12/2021] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Androgens are important modulators of immune cell function. The local generation of active androgens from circulating precursors is an important mediator of androgen action in peripheral target cells or tissues. We aimed to characterize the activation of classic and 11-oxygenated androgens in human peripheral blood mononuclear cells (PBMCs). METHODS PBMCs were isolated from healthy male donors and incubated ex vivo with precursors and active androgens of the classic and 11-oxygenated androgen pathways. Steroids were quantified by liquid chromatography-tandem mass spectrometry. The expression of genes encoding steroid-metabolizing enzymes was assessed by quantitative PCR. RESULTS PBMCs generated eight-fold higher amounts of the active 11-oxygenated androgen 11-ketotestosterone than the classic androgen testosterone from their respective precursors. We identified the enzyme AKR1C3 as the major reductive 17β-hydroxysteroid dehydrogenase in PBMCs responsible for both conversions and found that within the PBMC compartment natural killer cells are the major site of AKRC13 expression and activity. Steroid 5α-reductase type 1 catalyzed the 5α-reduction of classic but not 11-oxygenated androgens in PBMCs. Lag time prior to the separation of cellular components from whole blood increased serum 11-ketotestosterone concentrations in a time-dependent fashion, with significant increases detected from two hours after blood collection. CONCLUSIONS 11-Oxygenated androgens are the preferred substrates for androgen activation by AKR1C3 in PBMCs, primarily conveyed by natural killer cell AKR1C3 activity, yielding 11-ketotestosterone the major active androgen in PBMCs. Androgen metabolism by PBMCs can affect the results of serum 11-ketotestosterone measurements, if samples are not separated in a timely fashion. SIGNIFICANCE STATEMENT We show that human peripheral blood mononuclear cells (PBMCs) preferentially activate 11-ketotestosterone rather than testosterone when incubated with precursors of both the classic and the adrenal-derived 11-oxygenated androgen biosynthesis pathways. We demonstrate that this activity is catalyzed by the enzyme AKR1C3, which we found to primarily reside in natural killer cells, major contributors to the anti-viral immune defense. This potentially links intracrine 11-oxygenated androgen generation to the previously observed decreased NK cell cytotoxicity and increased infection risk in primary adrenal insufficiency. In addition, we show that PBMCs continue to generate 11-ketotestosterone if the cellular component of whole blood samples is not removed in a timely fashion, which could affect measurements of this active androgen in routine clinical biochemistry.
Collapse
Affiliation(s)
- Lina Schiffer
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Alicia Bossey
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Punith Kempegowda
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Angela E Taylor
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Ildem Akerman
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | | | - Karl-Heinz Storbeck
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
- Department of Biochemistry, Stellenbosch University, Stellenbosch, South Africa
| | - Wiebke Arlt
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
- National Institute for Health Research (NIHR), Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Birmingham, UK
- Correspondence should be addressed to W Arlt;
| |
Collapse
|
41
|
Najjar RS, Turner CG, Wong BJ, Feresin RG. Berry-Derived Polyphenols in Cardiovascular Pathologies: Mechanisms of Disease and the Role of Diet and Sex. Nutrients 2021; 13:nu13020387. [PMID: 33513742 PMCID: PMC7911141 DOI: 10.3390/nu13020387] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/21/2021] [Accepted: 01/22/2021] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular disease (CVD) prevalence, pathogenesis, and manifestation is differentially influenced by biological sex. Berry polyphenols target several signaling pathways pertinent to CVD development, including inflammation, oxidative stress, and cardiac and vascular remodeling, and there are innate differences in these pathways that also vary by sex. There is limited research systematically investigating sex differences in berry polyphenol effects on these pathways, but there are fundamental findings at this time that suggest a sex-specific effect. This review will detail mechanisms within these pathological pathways, how they differ by sex, and how they may be individually targeted by berry polyphenols in a sex-specific manner. Because of the substantial polyphenolic profile of berries, berry consumption represents a promising interventional tool in the treatment and prevention of CVD in both sexes, but the mechanisms in which they function within each sex may vary.
Collapse
Affiliation(s)
- Rami S. Najjar
- Department of Nutrition, Georgia State University, Atlanta, GA 30302, USA;
| | - Casey G. Turner
- Department of Kinesiology and Health, Georgia State University, Atlanta, GA 30302, USA; (C.G.T.); (B.J.W.)
| | - Brett J. Wong
- Department of Kinesiology and Health, Georgia State University, Atlanta, GA 30302, USA; (C.G.T.); (B.J.W.)
| | - Rafaela G. Feresin
- Department of Nutrition, Georgia State University, Atlanta, GA 30302, USA;
- Correspondence:
| |
Collapse
|
42
|
Abancens M, Bustos V, Harvey H, McBryan J, Harvey BJ. Sexual Dimorphism in Colon Cancer. Front Oncol 2020; 10:607909. [PMID: 33363037 PMCID: PMC7759153 DOI: 10.3389/fonc.2020.607909] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 11/09/2020] [Indexed: 12/12/2022] Open
Abstract
A higher incidence of colorectal cancer (CRC) is found in males compared to females. Young women (18-44 years) with CRC have a better survival outcome compared to men of the same age or compared to older women (over 50 years), indicating a global incidence of sexual dimorphism in CRC rates and survival. This suggests a protective role for the sex steroid hormone estrogen in CRC development. Key proliferative pathways in CRC tumorigenesis exhibit sexual dimorphism, which confer better survival in females through estrogen regulated genes and cell signaling. Estrogen regulates the activity of a class of Kv channels (KCNQ1:KCNE3), which control fundamental ion transport functions of the colon and epithelial mesenchymal transition through bi-directional interactions with the Wnt/β-catenin signalling pathway. Estrogen also modulates CRC proliferative responses in hypoxia via the novel membrane estrogen receptor GPER and HIF1A and VEGF signaling. Here we critically review recent clinical and molecular insights into sexual dimorphism of CRC biology modulated by the tumor microenvironment, estrogen, Wnt/β-catenin signalling, ion channels, and X-linked genes.
Collapse
Affiliation(s)
- Maria Abancens
- Department of Molecular Medicine, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin, Ireland
- Department of Surgery, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin, Ireland
| | - Viviana Bustos
- Departamento de Acuicultura y Recursos Agroalimentarios, Programa Fitogen, Universidad de Los Lagos, Osorno, Chile
| | - Harry Harvey
- Department of Medical Oncology, Cork University Hospital, Cork, Ireland
| | - Jean McBryan
- Department of Molecular Medicine, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin, Ireland
- Department of Surgery, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin, Ireland
| | - Brian J. Harvey
- Department of Molecular Medicine, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin, Ireland
- Centro de Estudios Cientificos CECs, Valdivia, Chile
| |
Collapse
|
43
|
Lim HJ. Autophagy in the uterine vessel microenvironment: Balancing vasoactive factors. Clin Exp Reprod Med 2020; 47:263-268. [PMID: 33227184 PMCID: PMC7711101 DOI: 10.5653/cerm.2020.04126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 10/14/2020] [Indexed: 11/23/2022] Open
Abstract
Autophagy, which has the literal meaning of self-eating, is a cellular catabolic process executed by arrays of conserved proteins in eukaryotes. Autophagy is dynamically ongoing at a basal level, presumably in all cells, and often carries out distinct functions depending on the cell type. Therefore, although a set of common genes and proteins is involved in this process, the outcome of autophagic activation or deficit requires scrutiny regarding how it affects cells in a specific pathophysiological context. The uterus is a complex organ that carries out multiple tasks under the influence of cyclic changes of ovarian steroid hormones. Several major populations of cells are present in the uterus, and the interactions among them drive complex physiological tasks. Mouse models with autophagic deficits in the uterus are very limited, but provide an initial glimpse at how autophagy plays a distinct role in different uterine tissues. Herein, we review recent research findings on the role of autophagy in the uterine mesenchyme in mouse models.
Collapse
|
44
|
Misiakiewicz-Has K, Zawiślak A, Pilutin A, Kolasa-Wołosiuk A, Szumilas P, Duchnik E, Wiszniewska B. Morphological and Functional Changes in Skin of Adult Male Rats Chronically Treated with Letrozole, a Nonsteroidal Inhibitor of Cytochrome P450 Aromatase. Acta Histochem Cytochem 2020; 53:99-111. [PMID: 33177782 PMCID: PMC7642481 DOI: 10.1267/ahc.20009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 07/08/2020] [Indexed: 11/22/2022] Open
Abstract
Skin is a target for hormones and a site of hormone production. Aromatase inhibitors such as letrozole reduce circulating estrogen. The aim of the study was to investigate the morphology of the dermis and immunoexpression of androgen receptor (AR), estrogen receptor α and β (ERα, ERβ), luteinizing hormone receptor (LHR), follicle-stimulating hormone receptor (FSHR), and cytochrome P450 aromatase (P450arom) in male rats with a deficit of estradiol. Experiments were performed on skin of 12 male rats. Rats in the experimental group received per os letrozole for 6 months. For morphological analysis, van Gieson, Sirius Red and orcein staining of sections was performed. In immunohistochemistry, reactions with specific antibodies (anti-P450arom, LHR, FSHR, ERα, ERβ) were used. In morphometric analysis, sections were stained with hematoxylin and eosin. Differences between groups were assessed by Mann-Whitney U-test. There were no differences in the diameter of collagen fibers. The dermis of letrozole-treated animals showed areas without collagen fibers, and expression of P450arom, ERα and ERβ was diminished in the skin of these animals. This study indicates that estrogens exert an effect via ERs that has a role in maintaining proper skin morphology in males, together with androgen. This is also the first documented expression of FSHR in the skin of male rats.
Collapse
Affiliation(s)
| | - Alicja Zawiślak
- Department of Interdisciplinary Dentistry, Pomeranian Medical University in Szczecin
| | - Anna Pilutin
- Department of Histology and Embryology, Pomeranian Medical University in Szczecin
| | | | - Paweł Szumilas
- Department of Social Medicine and Public Health, Pomeranian Medical University in Szczecin
| | - Ewa Duchnik
- Department of Dermatology and Venereology, Pomeranian Medical University in Szczecin
| | - Barbara Wiszniewska
- Department of Histology and Embryology, Pomeranian Medical University in Szczecin
| |
Collapse
|
45
|
Park JM, Lee YJ. Serum oestradiol levels are inversely associated with C-reactive protein levels in premenopausal women, but not postmenopausal women. J Int Med Res 2020; 48:300060520961228. [PMID: 33044103 PMCID: PMC7556179 DOI: 10.1177/0300060520961228] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE Epidemiological studies on the association of serum oestradiol levels and inflammatory markers have reported inconsistent and conflicting results. Therefore, we investigated the association between serum oestradiol and high-sensitivity C-reactive protein (CRP) levels in women on the basis of their menopausal status. METHODS This cross-sectional study examined the association between serum oestradiol and CRP levels on the basis of menopausal status in 151 premenopausal women aged 42.7 ± 6.7 years and 394 postmenopausal women aged 58.1 ± 6.7 years who participated in a health examination program. Multiple linear regression analysis was conducted using CRP levels as the dependent variable. RESULTS Multiple linear regression analysis showed that serum oestradiol levels were inversely associated with CRP levels in premenopausal women (β coefficient = -0.298) after adjusting for age, body mass index, smoking, mean arterial pressure, and levels of fasting plasma glucose, triglycerides, high-density lipoprotein cholesterol, aspartate aminotransferase, and alanine aminotransferase. However, this association was not found in postmenopausal women after adjusting for the same confounding factors. CONCLUSIONS Serum oestradiol levels are inversely associated with CRP levels in premenopausal women, but not in postmenopausal women. Lower oestrogenic activity may at least partly contribute to the pathogenesis of chronic inflammation, particularly in premenopausal women.
Collapse
Affiliation(s)
- Jae-Min Park
- Department of Family Medicine, Yonsei University College of Medicine, Gangnam Severance Hospital, Seoul, Korea.,Department of Medicine, Graduate School of Medicine, Yonsei University, Seoul, Korea
| | - Yong-Jae Lee
- Department of Family Medicine, Yonsei University College of Medicine, Gangnam Severance Hospital, Seoul, Korea
| |
Collapse
|
46
|
Androgen receptor signaling in the lungs mitigates inflammation and improves the outcome of influenza in mice. PLoS Pathog 2020; 16:e1008506. [PMID: 32645119 PMCID: PMC7373319 DOI: 10.1371/journal.ppat.1008506] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 07/21/2020] [Accepted: 03/27/2020] [Indexed: 01/06/2023] Open
Abstract
Circulating androgens can modulate immune cell activity, but the impact of androgens on viral pathogenesis remains unclear. Previous data demonstrate that testosterone reduces the severity of influenza A virus (IAV) infection in male mice by mitigating pulmonary inflammation rather than by affecting viral replication. To examine the immune responses mediated by testosterone to mitigate IAV-induced inflammation, adult male mice remained gonadally intact or were gonadectomized and treated with either placebo or androgen-filled (i.e., testosterone or dihydrotestosterone) capsules prior to sublethal IAV infection. Like intact males, treatment of gonadectomized males with androgens improved the outcome of IAV infection, which was not mediated by changes in the control of virus replication or pulmonary cytokine activity. Instead, androgens accelerated pulmonary leukocyte contraction to limit inflammation. To identify which immune cells were contracting in response to androgens, the composition of pulmonary cellular infiltrates was analyzed and revealed that androgens specifically accelerated the contraction of total pulmonary inflammatory monocytes during peak disease, as well as CD8+ T cells, IAV-specific CD8+ T numbers, cytokine production and degranulation by IAV-specific CD8+ T cells, and the influx of eosinophils into the lungs following clearance of IAV. Neither depletion of eosinophils nor adoptive transfer of CD8+ T cells could reverse the ability of testosterone to protect males against IAV suggesting these were secondary immunologic effects. The effects of testosterone on the contraction of immune cell numbers and activity were blocked by co-administration of the androgen receptor antagonist flutamide and mimicked by treatment with dihydrotestosterone, which was also able to reduce the severity of IAV in female mice. These data suggest that androgen receptor signaling creates a local pulmonary environment that promotes downregulation of detrimental inflammatory immune responses to protect against prolonged influenza disease.
Collapse
|
47
|
Chronic maternal interleukin-17 and autism-related cortical gene expression, neurobiology, and behavior. Neuropsychopharmacology 2020; 45:1008-1017. [PMID: 32074626 PMCID: PMC7162858 DOI: 10.1038/s41386-020-0640-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 02/06/2020] [Accepted: 02/11/2020] [Indexed: 12/18/2022]
Abstract
Chronic inflammation during pregnancy (e.g., preeclampsia, diabetes) is linked to increased risk for offspring neurodevelopmental disorders such as autism spectrum disorder (ASD). However, mediators of such exposures that could be targeted with maternal intervention are unclear, as few chronic gestational inflammation models have been tested. One potential mediator is interleukin-17 (IL-17), a pro-inflammatory cytokine implicated in neurodevelopmental disorders and gestational disease. To test chronic maternal IL-17 impacts on offspring, C57BL/6J dams were administered IL-17A continuously throughout pregnancy. Offspring were assessed for body weight; cortical volume, gene expression, and cellular composition; and adult behavior. IL-17A-condition offspring exhibited decreased somatic and cortical size at embryonic day 18 (E18) and as adults. mRNA sequencing of E18 cortex revealed 320 differentially expressed genes in males, but none in females. These were significantly enriched for ASD (Simons Foundation Autism Research Initiative), synaptic, and cell cycle genes. By adulthood, neocortical glial cell density and gene expression were decreased, while GABAergic synaptic gene expression was increased in males. Furthermore, IL-17A-condition male but not female offspring exhibited reduced anxiety-like behavior. Social approach deficits in males were negatively correlated with neocortical GABAergic synaptic gene expression. Chronic gestational IL-17A was sufficient to cause ASD-like phenotypes early and persistently in male offspring. This echoes the male bias, altered cortical development, and behavioral findings in ASD, suggesting that chronic maternal IL-17 contributes to offspring ASD pathogenesis. Furthermore, the trajectory from embryonically dysregulated synaptic and cell cycle genes to disrupted adult glia, inhibitory synapses, and behavior suggests a mechanism for chronic maternal IL-17 effects on offspring.
Collapse
|
48
|
Sex differences in leukocyte profile in ST-elevation myocardial infarction patients. Sci Rep 2020; 10:6851. [PMID: 32321989 PMCID: PMC7176674 DOI: 10.1038/s41598-020-63185-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 03/23/2020] [Indexed: 12/18/2022] Open
Abstract
Background: Whether sex differences exist in the inflammatory response after ST-elevation myocardial infarction (STEMI) remains to be elucidated. We studied leukocyte profiles and their prognostic value in men and women presenting with STEMI. Methods: From a total of 552 consecutive STEMI patients, blood samples were collected at hospital admission. Linear regression was used to assess the relationship between leukocyte profiles and enzymatic infarct size. Cox regression was used to assess the association between leukocyte profiles and one-year mortality. Results: Women presented with higher lymphocyte counts (2.3·109 cells/L (IQR 1.6–3.1) vs. 1.8·109 cells/L (IQR 1.4–2.5), p = 3.00 ∙ 10−4) and percentages (21.1% (IQR 14.4–28.1) vs. 17.1% (IQR 12.3–24.3), p = 0.004). Lymphocyte to monocyte ratio (LMR) was also higher in women (3.25 (IQR 2.56–4.5) vs. 2.68 (IQR 2.08–3.59), p = 7.28 ∙ 10−7). Higher LMR was associated with lower peak CK-MB (β = −0.27 (95% CI: −0.50, −0.03), p = 0.026), lower peak troponin T (β = −0.45 (95% CI: −0.77, −0.13), p = 0.006) and lower one-year mortality risk (HR 0.35 (95% CI: 0.13, 0.96), p = 0.042). Conclusion: At admission for STEMI, women present with higher lymphocyte count and LMR. Higher LMR is associated with smaller infarct size and decreased one-year mortality risk and could be used as a biomarker to predict outcome.
Collapse
|
49
|
Tuku B, Stanelle-Bertram S, Sellau J, Beck S, Bai T, Kouassi NM, Preuß A, Hoenow S, Renné T, Lotter H, Gabriel G. Testosterone Protects Against Severe Influenza by Reducing the Pro-Inflammatory Cytokine Response in the Murine Lung. Front Immunol 2020; 11:697. [PMID: 32431696 PMCID: PMC7216738 DOI: 10.3389/fimmu.2020.00697] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 03/27/2020] [Indexed: 11/21/2022] Open
Abstract
Influenza A virus pathogenesis may differ between men and women. The 2009 H1N1 influenza pandemic resulted in more documented hospitalizations in women compared to men. In this study, we analyzed the impact of male sex hormones on pandemic 2009 H1N1 influenza A virus disease outcome. In a murine infection model, we could mimic the clinical findings with female mice undergoing severe and even fatal 2009 H1N1 influenza compared to male mice. Treatment of female mice with testosterone could rescue the majority of mice from lethal influenza. Improved disease outcome in testosterone treated female mice upon 2009 H1N1 influenza A virus infection did not affect virus titers in the lung compared to carrier-treated females. However, reduction in IL-1β cytokine expression levels strongly correlated with reduced lung damage and improved influenza disease outcome in female mice upon testosterone treatment. In contrast, influenza disease outcome was not affected between castrated male mice and non-castrated controls. Here, influenza infection resulted in reduction of testosterone expression in male mice. These findings show that testosterone has protective functions on the influenza infection course. However, 2009 H1N1 influenza viruses seem to have evolved yet unknown mechanisms to reduce testosterone expression in males. These data will support future antiviral strategies to treat influenza taking sex-dependent immunopathologies into consideration.
Collapse
Affiliation(s)
- Berfin Tuku
- Department Viral Zoonoses - One Health, Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Stephanie Stanelle-Bertram
- Department Viral Zoonoses - One Health, Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Julie Sellau
- Research Group Molecular Infection Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Sebastian Beck
- Department Viral Zoonoses - One Health, Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Tian Bai
- Department Viral Zoonoses - One Health, Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Nancy Mounogou Kouassi
- Department Viral Zoonoses - One Health, Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Annette Preuß
- Department Viral Zoonoses - One Health, Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Stefan Hoenow
- Research Group Molecular Infection Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Thomas Renné
- Institute for Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hanna Lotter
- Research Group Molecular Infection Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Gülsah Gabriel
- Department Viral Zoonoses - One Health, Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany.,Institute of Virology, University of Veterinary Medicine Hannover, Hanover, Germany
| |
Collapse
|
50
|
Kim S, Song S, Kim YS, Yang SY, Lee JE. The association between predicted inflammatory status and colorectal adenoma. Sci Rep 2020; 10:2433. [PMID: 32051482 PMCID: PMC7016133 DOI: 10.1038/s41598-020-59271-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 01/27/2020] [Indexed: 02/08/2023] Open
Abstract
We developed a diet and lifestyle score based on high sensitivity C-reactive protein (hsCRP), and investigated its association with odds of adenoma. We performed stepwise linear regression to develop the predicted hsCRP score among 23,330 participants in the Health Examinee Study and examined its association with colorectal adenoma among 1,711 participants in a cross-sectional study of colorectal adenoma. We estimated odds ratios (ORs) and 95% confidence intervals (CIs) of colorectal adenoma using logistic regression models. Variances in hsCRP explained by body mass index were 61.1% in men and 64.5% in women in the prediction model. The increasing predicted hsCRP score was positively associated with colorectal adenoma (ORquartile 4 VS quartile 1 1.71, 95% CI: 1.12–2.62; Ptrend = 0.011 in men; ORquartile 4 VS quartile 1 2.86, 95% CI: 1.26–6.49; Ptrend = 0.019 in women). In subgroups, the associations differed by age and menopausal status among women, with stronger associations among women aged less than 50 years (OR≥median VS <median 3.74, 95% CI: 1.77–7.90, p for interaction 0.014) or premenopausal women (OR≥median vs <median 4.21, 95% CI: 2.12–8.36, p for interaction <0.001). The associations were more pronounced in the advanced or distal colon/rectum in men and in the advanced or proximal colon in women. The associations were attenuated after further adjustment for body mass index. In conclusion, we found that the predicted hsCRP score was positively associated with colorectal adenoma, suggesting that diet and lifestyle lowering inflammation may be a strategy to prevent colorectal neoplasia.
Collapse
Affiliation(s)
- Sejin Kim
- Department of Food and Nutrition, College of Human Ecology, Seoul National University, Seoul, Republic of Korea
| | - Sihan Song
- Department of Food and Nutrition, College of Human Ecology, Seoul National University, Seoul, Republic of Korea
| | - Young Sun Kim
- Department of Internal Medicine, Healthcare System Gangnam Center, Seoul National University Hospital, Seoul, Republic of Korea
| | - Sun Young Yang
- Department of Internal Medicine, Healthcare System Gangnam Center, Seoul National University Hospital, Seoul, Republic of Korea.
| | - Jung Eun Lee
- Department of Food and Nutrition, College of Human Ecology, Seoul National University, Seoul, Republic of Korea. .,Research Institute of Human Ecology, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea.
| |
Collapse
|