1
|
Frank J, Tehrani L, Gamer J, Van Booven DJ, Ballarin S, Rossman R, Edelstein A, Uppalati S, Reuthebuck A, Collado F, Klimas NG, Nathanson L. Gulf War Illness Induced Sex-Specific Transcriptional Differences Under Stressful Conditions. Int J Mol Sci 2025; 26:3610. [PMID: 40332133 PMCID: PMC12026906 DOI: 10.3390/ijms26083610] [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: 02/15/2025] [Revised: 04/01/2025] [Accepted: 04/03/2025] [Indexed: 05/08/2025] Open
Abstract
Gulf War Illness (GWI) is a multi-symptom disorder affecting 1990-1991 Persian Gulf War veterans and is characterized by post-exertional malaise, neurological symptoms, immune deregulation, and exhaustion. Causation is not understood, and effective diagnostics and therapies have not yet been developed. In this work, we analyzed stress-related, sex-specific transcriptomic shifts in GWI subjects and healthy controls through RNA sequencing of peripheral blood mononuclear cells (PBMCs). Blood samples at baseline (T0), at maximal exertion (T1), and four hours post-exertion (T2) were analyzed. In female subjects with GWI, pathways associated with pro-inflammatory processes were found to be deregulated, and in male GWI subjects, pathways related to IL-12 signaling and lymphocytic activation were deregulated at T1 compared to T0. During recovery from stress, pathways corresponding to immune responses and microglial cell activation were altered in female GWI subjects, and apoptotic signaling changed in males with GWI. Documented sex-specific immune deregulation leads to finding better biomarkers. Targeting sex-specific transcriptomic markers of the disease could lead to new therapies for GWI.
Collapse
Affiliation(s)
- Joshua Frank
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (J.F.); (F.C.); (N.G.K.)
| | - Lily Tehrani
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (L.T.); (J.G.); (S.B.); (R.R.); (A.E.)
| | - Jackson Gamer
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (L.T.); (J.G.); (S.B.); (R.R.); (A.E.)
| | - Derek J. Van Booven
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL 33146, USA;
| | - Sarah Ballarin
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (L.T.); (J.G.); (S.B.); (R.R.); (A.E.)
| | - Raquel Rossman
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (L.T.); (J.G.); (S.B.); (R.R.); (A.E.)
| | - Abraham Edelstein
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (L.T.); (J.G.); (S.B.); (R.R.); (A.E.)
| | - Sadhika Uppalati
- Halmos College of Arts and Sciences, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (S.U.); (A.R.)
| | - Ana Reuthebuck
- Halmos College of Arts and Sciences, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (S.U.); (A.R.)
| | - Fanny Collado
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (J.F.); (F.C.); (N.G.K.)
| | - Nancy G. Klimas
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (J.F.); (F.C.); (N.G.K.)
- Department of Veterans Affairs, Miami VA Healthcare System, Geriatric Research Education and Clinical Center (GRECC), Miami, FL 33125, USA
| | - Lubov Nathanson
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (J.F.); (F.C.); (N.G.K.)
| |
Collapse
|
2
|
Yamamoto C, Kobashi Y, Kawamura T, Nishikawa Y, Saito H, Oguro F, Zhao T, Takita M, Sawano T, Ozaki A, Abe T, Ito N, Kaneko Y, Nakayama A, Wakui M, Kodama T, Tsubokura M. Group of longitudinal adverse event patterns after the fourth dose of COVID-19 vaccination with a latent class analysis. Front Public Health 2024; 12:1406315. [PMID: 39139673 PMCID: PMC11320210 DOI: 10.3389/fpubh.2024.1406315] [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: 04/08/2024] [Accepted: 07/16/2024] [Indexed: 08/15/2024] Open
Abstract
Introduction Vaccination has been implemented as a useful measure to combat the COVID-19 pandemic. However, there is a tendency for individuals to avoid vaccination due to the possibility of adverse events, making it important to investigate the relationship between COVID-19 vaccines and their adverse events. This study explored longitudinal adverse event patterns and factors that influence adverse events following the second to fourth doses of the COVID-19 vaccine through a latent class analysis. Methods Participants were recruited from the Fukushima Prefecture and included individuals who had completed four doses of the COVID-19 mRNA vaccine. This study utilized data from questionnaire surveys and blood collection conducted between September 2021 and November 2022. In the questionnaire, factors such as sex, age, medical history, medication, type of vaccine administered, and adverse events following vaccination were recorded. Additionally, in the blood data, serological tests [IgG(S)] and cellular immune responses (T-spot) were measured. Descriptive statistics, latent class analysis, multivariable logistic regression, and multiple regression analyses were performed to identify the longitudinal adverse event patterns and influencing factors. By analyzing adverse events over time, we identified two distinct groups: those less prone to experiencing adverse events (Group 1) and those more susceptible (Group 2) to latent class analysis. Results A total of 1,175 participants were included after excluding those without any adverse events. The median age of the participants in Group 1 was 70 years, and in Group 2 it was 51 years. The proportion of female participants was 298 in Group 1 and 353 in Group 2. Patients in Group 2 were significantly younger (p < 0.001) and more likely to be female (p < 0.001) than those in Group 1. Furthermore, the median IgG(S) value after the fourth vaccination was 3,233 AU/mL in Group 1 and 4,059.39 AU/mL in Group 2. The median T-spot value was 15.4 in Group 1 and 28.5 in Group 2. Group 2 showed significantly higher IgG(S) and T-spot values after the fourth vaccination (p < 0.001). Discussion Our findings suggest that factors other than age, particularly sex and a history of allergies, significantly influence the likelihood of experiencing adverse events. Groups categorized by latent class analysis for longitudinal adverse events are expected to be valuable for optimizing vaccination strategies and formulating public health measures.
Collapse
Affiliation(s)
- Chika Yamamoto
- Department of Radiation Health Management, Fukushima Medical University School of Medicine, Fukushima City, Fukushima, Japan
| | - Yurie Kobashi
- Department of Radiation Health Management, Fukushima Medical University School of Medicine, Fukushima City, Fukushima, Japan
- Department of General Internal Medicine, Hirata Central Hospital, Hirata, Fukushima, Japan
| | - Takeshi Kawamura
- Proteomics Laboratory, Isotope Science Center, The University of Tokyo, Tokyo, Japan
- Laboratory for Systems Biology and Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Yoshitaka Nishikawa
- Department of General Internal Medicine, Hirata Central Hospital, Hirata, Fukushima, Japan
| | - Hiroaki Saito
- Department of Radiation Health Management, Fukushima Medical University School of Medicine, Fukushima City, Fukushima, Japan
- Department of Internal Medicine, Soma Central Hospital, Soma, Fukushima, Japan
| | - Fumiya Oguro
- Department of General Internal Medicine, Hirata Central Hospital, Hirata, Fukushima, Japan
| | - Tianchen Zhao
- Department of Radiation Health Management, Fukushima Medical University School of Medicine, Fukushima City, Fukushima, Japan
| | - Morihito Takita
- Department of Radiation Health Management, Fukushima Medical University School of Medicine, Fukushima City, Fukushima, Japan
| | - Toyoaki Sawano
- Department of Surgery, Jyoban Hospital, Iwaki, Fukushima, Japan
| | - Akihiko Ozaki
- Department of Breast and Thyroid Surgery, Jyoban Hospital, Iwaki, Fukushima, Japan
| | - Toshiki Abe
- Department of Radiation Health Management, Fukushima Medical University School of Medicine, Fukushima City, Fukushima, Japan
| | - Naomi Ito
- Department of Radiation Health Management, Fukushima Medical University School of Medicine, Fukushima City, Fukushima, Japan
| | - Yudai Kaneko
- Laboratory for Systems Biology and Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
- Medical & Biological Laboratories Co., Ltd, Minato-ku, Tokyo, Japan
| | - Aya Nakayama
- Proteomics Laboratory, Isotope Science Center, The University of Tokyo, Tokyo, Japan
| | - Masatoshi Wakui
- Department of Laboratory Medicine, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Tatsuhiko Kodama
- Laboratory for Systems Biology and Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Masaharu Tsubokura
- Department of Radiation Health Management, Fukushima Medical University School of Medicine, Fukushima City, Fukushima, Japan
- Department of General Internal Medicine, Hirata Central Hospital, Hirata, Fukushima, Japan
- Minamisoma Municipal General Hospital, Minamisoma, Fukushima, Japan
| |
Collapse
|
3
|
Feng Q, Shigesi N, Guan J, Rahmioglu N, Bafadhel M, Paddon K, Hubbard C, Zondervan K, Becker C, Hellner K. Elevated basophil count is associated with increased risk of endometriosis. REPRODUCTION AND FERTILITY 2024; 5:RAF-23-0090. [PMID: 39012084 PMCID: PMC11378143 DOI: 10.1530/raf-23-0090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 07/15/2024] [Indexed: 07/17/2024] Open
Abstract
Immunological dysregulation plays a fundamental role in the inflammatory aspects of endometriosis. Circulating blood leukocytes, one of the most abundant immune cell populations in the human body, have been shown diagnostic significance in some diseases. Nevertheless, the association between peripheral blood leukocyte counts and endometriosis remains unexplored to date. We analysed two targeted study cohorts: a tertiary centre cohort (Endometriosis at Oxford University [ENDOX] study, 325 cases/177 controls) and a large-scale population study (UK Biobank [UKBB], 1537 cases/6331 controls). In both datasets, peripheral venous blood sample results were retrieved and counts of leukocyte subpopulations, including neutrophils, lymphocytes, monocytes, eosinophils and basophils analysed. Logistic regression models were used to investigate the association of leukocyte subtype alterations with endometriosis status, adjusting for confounding factors. We demonstrate that higher blood basophil level is associated with increased odds of endometriosis. This association was first discovered in the ENDOX cohort (basophils >0.04 x10^9/L: OR 1.65 [95%CI:1.06-2.57], P trend = 0.025) and replicated in the UKBB dataset (basophils >0.04 x10^9/L: OR 1.26 [95%CI:1.09-1.45], P trend = 0.001). Notably, women with basophil counts in the upper tercile had significantly increased odds of having stage III/IV endometriosis (ENDOX study: OR = 2.30, 95% CI [1.25 to 4.22], P trend = 0.007; UKBB study (OR = 1.40, 95% CI [1.07 to 1.85], P trend = 0.015). None of the other leukocyte subtypes showed an association. Our findings suggest an association between inflammatory responses and the pathogenesis of endometriosis; future studies are warranted to investigate whether the association is causal.
Collapse
Affiliation(s)
- Qian Feng
- Q Feng, Monash University, Clayton, Australia
| | - Nina Shigesi
- N Shigesi, Oxford, United Kingdom of Great Britain and Northern Ireland
| | | | - Nilufer Rahmioglu
- N Rahmioglu, University of Oxford, Oxford, United Kingdom of Great Britain and Northern Ireland
| | - Mona Bafadhel
- M Bafadhel, King's College London, London, United Kingdom of Great Britain and Northern Ireland
| | - Kevin Paddon
- K Paddon, Oxford, United Kingdom of Great Britain and Northern Ireland
| | - Carol Hubbard
- C Hubbard, Oxford, United Kingdom of Great Britain and Northern Ireland
| | - Krina Zondervan
- K Zondervan, Women's Health, University of Oxford, Oxford, United Kingdom of Great Britain and Northern Ireland
| | - Christian Becker
- C Becker, Oxford, United Kingdom of Great Britain and Northern Ireland
| | - Karin Hellner
- K Hellner, University of Oxford, Oxford, OX1 2JD, United Kingdom of Great Britain and Northern Ireland
| |
Collapse
|
4
|
Zhao J, Wang Q, Tan AF, Loh CJL, Toh HC. Sex differences in cancer and immunotherapy outcomes: the role of androgen receptor. Front Immunol 2024; 15:1416941. [PMID: 38863718 PMCID: PMC11165033 DOI: 10.3389/fimmu.2024.1416941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 05/16/2024] [Indexed: 06/13/2024] Open
Abstract
Across the wide range of clinical conditions, there exists a sex imbalance where biological females are more prone to autoimmune diseases and males to some cancers. These discrepancies are the combinatory consequence of lifestyle and environmental factors such as smoking, alcohol consumption, obesity, and oncogenic viruses, as well as other intrinsic biological traits including sex chromosomes and sex hormones. While the emergence of immuno-oncology (I/O) has revolutionised cancer care, the efficacy across multiple cancers may be limited because of a complex, dynamic interplay between the tumour and its microenvironment (TME). Indeed, sex and gender can also influence the varying effectiveness of I/O. Androgen receptor (AR) plays an important role in tumorigenesis and in shaping the TME. Here, we lay out the epidemiological context of sex disparity in cancer and then review the current literature on how AR signalling contributes to such observation via altered tumour development and immunology. We offer insights into AR-mediated immunosuppressive mechanisms, with the hope of translating preclinical and clinical evidence in gender oncology into improved outcomes in personalised, I/O-based cancer care.
Collapse
Affiliation(s)
- Junzhe Zhao
- Duke-NUS Medical School, Singapore, Singapore
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Qian Wang
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
- Department of Medical Oncology Cancer Hospital of China Medical University/Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, China
| | | | - Celestine Jia Ling Loh
- Duke-NUS Medical School, Singapore, Singapore
- Sengkang General Hospital, Singapore, Singapore
| | - Han Chong Toh
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| |
Collapse
|
5
|
Anger JT, Case LK, Baranowski AP, Berger A, Craft RM, Damitz LA, Gabriel R, Harrison T, Kaptein K, Lee S, Murphy AZ, Said E, Smith SA, Thomas DA, Valdés Hernández MDC, Trasvina V, Wesselmann U, Yaksh TL. Pain mechanisms in the transgender individual: a review. FRONTIERS IN PAIN RESEARCH 2024; 5:1241015. [PMID: 38601924 PMCID: PMC11004280 DOI: 10.3389/fpain.2024.1241015] [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: 06/15/2023] [Accepted: 01/25/2024] [Indexed: 04/12/2024] Open
Abstract
Specific Aim Provide an overview of the literature addressing major areas pertinent to pain in transgender persons and to identify areas of primary relevance for future research. Methods A team of scholars that have previously published on different areas of related research met periodically though zoom conferencing between April 2021 and February 2023 to discuss relevant literature with the goal of providing an overview on the incidence, phenotype, and mechanisms of pain in transgender patients. Review sections were written after gathering information from systematic literature searches of published or publicly available electronic literature to be compiled for publication as part of a topical series on gender and pain in the Frontiers in Pain Research. Results While transgender individuals represent a significant and increasingly visible component of the population, many researchers and clinicians are not well informed about the diversity in gender identity, physiology, hormonal status, and gender-affirming medical procedures utilized by transgender and other gender diverse patients. Transgender and cisgender people present with many of the same medical concerns, but research and treatment of these medical needs must reflect an appreciation of how differences in sex, gender, gender-affirming medical procedures, and minoritized status impact pain. Conclusions While significant advances have occurred in our appreciation of pain, the review indicates the need to support more targeted research on treatment and prevention of pain in transgender individuals. This is particularly relevant both for gender-affirming medical interventions and related medical care. Of particular importance is the need for large long-term follow-up studies to ascertain best practices for such procedures. A multi-disciplinary approach with personalized interventions is of particular importance to move forward.
Collapse
Affiliation(s)
- Jennifer T. Anger
- Department of Urology, University of California San Diego, San Diego, CA, United States
| | - Laura K. Case
- Department of Anesthesiology, University of California San Diego, San Diego, CA, United States
| | - Andrew P. Baranowski
- Pelvic Pain Medicine and Neuromodulation, University College Hospital Foundation Trust, University College London, London, United Kingdom
| | - Ardin Berger
- Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA, United States
| | - Rebecca M. Craft
- Department of Psychology, Washington State University, Pullman, WA, United States
| | - Lyn Ann Damitz
- Division of Plastic and Reconstructive Surgery, University of North Carolina, Chapel Hill, NC, United States
| | - Rodney Gabriel
- Division of Regional Anesthesia, University of California San Diego, San Diego, CA, United States
| | - Tracy Harrison
- Department of OB/GYN & Reproductive Sciences, University of California San Diego, San Diego, CA, United States
| | - Kirsten Kaptein
- Division of Plastic Surgery, University of California San Diego, San Diego, CA, United States
| | - Sanghee Lee
- Department of Urology, University of California San Diego, San Diego, CA, United States
| | - Anne Z. Murphy
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
| | - Engy Said
- Division of Regional Anesthesia, University of California San Diego, San Diego, CA, United States
| | - Stacey Abigail Smith
- Division of Infection Disease, The Hope Clinic of Emory University, Atlanta, GA, United States
| | - David A. Thomas
- Office of Research on Women's Health, National Institutes of Health, Bethesda, MD, United States
| | - Maria del C. Valdés Hernández
- Department of Neuroimaging Sciences, Center for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Victor Trasvina
- Department of Urology, University of California San Diego, San Diego, CA, United States
| | - Ursula Wesselmann
- Departments of Anesthesiology and Perioperative Medicine/Division of Pain Medicine, Neurology and Psychology, and Consortium for Neuroengineering and Brain-Computer Interfaces, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Tony L. Yaksh
- Department of Anesthesiology, University of California San Diego, San Diego, CA, United States
| |
Collapse
|
6
|
Alvergne A, Boniface E, Darney B, Shea A, Weber K, Ventola C, Vitzthum VJ, Edelman A. Associations Among Menstrual Cycle Length, Coronavirus Disease 2019 (COVID-19), and Vaccination. Obstet Gynecol 2024; 143:83-91. [PMID: 37562052 PMCID: PMC10715707 DOI: 10.1097/aog.0000000000005343] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 06/08/2023] [Accepted: 06/15/2023] [Indexed: 08/12/2023]
Abstract
OBJECTIVE To assess whether coronavirus disease 2019 (COVID-19) is associated with menstrual cycle length changes and, if so, how that compares with those undergoing vaccination or no event (control). METHODS We conducted a retrospective cohort analysis in which we analyzed prospectively tracked cycle-length data from users of a period tracker application who also responded to a survey regarding COVID-19 symptoms and vaccination. We restricted our sample to users aged 16-45 years, with normal cycle lengths (24-38 days) and regular tracking behavior during the five cycles around COVID-19 symptoms or vaccination or a similar time period for those experiencing no event (control group). We calculated the within-user change in cycle length (days) from the three consecutive cycles preevent average (either vaccination, disease, or neither; cycles 1-3) to the event (cycle 4) and postevent (cycle 5) cycles. We used mixed-effects models to estimate the age- and country-adjusted difference in change in cycle length across the groups. RESULTS We included 6,514 users from 110 countries representing 32,570 cycles (COVID-19 symptoms: 1,450; COVID-19 vaccination: 4,643; control: 421). The COVID-19 cohort experienced a 1.45-day adjusted increase in cycle length during cycle 4 (COVID-19) compared with their three preevent cycles (95% CI 0.86-2.04). The vaccinated group experienced a 1.14-day adjusted increase in cycle length during cycle 4 (COVID-19 vaccine) compared with their preevent average (95% CI 0.60-1.69). The control group (neither vaccine nor disease) experienced a 0.68-day decrease (95% CI -1.18 to -0.19) in a similar time period. Post hoc tests showed no significant differences in the magnitude of changes between the COVID-19 and vaccination cohorts. In both cohorts, cycle length changes disappeared in the postevent cycle. CONCLUSION Experiencing COVID-19 is associated with a small change in cycle length similar to COVID-19 vaccination. These changes resolve quickly within the next cycle.
Collapse
Affiliation(s)
- Alexandra Alvergne
- Institute for Evolutionary Sciences, Montpellier University, Montpellier, France; the School of Anthropology and Museum Ethnography, University of Oxford, Oxford, United Kingdom; the Department of Obstetrics and Gynecology, Oregon Health & Science University, and the OHSU-PSU School of Public Health, Portland, Oregon; the National Institute of Public Health (INSP), Center for Population Health (CISP), Cuernavaca, Morelos, Mexico; and Clue by BioWink GmbH, Berlin, Germany
| | | | | | | | | | | | | | | |
Collapse
|
7
|
Maddalon A, Cari L, Iulini M, Alhosseini MN, Galbiati V, Marinovich M, Nocentini G, Corsini E. Impact of endocrine disruptors on peripheral blood mononuclear cells in vitro: role of gender. Arch Toxicol 2023; 97:3129-3150. [PMID: 37676302 PMCID: PMC10567873 DOI: 10.1007/s00204-023-03592-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 08/24/2023] [Indexed: 09/08/2023]
Abstract
Humans can be exposed to endocrine disruptors (EDs) in numerous ways. EDs can interfere with endogenous hormones at different levels, resulting in numerous adverse human health outcomes, including immunotoxicity. In this regard, this study aimed to investigate in vitro the possible effects of EDs on immune cells and possible gender differences. Peripheral blood mononuclear cells from healthy humans, both males and females, were exposed to 6 different EDs, namely atrazine (herbicide), cypermethrin (insecticide), diethyl phthalate (plasticizer), 17α-ethynylestradiol (contraceptive drug), perfluorooctanesulfonic acid (persistent organic pollutant), and vinclozolin (fungicide). We evaluated the effect of EDs on RACK1 (receptor for activated C kinase 1) expression, considering it as a bridge between the endocrine and the immune system, and putatively used as screening tool of immunotoxic effects of EDs. The exposure to EDs resulted at different extent in alteration in RACK1 expression, pro-inflammatory activity, natural killer lytic ability, and lymphocyte differentiation, with sex-related differences. In particular, diethyl phthalate and perfluorooctanesulfonic acid resulted the most active EDs tested, with gender differences in terms of effects and magnitude. The results from our study evidenced the ability of EDs to directly affect immune cells.
Collapse
Affiliation(s)
- Ambra Maddalon
- Laboratory of Toxicology, Department of Pharmacological and Biomolecular Sciences, Rodolfo Paoletti', Università Degli Studi Di Milano, Via Balzaretti 9, 20133, Milan, Italy
| | - Luigi Cari
- Department of Medicine and Surgery, Section of Pharmacology, Università Degli Studi Di Perugia, Building D, Severi Square 1, 06129, Perugia, Italy
| | - Martina Iulini
- Laboratory of Toxicology, Department of Pharmacological and Biomolecular Sciences, Rodolfo Paoletti', Università Degli Studi Di Milano, Via Balzaretti 9, 20133, Milan, Italy
| | - Mahdieh Naghavi Alhosseini
- Department of Medicine and Surgery, Section of Pharmacology, Università Degli Studi Di Perugia, Building D, Severi Square 1, 06129, Perugia, Italy
| | - Valentina Galbiati
- Laboratory of Toxicology, Department of Pharmacological and Biomolecular Sciences, Rodolfo Paoletti', Università Degli Studi Di Milano, Via Balzaretti 9, 20133, Milan, Italy
| | - Marina Marinovich
- Laboratory of Toxicology, Department of Pharmacological and Biomolecular Sciences, Rodolfo Paoletti', Università Degli Studi Di Milano, Via Balzaretti 9, 20133, Milan, Italy
| | - Giuseppe Nocentini
- Department of Medicine and Surgery, Section of Pharmacology, Università Degli Studi Di Perugia, Building D, Severi Square 1, 06129, Perugia, Italy.
| | - Emanuela Corsini
- Laboratory of Toxicology, Department of Pharmacological and Biomolecular Sciences, Rodolfo Paoletti', Università Degli Studi Di Milano, Via Balzaretti 9, 20133, Milan, Italy
| |
Collapse
|
8
|
Cignarella A, Vegeto E, Bolego C, Trabace L, Conti L, Ortona E. Sex-oriented perspectives in immunopharmacology. Pharmacol Res 2023; 197:106956. [PMID: 37820857 DOI: 10.1016/j.phrs.2023.106956] [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: 08/07/2023] [Revised: 09/27/2023] [Accepted: 10/08/2023] [Indexed: 10/13/2023]
Abstract
Several immunopharmacological agents are effective in the treatment of cancer and immune-mediated conditions, with a favorable impact on life expectancy and clinical outcomes for a large number of patients. Nevertheless, response variation and undesirable effects of these drugs represent major issues, and overall efficacy remains unpredictable. Males and females show a distinct difference in immune system responses, with females generally mounting stronger responses to a variety of stimuli. Therefore, exploring sex differences in the efficacy and safety of immunopharmacological agents would strengthen the practice of precision medicine. As a pharmacological target highlight, programmed cell death 1 ligand 1 (PD-L1) is the first functionally characterized ligand of the coinhibitory programmed death receptor 1 (PD-1). The PD-L1/PD-1 crosstalk plays an important role in the immune response and is relevant in cancer, infectious and autoimmune disease. Sex differences in the response to immune checkpoint inhibitors are well documented, with male patients responding better than female patients. Similarly, higher efficacy of and adherence to tumor necrosis factor inhibitors in chronic inflammatory conditions including rheumatoid arthritis and Crohn's disease have been reported in male patients. The pharmacological basis of sex-specific responses to immune system modulating drugs is actively investigated in other settings such as stroke and type 1 diabetes. Advances in therapeutics targeting the endothelium could soon be wielded against autoimmunity and metabolic disorders. Based on the established sexual dimorphism in immune-related pathophysiology and disease presentation, sex-specific immunopharmacological protocols should be integrated into clinical guidelines.
Collapse
Affiliation(s)
| | - Elisabetta Vegeto
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - Chiara Bolego
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Luigia Trabace
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Lucia Conti
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Elena Ortona
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
9
|
Kawji Y, Almoaswes H, Bise C, Kawji L, Murphy A, Reed TD, Klapper RJ, Ahmadzadeh S, Shekoohi S, Cornett EM, Kaye AD. Electronic Health Record Recording of Patient Pain: Challenges and Discrepancies. Curr Pain Headache Rep 2023; 27:737-745. [PMID: 37740879 DOI: 10.1007/s11916-023-01170-x] [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] [Accepted: 09/10/2023] [Indexed: 09/25/2023]
Abstract
PURPOSE OF REVIEW In the present review, various categories of pain, clinician-observed pain scales, and patient-reported pain scales are evaluated to better understand factors that impact patient pain perceptions. Additionally, the expansion of areas that require further research to determine the optimal way to evaluate pain scale data for treatment and management are discussed. RECENT FINDINGS Electronic health record (EHR) data provides a starting point for evaluating whether patient predictors influence postoperative pain. There are several ways to assess pain and choosing the most effective form of pain treatment. Identifying individuals at high risk for severe postoperative pain enables more effective pain treatment. However, there are discrepancies in patient pain reporting dependent on instruments used to measure pain and their storage in the EHR. Additionally, whether administered by a physician or another healthcare practitioner, differences in patient pain perception occur. While each scale has distinct advantages and limitations, pain scale data is a valuable therapeutic tool for assisting clinicians in providing patients with optimal pain control. Accurate assessment of patient pain perceptions by data extraction from electronic health records provides a potential for pain alleviation improvement. Predicting high-risk postoperative pain syndromes is a difficult clinical challenge. Numerous studies have been conducted on factors that impact pain prediction. Postoperative pain is significantly predicted by the kind of operation, the existence of prior discomfort, patient anxiety, and age.
Collapse
Affiliation(s)
- Yasmeen Kawji
- School of Medicine, Louisiana State University Health Sciences Center New Orleans, 433 Bolivar Street, New Orleans, LA, 70112, USA
| | - Hanna Almoaswes
- School of Medicine, Louisiana State University Health Sciences Center New Orleans, 433 Bolivar Street, New Orleans, LA, 70112, USA
| | - Claire Bise
- School of Medicine, Louisiana State University Health Sciences Center New Orleans, 433 Bolivar Street, New Orleans, LA, 70112, USA
| | - Lena Kawji
- Department of Internal Medicine, School of Medicine, Louisiana State University Health Sciences Center at Shreveport, 1501 Kings Highway, Shreveport, LA, 71103, USA
| | - Adrienne Murphy
- School of Medicine, Louisiana State University Health Sciences Center New Orleans, 433 Bolivar Street, New Orleans, LA, 70112, USA
| | - Tanner D Reed
- School of Medicine, Louisiana State University Health Sciences Center New Orleans, 433 Bolivar Street, New Orleans, LA, 70112, USA
| | - Rachel J Klapper
- Department of Radiology, Louisiana State University Health Sciences Center at Shreveport, 1501 Kings Highway, Shreveport, LA, 71103, USA
| | - Shahab Ahmadzadeh
- Department of Anesthesiology, Louisiana State University Health Sciences Center at Shreveport, 1501 Kings Highway, Shreveport, LA, 71103, USA
| | - Sahar Shekoohi
- Department of Anesthesiology, Louisiana State University Health Sciences Center at Shreveport, 1501 Kings Highway, Shreveport, LA, 71103, USA.
| | - Elyse M Cornett
- Department of Anesthesiology, Louisiana State University Health Sciences Center at Shreveport, 1501 Kings Highway, Shreveport, LA, 71103, USA
| | - Alan D Kaye
- Department of Anesthesiology, Louisiana State University Health Sciences Center at Shreveport, 1501 Kings Highway, Shreveport, LA, 71103, USA
- Departments of Anesthesiology and Pharmacology, Toxicology, and Neurosciences, Louisiana State University Health Sciences Center at Shreveport, 1501 Kings Highway, Shreveport, LA, 71103, USA
| |
Collapse
|
10
|
Wang J, Yin T, Liu S. Dysregulation of immune response in PCOS organ system. Front Immunol 2023; 14:1169232. [PMID: 37215125 PMCID: PMC10196194 DOI: 10.3389/fimmu.2023.1169232] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 04/24/2023] [Indexed: 05/24/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is the most common reproductive endocrine disorder affecting women, which can lead to infertility. Infertility, obesity, hirsutism, acne, and irregular menstruation are just a few of the issues that PCOS can be linked to. PCOS has a complicated pathophysiology and a range of clinical symptoms. Chronic low-grade inflammation is one of the features of PCOS. The inflammatory environment involves immune and metabolic disturbances. Numerous organ systems across the body, in addition to the female reproductive system, have been affected by the pathogenic role of immunological dysregulation in PCOS in recent years. Insulin resistance and hyperandrogenism are associated with immune cell dysfunction and cytokine imbalance. More importantly, obesity is also involved in immune dysfunction in PCOS, leading to an inflammatory environment in women with PCOS. Hormone, obesity, and metabolic interactions contribute to the pathogenesis of PCOS. Hormone imbalance may also contribute to the development of autoimmune diseases. The aim of this review is to summarize the pathophysiological role of immune dysregulation in various organ systems of PCOS patients and provide new ideas for systemic treatment of PCOS in the future.
Collapse
Affiliation(s)
- Jingxuan Wang
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Tailang Yin
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Su Liu
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Shenzhen Zhongshan Urology Hospital, Shenzhen, China
| |
Collapse
|
11
|
Avin KG, Dominguez JM, Chen NX, Hato T, Myslinski JJ, Gao H, Liu Y, McKinley TO, Brown KM, Moe SM, Natoli RM. Single-cell RNAseq provides insight into altered immune cell populations in human fracture nonunions. J Orthop Res 2023; 41:1060-1069. [PMID: 36200412 PMCID: PMC10335365 DOI: 10.1002/jor.25452] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 09/16/2022] [Accepted: 09/22/2022] [Indexed: 02/04/2023]
Abstract
Nonunion describes bone fractures that fail to heal, resulting in the fracture callus failing to fully ossify or, in atrophic cases, not forming altogether. Fracture healing is regulated, in part, by the balance of proinflammatory and anti-inflammatory processes occurring within the bone marrow and surface cell populations. We sought to further understand the role of osteoimmunology (i.e., study of the close relationship between the immune system and bone) by examining immune cell gene expression via single-cell RNA sequencing of intramedullary canal tissue obtained from human patients with femoral nonunions. Intramedullary canal tissue samples obtained by reaming were collected at the time of surgical repair for femur fracture nonunion (n = 5) or from native bone controls when harvesting autologous bone graft (n = 4). Cells within the samples were isolated and analyzed using the Chromium Single-Cell System (10x Genomics Inc.) and Illumina sequencers. Twenty-three distinct cell clusters were identified, with higher cell proportions in the nonunion samples for monocytes and CD14 + dendritic cells (DCs), and lower proportions of T cells, myelocytes, and promyelocytes in nonunion samples. Gene expression differences were identified in each of the cell clusters from cell types associated with osteoimmunology, including CD14 + DC, monocytes, T cells, promyelocytes, and myelocytes. These results provide human-derived gene profiles that can further our understanding of pathways that may be a cause or a consequence of nonunion, providing the clinical rationale to focus on specific components of osteoimmunology. Clinical significance: The novel single-cell approach may lead to clinically relevant diagnostic biomarkers during earlier stages of nonunion development and/or investigation into therapeutic options.
Collapse
Affiliation(s)
- Keith G. Avin
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University, Indianapolis, Indiana, USA
- Division of Nephrology, School of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - James M. Dominguez
- Division of Nephrology, School of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Neal X. Chen
- Division of Nephrology, School of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Takashi Hato
- Division of Nephrology, School of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Jered J. Myslinski
- Division of Nephrology, School of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Hongyu Gao
- Department of Medical and Molecular Genetics, School of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Yunlong Liu
- Department of Medical and Molecular Genetics, School of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Todd O. McKinley
- Department of Orthopaedic Surgery, School of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Krista M. Brown
- Department of Orthopaedic Surgery, School of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Sharon M. Moe
- Division of Nephrology, School of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Roman M. Natoli
- Department of Orthopaedic Surgery, School of Medicine, Indiana University, Indianapolis, Indiana, USA
| |
Collapse
|
12
|
Abstract
Sex and gender play a pivotal role in health and disease. Differences can be identified in symptoms, biomarkers, lifetime experiences of diseases, incidence, prevalence, therapeutic options, health-related behavior, and resiliency. However, awareness of sex and gender differences in medicine is still limited. Systematic implementation of sex and gender-sensitive research is not yet the norm, resulting in gaps in evidence especially in the diagnosis and treatment of diseases in women. For decades research has predominantly included male persons and animals, leading to a lack of information about symptoms in female individuals or the classification of their symptoms as "atypical". Currently, the inclusion of female participants in clinical marketing access trials is mandatory. However, this does not automatically translate into sex-disaggregated analyses potentially limiting the discovery of sex-specific targeted therapeutic schemes. Consistent consideration of sex and gender in planning, conducting, analyzing, and dissemination of pharmacological research projects is an important prerequisite for closing the gender data gap. Targeted implementation strategies might help to include sex and gender aspects in different parts of the health system and thereby support the improvement of health care for all patients. Health economic aspects could be a further drive for the implementation of sex- and gender-sensitive medicine.The current chapter focuses on the role of sex and gender in biomedical research and, consequently, their potential role in pharmacology. We will explore the commonly used terminology in the field, the historical development of sex and gender-sensitive medicine (SGSM), the relevance of sex and gender to research and clinical practice and conclude with an outlook on future developments in the field.
Collapse
Affiliation(s)
- Eva Becher
- Sex- and Gender-Sensitive Medicine Unit, Medical Faculty OWL, Bielefeld University, Bielefeld, Germany
| | - Sabine Oertelt-Prigione
- Sex- and Gender-Sensitive Medicine Unit, Medical Faculty OWL, Bielefeld University, Bielefeld, Germany.
- Gender Unit, Departement of Primary and Community Care, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
13
|
Park MY, Park SY, Hartog A, van Hoffen E, Kardinaal A, Kim J, Choi HJ, Kwon O, Kim JY. Study protocol for cholera vaccination as a model to measure the inflammatory response in the gut: A case of modulation with a Lactobacillus plantarum K8 lysate. PLoS One 2023; 18:e0281817. [PMID: 36809275 PMCID: PMC9942990 DOI: 10.1371/journal.pone.0281817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 01/20/2023] [Indexed: 02/23/2023] Open
Abstract
It is crucial for human health that the immune system of the gastrointestinal tract works effectively. Dietary modulation is one of the factors that regulate the immune response in the gut. This study aims to develop a safe human challenge model to study gastrointestinal inflammation and immune function. This study focuses on evaluating gut stimulation induced by the oral cholera vaccine in healthy people. In addition, this paper describes the study design for assessing the efficacy and safety of a probiotic lysate, identifying whether functional ingredients in food can modulate inflammatory response induced by oral cholera vaccine. Forty-six males aged 20 to 50 with healthy bowel habits will be randomly allocated to the placebo or intervention group. Participants will consume 1 capsule of probiotic lysate or placebo twice daily for 6 weeks, take oral cholera vaccines on visit 2 (day 15) and visit 5 (day 29). The level of fecal calprotectin, a marker of gut inflammation, will be the primary outcome. The changes of cholera toxin-specific antibody levels and local/systemic inflammatory responses will be evaluated in blood. The purpose of this study is to evaluate gut stimulation of the oral cholera vaccine and investigate the effect of a probiotic lysate on improving the mild inflammatory response induced by the vaccine or supporting the immune response in healthy subjects. Trial registration: * This trial is registered in the International Clinical Trials Registry Platform of WHO (ICTRP, registration number: KCT0002589).
Collapse
Affiliation(s)
- Min Young Park
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul, Republic of Korea
| | - Soo-yeon Park
- Department of Food Science and Technology, Seoul National University of Science and Technology, Seoul, Republic of Korea
| | - Anita Hartog
- Department of Health, NIZO, Ede, The Netherlands
| | | | | | - Joohee Kim
- BiofoodCRO Co., Ltd., Seoul, Republic of Korea
| | - Hee Jung Choi
- Division of Infectious Diseases, Office of Infection Control, Ewha Woman’s University Medical Center, Seoul, Republic of Korea
| | - Oran Kwon
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul, Republic of Korea
- System Health & Engineering Major in Graduate School, Ewha Womans University, Seoul, Republic of Korea
- * E-mail: (OK); (JYK)
| | - Ji Yeon Kim
- Department of Food Science and Technology, Seoul National University of Science and Technology, Seoul, Republic of Korea
- Department of Nutritional Science and Food Management, Graduate Program in System Health Science and Engineering, Ewha Womans University, Seoul, Republic of Korea
- * E-mail: (OK); (JYK)
| |
Collapse
|
14
|
White AA, Lin A, Bickendorf X, Cavve BS, Moore JK, Siafarikas A, Strickland DH, Leffler J. Potential immunological effects of gender-affirming hormone therapy in transgender people - an unexplored area of research. Ther Adv Endocrinol Metab 2022; 13:20420188221139612. [PMID: 36533187 PMCID: PMC9747891 DOI: 10.1177/20420188221139612] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 10/31/2022] [Indexed: 12/14/2022] Open
Abstract
There are well-described sex-based differences in how the immune system operates. In particular, cisgender (cis) females have a more easily activated immune system; associated with an increased prevalence of autoimmune diseases and adverse events following vaccinations. Conversely, cis males have a higher threshold for immune activation, and are more prone to certain infectious diseases, such as coronavirus disease (COVID-19). Oestrogen and testosterone have immune-modulatory properties, and it is likely that these contribute to the sexual dimorphism of the immune system. There are also important immune-related genes located on the X chromosome, such as toll-like receptor (TLR) 7/8; and the mosaic bi-allelic expression of such genes may contribute to the state of immune hyperactivation in cis females. The scientific literature strongly suggests that sex-based differences in the functioning of the immune system are related to both X-linked genes and immune modulation by sex hormones. However, it is currently not clear how this impacts transgender (trans) people receiving gender-affirming hormonal therapy. Moreover, it is estimated that in Australia, at least 2.3% of adolescents identify as trans and/or gender diverse, and referrals to specialist gender-affirming care are increasing each year. Despite the improving social awareness of trans people, they remain chronically underrepresented in the scientific literature. In addition, a small number of case studies describe new onset autoimmune disorders in adult trans females following oestrogen use. However, there is currently minimal long-term research with an immunological focus on trans people. Therefore, to ensure the positive health outcomes of trans people, it is crucial that the role of sex hormones in immune modulation is investigated further.
Collapse
Affiliation(s)
- Alice A. White
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
| | - Ashleigh Lin
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
| | - Xander Bickendorf
- Telethon Kids Institute, University of Western Australia, WA, Australia
- Gender Diversity Service, Child and Adolescent Health Service, Nedlands, WA, Australia
| | - Blake S. Cavve
- Gender Diversity Service, Child and Adolescent Health Service, Nedlands, WA, Australia
| | - Julia K. Moore
- Gender Diversity Service, Child and Adolescent Health Service, Nedlands, WA, Australia
- School of Psychiatry and Clinical Neurosciences, University of Western Australia, Nedlands, WA, Australia
| | - Aris Siafarikas
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
- Gender Diversity Service, Child and Adolescent Health Service, Nedlands, WA, Australia
- Paediatrics, Medical School, The University of Western Australia, Nedlands, WA, Australia
| | | | - Jonatan Leffler
- Telethon Kids Institute, University of Western Australia, Perth Children’s Hospital, 15 Hospital Ave., Nedlands, WA 6009, Australia
| |
Collapse
|
15
|
Pátková Ž, Schwambergová D, Třebická Fialová J, Třebický V, Stella D, Kleisner K, Havlíček J. Attractive and healthy-looking male faces do not show higher immunoreactivity. Sci Rep 2022; 12:18432. [PMID: 36319732 PMCID: PMC9626598 DOI: 10.1038/s41598-022-22866-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 10/20/2022] [Indexed: 01/01/2023] Open
Abstract
Previous research has indicated that facial attractiveness may provide cues to the functioning of the immune system. Mating with individuals who have a more effective immune system could lead to a higher reproductive success. Our main aim was to test a possible association between immunoreactivity (stimulated by vaccination) and perceived facial attractiveness and healthiness. We experimentally activated the immune system of healthy men using vaccination against hepatitis A/B and meningococcus and measured levels of specific antibodies (markers of immune system reactivity) before and 30 days after the vaccination. Further, 1 day before the vaccination, we collected their facial photographs that were judged by females for attractiveness, healthiness, and facial skin patches for healthiness. In view of its proposed connection with the functioning of the immune system, we also measured skin colouration (both from the facial photographs and in vivo using a spectrophotometer) and we assessed its role in attractiveness and healthiness judgements. Moreover, we measured the levels of steroid hormones (testosterone and cortisol) and the percentage of adipose tissue, because both are known to have immunomodulatory properties and are related to perceived facial attractiveness and healthiness. We found no significant associations between antibody levels induced by vaccination and perceived facial attractiveness, facial healthiness, or skin healthiness. We also found no significant connections between steroid hormone levels, the amount of adipose tissue, rated characteristics, and antibody levels, except for a small negative effect of cortisol levels on perceived facial healthiness. Higher forehead redness was perceived as less attractive and less healthy and higher cheek patch redness was perceived as less healthy, but no significant association was found between antibody levels and facial colouration. Overall, our results suggest that perceived facial attractiveness, healthiness, and skin patch healthiness provide limited cues to immunoreactivity, and perceived characteristics seem to be related only to cortisol levels and facial colouration.
Collapse
Affiliation(s)
- Žaneta Pátková
- Faculty of Science, Charles University, Viničná 7, 128 44, Prague, Czech Republic.
| | - Dagmar Schwambergová
- Faculty of Science, Charles University, Viničná 7, 128 44, Prague, Czech Republic
| | | | - Vít Třebický
- Faculty of Physical Education and Sport, Charles University, José Martího 269, 162 52, Prague, Czech Republic
| | - David Stella
- Faculty of Science, Charles University, Viničná 7, 128 44, Prague, Czech Republic
- Global Change Research Institute of the Czech Academy of Sciences, Bělidla 986/4a, 603 00, Brno, Czech Republic
| | - Karel Kleisner
- Faculty of Science, Charles University, Viničná 7, 128 44, Prague, Czech Republic
| | - Jan Havlíček
- Faculty of Science, Charles University, Viničná 7, 128 44, Prague, Czech Republic
| |
Collapse
|
16
|
Sayaf K, Gabbia D, Russo FP, De Martin S. The Role of Sex in Acute and Chronic Liver Damage. Int J Mol Sci 2022; 23:10654. [PMID: 36142565 PMCID: PMC9505609 DOI: 10.3390/ijms231810654] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/07/2022] [Accepted: 09/09/2022] [Indexed: 11/16/2022] Open
Abstract
Acute and chronic hepatic damages are caused by xenobiotics or different diseases affecting the liver, characterized by different etiologies and pathological features. It has been demonstrated extensively that liver damage progresses differently in men and women, and some chronic liver diseases show a more favorable prognosis in women than in men. This review aims to update the most recent advances in the comprehension of the molecular basis of the sex difference observed in both acute and chronic liver damage. With this purpose, we report experimental studies on animal models and clinical observations investigating both acute liver failure, e.g., drug-induced liver injury (DILI), and chronic liver diseases, e.g., viral hepatitis, alcoholic liver disease (ALD), non-alcoholic fatty liver disease (NAFLD), autoimmune liver diseases, and hepatocellular carcinoma (HCC).
Collapse
Affiliation(s)
- Katia Sayaf
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35131 Padova, Italy
| | - Daniela Gabbia
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy
| | - Francesco Paolo Russo
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35131 Padova, Italy
- Gastroenterology and Multivisceral Transplant Units, Azienda Ospedale—Università di Padova, 35131 Padova, Italy
| | - Sara De Martin
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy
| |
Collapse
|
17
|
Premenopausal and postmenopausal women during the COVID-19 pandemic. PRZEGLAD MENOPAUZALNY = MENOPAUSE REVIEW 2022; 21:200-206. [PMID: 36254124 PMCID: PMC9551364 DOI: 10.5114/pm.2022.118695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 03/19/2022] [Indexed: 01/09/2023]
Abstract
The current global COVID-19 mortality rate is estimated to be around 3.4%; however, it is dependent on age, sex, and comorbidities. Epidemiological evidence shows gender disparities in COVID-19 severity and fatality, with non-menopausal females having milder severity and better outcomes than age-matched males. However, the difference vanishes when comparing postmenopausal women with age-matched men. It has been suggested that, to some extent, this is due to the protective role of female hormones, such as anti-Müllerian hormone and oestradiol (E2), in non-menopausal women. Oestrogens have been hypothesized to be crucial in modulating viral infection and the progression of the disease via an action on immune/inflammatory responses and angiotensin-converting enzyme type 2 expression. Hence, the most likely explanation is that, because the levels of oestrogen in females after menopause decrease, oestrogen no longer offers a beneficial effect as seen in younger females. The COVID-19 pandemic has highlighted the serious negative effects arising from the state of E2 deficiency. Therefore, hormone replacement therapy gains further support as the damaging effect of the decline in ovarian function affects many biological systems, and recently with the COVID-19 pandemic, oestrogen's vital role within the immune system has become quite clear. However, additional clinical investigations regarding hormone replacement therapy are urgently needed to further verify the protective and therapeutic effects of E2 on menopausal women with COVID-19.
Collapse
|
18
|
Nytrova P, Dolezal O. Sex bias in multiple sclerosis and neuromyelitis optica spectrum disorders: How it influences clinical course, MRI parameters and prognosis. Front Immunol 2022; 13:933415. [PMID: 36016923 PMCID: PMC9396644 DOI: 10.3389/fimmu.2022.933415] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 07/19/2022] [Indexed: 11/13/2022] Open
Abstract
This review is a condensed summary of representative articles addressing the sex/gender bias in multiple sclerosis (MS) and neuromyelitis optica spectrum disorders (NMOSD). The strong effects of sex on the incidence and possibly also the activity and progression of these disorders should be implemented in the evaluation of any phase of clinical research and also in treatment choice consideration in clinical practice and evaluation of MRI parameters. Some relationships between clinical variables and gender still remain elusive but with further understanding of sex/gender-related differences, we should be able to provide appropriate patient-centered care and research.
Collapse
Affiliation(s)
- Petra Nytrova
- Department of Neurology and Centre of Clinical Neuroscience, First Faculty of Medicine, Charles University in Prague and General University Hospital, Prague, Czechia
- *Correspondence: Petra Nytrova,
| | - Ondrej Dolezal
- Department of Neurology, Dumfries and Galloway Royal Infirmary, NHS Scotland, Dumfries, United Kingdom
| |
Collapse
|
19
|
Averyanova M, Vishnyakova P, Yureneva S, Yakushevskaya O, Fatkhudinov T, Elchaninov A, Sukhikh G. Sex hormones and immune system: Menopausal hormone therapy in the context of COVID-19 pandemic. Front Immunol 2022; 13:928171. [PMID: 35983046 PMCID: PMC9379861 DOI: 10.3389/fimmu.2022.928171] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
The fatal outcomes of COVID-19 are related to the high reactivity of the innate wing of immunity. Estrogens could exert anti-inflammatory effects during SARS-CoV-2 infection at different stages: from increasing the antiviral resistance of individual cells to counteracting the pro-inflammatory cytokine production. A complex relationship between sex hormones and immune system implies that menopausal hormone therapy (MHT) has pleiotropic effects on immunity in peri- and postmenopausal patients. The definite immunological benefits of perimenopausal MHT confirm the important role of estrogens in regulation of immune functionalities. In this review, we attempt to explore how sex hormones and MHT affect immunological parameters of the organism at different level (in vitro, in vivo) and what mechanisms are involved in their protective response to the new coronavirus infection. The correlation of sex steroid levels with severity and lethality of the disease indicates the potential of using hormone therapy to modulate the immune response and increase the resilience to adverse outcomes. The overall success of MHT is based on decades of experience in clinical trials. According to the current standards, MHT should not be discontinued in COVID-19 with the exception of critical cases.
Collapse
Affiliation(s)
- Marina Averyanova
- National Medical Research Center for Obstetrics, Gynecology and Perinatology named after Academician V. I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia
| | - Polina Vishnyakova
- National Medical Research Center for Obstetrics, Gynecology and Perinatology named after Academician V. I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia
- Peoples’ Friendship University of Russia, Medical Institute, Moscow, Russia
| | - Svetlana Yureneva
- National Medical Research Center for Obstetrics, Gynecology and Perinatology named after Academician V. I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia
| | - Oksana Yakushevskaya
- National Medical Research Center for Obstetrics, Gynecology and Perinatology named after Academician V. I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia
| | - Timur Fatkhudinov
- Peoples’ Friendship University of Russia, Medical Institute, Moscow, Russia
- A. P. Avtsyn Research Institute of Human Morphology, Laboratory of Growth and Development, Moscow, Russia
| | - Andrey Elchaninov
- National Medical Research Center for Obstetrics, Gynecology and Perinatology named after Academician V. I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia
| | - Gennady Sukhikh
- National Medical Research Center for Obstetrics, Gynecology and Perinatology named after Academician V. I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia
| |
Collapse
|
20
|
Sex Steroids Effects on Asthma: A Network Perspective of Immune and Airway Cells. Cells 2022; 11:cells11142238. [PMID: 35883681 PMCID: PMC9318292 DOI: 10.3390/cells11142238] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 07/13/2022] [Accepted: 07/17/2022] [Indexed: 11/17/2022] Open
Abstract
A multitude of evidence has suggested the differential incidence, prevalence and severity of asthma between males and females. A compilation of recent literature recognized sex differences as a significant non-modifiable risk factor in asthma pathogenesis. Understanding the cellular and mechanistic basis of sex differences remains complex and the pivotal point of this ever elusive quest, which remains to be clarified in the current scenario. Sex steroids are an integral part of human development and evolution while also playing a critical role in the conditioning of the immune system and thereby influencing the function of peripheral organs. Classical perspectives suggest a pre-defined effect of sex steroids, generalizing estrogens popularly under the “estrogen paradox” due to conflicting reports associating estrogen with a pro- and anti-inflammatory role. On the other hand, androgens are classified as “anti-inflammatory,” serving a protective role in mitigating inflammation. Although considered mainstream and simplistic, this observation remains valid for numerous reasons, as elaborated in the current review. Women appear immune-favored with stronger and more responsive immune elements than men. However, the remarkable female predominance of diverse autoimmune and allergic diseases contradicts this observation suggesting that hormonal differences between the sexes might modulate the normal and dysfunctional regulation of the immune system. This review illustrates the potential relationship between key elements of the immune cell system and their interplay with sex steroids, relevant to structural cells in the pathophysiology of asthma and many other lung diseases. Here, we discuss established and emerging paradigms in the clarification of observed sex differences in asthma in the context of the immune system, which will deepen our understanding of asthma etiopathology.
Collapse
|
21
|
Thomas N, Gurvich C, Huang K, Gooley PR, Armstrong CW. The underlying sex differences in neuroendocrine adaptations relevant to Myalgic Encephalomyelitis Chronic Fatigue Syndrome. Front Neuroendocrinol 2022; 66:100995. [PMID: 35421511 DOI: 10.1016/j.yfrne.2022.100995] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 02/27/2022] [Accepted: 03/29/2022] [Indexed: 11/16/2022]
Abstract
INTRODUCTION Myalgic Encephalomyelitis/ Chronic Fatigue Syndrome (ME/CFS) is a complex multisystem disease characterised by severe and disabling new-onset symptoms of post-exertional malaise (PEM), fatigue, brain fog, and sleep dysfunction that lasts for at least six months. Accumulating evidence suggests that sex and endocrine events have a significant influence on symptom onset and moderation of ME/CFS, with female sex being one of the most consistent and credible predictive risk factors associated with diagnosis. Such sex differences suggest sex chromosomes and sex steroids may play a part in the development of the condition or moderation of symptoms, although this has yet to be explored in detail. METHODS/AIMS This narrative review outlines sex differences in ME/CFS in terms of vulnerability factors and clinical phenotype and explores the known sex differences in neuroendocrine systems affected in ME/CFS and how this may relate to disease risk, onset, pathophysiology, and potential treatment avenues. CONCLUSIONS There is clear evidence of a sex dimorphism with regards to prevalence (3:1 female preponderance), clinical phenotypes, and aetiological triggers prior to symptom onset of ME/CFS. Endocrinological events, particularly those throughout the female lifespan, are associated with ME/CFS and include reproductive menstrual cycle fluctuations, pregnancy, post-partum and perimenopause. Further, there is evidence for gonadal sex, adrenal stress and renal neuroendocrine systems as implicated in ME/CFS, including changes in estrogen, progesterone compounds, aldosterone, and cortisol levels, of which there are established sex differences. The broad effects of steroid hormones on the physiological systems may also speak to the diversity of ME/CFS symptomatology observed in patients. Further attention must be paid to sex, age, and steroid biology in ME/CFS.
Collapse
Affiliation(s)
- Natalie Thomas
- Department of Biochemistry & Pharmacology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Australia.
| | - Caroline Gurvich
- Department of Psychiatry, Faculty of Medicine, Nursing and Health Sciences, Monash University, Australia
| | - Katherine Huang
- Department of Biochemistry & Pharmacology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Australia
| | - Paul R Gooley
- Department of Biochemistry & Pharmacology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Australia
| | - Christopher W Armstrong
- Department of Biochemistry & Pharmacology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Australia
| |
Collapse
|
22
|
Valle-Millares D, Brochado-Kith Ó, Gómez-Sanz A, Martín-Carbonero L, Ryan P, De Los Santos I, Castro JM, Troya J, Mayoral-Muñoz M, Cuevas G, Martínez-Román P, Sanz-Sanz J, Muñoz-Muñoz M, Jiménez-Sousa MÁ, Resino S, Briz V, Fernández-Rodríguez A. HCV eradication with DAAs differently affects HIV males and females: A whole miRNA sequencing characterization. Biomed Pharmacother 2021; 145:112405. [PMID: 34781145 DOI: 10.1016/j.biopha.2021.112405] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/31/2021] [Accepted: 11/03/2021] [Indexed: 12/12/2022] Open
Abstract
Gender-specific consequences after HCV eradication are unexplored. MicroRNAs (miRNAs) play a crucial role in the immune response against viral infections. However, few have highlighted miRNA role in sex-biased disease or therapy response. We aim to assess gender differences reflected in the miRNA expression of HIV/HCV-coinfected patients who achieve sustained virological response (SVR) with direct acting antivirals (DAAs). We conducted a prospective study of miRNA expression in PBMCs from 28 chronic HIV/HCV-coinfected patients (HIV/HCV) at baseline and after achieving SVR with DAAs. Sixteen HIV-monoinfected patients (HIV) and 36 healthy controls (HC) were used as controls. Identification of significant differentially expressed (SDE) miRNAs was performed with generalized linear model and mixed GLMs. We also explored putative dysregulated biological pathways. At baseline, the HIV/HCV patients showed differences in the miRNA profile concerning the HIV group (165 and 102 SDE miRNAs for males and females, respectively). Gender-stratified analysis of HIV/HCV group at baseline versus at SVR achievement showed higher differences in males (80 SDE miRNAs) than in females (55 SDE miRNAs). After SVR, HIV/HCV group showed similar values to HIV individuals, especially in females (1 SDE miRNA). However, ten miRNAs in males remained dysregulated, which were mainly involved in cancer, fatty acid, and inflammatory pathways. Taken together, our results show gender-biased dysregulation in the miRNA expression profile of PBMCs after HCV eradication with DAAs. These differences were normalized in females, while miRNA profile and their target-related pathways in males lack of normalization, which may be related to a high-risk of developing liver-related complications.
Collapse
Affiliation(s)
- Daniel Valle-Millares
- Unit of Viral Infection and Immunity, National Center for Microbiology, Institute of Health Carlos III, Majadahonda, Madrid, Spain
| | - Óscar Brochado-Kith
- Unit of Viral Infection and Immunity, National Center for Microbiology, Institute of Health Carlos III, Majadahonda, Madrid, Spain
| | - Alicia Gómez-Sanz
- Unit of Viral Infection and Immunity, National Center for Microbiology, Institute of Health Carlos III, Majadahonda, Madrid, Spain
| | | | - Pablo Ryan
- Internal Medicine Service, Infanta Leonor Teaching Hospital, Madrid, Spain
| | - Ignacio De Los Santos
- Servicio de Medicina Interna-Infecciosas, Hospital Universitario de La Princesa, Madrid, Spain
| | - Juan M Castro
- Hospital La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
| | - Jesús Troya
- Internal Medicine Service, Infanta Leonor Teaching Hospital, Madrid, Spain
| | | | - Guillermo Cuevas
- Internal Medicine Service, Infanta Leonor Teaching Hospital, Madrid, Spain
| | - Paula Martínez-Román
- Unit of Viral Infection and Immunity, National Center for Microbiology, Institute of Health Carlos III, Majadahonda, Madrid, Spain
| | - Jesús Sanz-Sanz
- Servicio de Medicina Interna-Infecciosas, Hospital Universitario de La Princesa, Madrid, Spain
| | - María Muñoz-Muñoz
- Department of Animal Breeding, Instituto Nacional de Investigación y Alimentación Agraria y Alimentaria (INIA), Madrid, Spain
| | - María Á Jiménez-Sousa
- Unit of Viral Infection and Immunity, National Center for Microbiology, Institute of Health Carlos III, Majadahonda, Madrid, Spain
| | - Salvador Resino
- Unit of Viral Infection and Immunity, National Center for Microbiology, Institute of Health Carlos III, Majadahonda, Madrid, Spain
| | - Verónica Briz
- Laboratory of Reference and Research on Viral Hepatitis, National Center for Microbiology, Institute of Health Carlos III, Majadahonda, Madrid, Spain
| | - Amanda Fernández-Rodríguez
- Unit of Viral Infection and Immunity, National Center for Microbiology, Institute of Health Carlos III, Majadahonda, Madrid, Spain; Department of Medicine, Alfonso X el Sabio, Villanueva de la Cañada, 28691 Madrid, Spain.
| |
Collapse
|
23
|
Caffo O, Messina M, Veccia A, Kinspergher S, Maines F, Messina C. Severe acute respiratory syndrome coronavirus 2 infection in patients with prostate cancer: A critical review. Crit Rev Oncol Hematol 2021; 167:103491. [PMID: 34626792 PMCID: PMC8492888 DOI: 10.1016/j.critrevonc.2021.103491] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/30/2021] [Accepted: 10/01/2021] [Indexed: 12/27/2022] Open
Abstract
Real-world data suggest a possible interplay between androgen deprivation therapy (ADT) and susceptibility to and the severity of SARS-CoV-2 infection. As ADT is the backbone of prostate cancer treatment, various authors have evaluated different patient cohorts but the evidence provided is conflicting. The aim of this review is to assess the available publications concerning the role of ADT in preventing or reducing the severity of SARS-CoV-2 infection. After a literature search we identified four full papers, five letters, and four meeting abstracts, but these used different search methods and the quality of the evidence varied. They frequently had different endpoints, did not report the status of the prostate cancer patients and evaluated heterogeneous populations. The available data do not support the view that ADT protects against SARS-CoV-2 infection. Larger and more precise studies are warranted, considering variables that affect infection outcomes as these significantly influence the reliability of the findings.
Collapse
Affiliation(s)
- Orazio Caffo
- Medical OncologyDepartments of Santa Chiara Hospital, Trento, Italy,Corresponding author at: Department of Medical Oncology, Santa Chiara Hospital, Largo Medaglie d’Oro 9, Trento, 38122, Italy
| | - Marco Messina
- Medical Oncology Departments of Civic Hospital, Palermo, Italy
| | - Antonello Veccia
- Medical OncologyDepartments of Santa Chiara Hospital, Trento, Italy
| | | | - Francesca Maines
- Medical OncologyDepartments of Santa Chiara Hospital, Trento, Italy
| | - Carlo Messina
- Medical Oncology Departments of Civic Hospital, Palermo, Italy
| |
Collapse
|
24
|
Cocomazzi G, Piazzolla V, Squillante MM, Antinucci S, Giambra V, Giuliani F, Maiorana A, Serra N, Mangia A. Early Serological Response to BNT162b2 mRNA Vaccine in Healthcare Workers. Vaccines (Basel) 2021; 9:913. [PMID: 34452038 PMCID: PMC8402545 DOI: 10.3390/vaccines9080913] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 08/03/2021] [Accepted: 08/13/2021] [Indexed: 12/16/2022] Open
Abstract
PURPOSE Clinical significance and durability of serological response after mRNA COVID-19 vaccines is under investigation. Data on early virological response are limited. To iden-tify potential predictors of antibody durability, circulating antibody levels were longitudinally ex-plored in healthcare workers included in a follow-up program for SARS-CoV-2 infection. Meth-ods: Subjects meeting the inclusion criteria signed an informed consent. Serum samples were col-lected at baseline, before the first BNT162b2 vaccine, at days 7, 21, 31, 90, and 180 days after the first dose. Serological evaluation was performed by QuantiVac Euroimmune anti-S1 antibody as-say. Only subjects followed-up until day 90 are here considered. RESULTS Of 340 taken into consid-eration, 265 subjects were naive, and 75 COVID-19 experienced. The former showed a progres-sive increase in their antibody levels before day 90 decline, while the latter showed antibody levels reaching a plateau at day 7 and slightly declining at day 90. All showed antibody levels higher than the assay cut-off at day 31 and 90. Among naive, 108 had an early response whose predic-tors were younger age and female gender (OR 0.94, 95% CI 0.91-0.96, p < 0.0001; and OR 2.58, 95% CI 1.48-4.51, p = 0.0009). Naive subjects experienced a day 30/90 decline in antibody levels, whereas experienced did not. Early response was an independent predictor of higher day 30/90 antibody levels decline (OR = 2.05, 95% CI 1.04-4.02; p = 0.037). CONCLUSIONS Our results suggest that in healthcare workers early response might be inversely associated with antibody levels 90 days after BNT162b2 vaccine.
Collapse
Affiliation(s)
- Giovanna Cocomazzi
- Liver Unit, Fondazione IRCCS “Casa Sollievo della Sofferenza”, 71013 San Giovanni Rotondo (Fg), Italy; (G.C.); (V.P.); (M.M.S.)
| | - Valeria Piazzolla
- Liver Unit, Fondazione IRCCS “Casa Sollievo della Sofferenza”, 71013 San Giovanni Rotondo (Fg), Italy; (G.C.); (V.P.); (M.M.S.)
| | - Maria Maddalena Squillante
- Liver Unit, Fondazione IRCCS “Casa Sollievo della Sofferenza”, 71013 San Giovanni Rotondo (Fg), Italy; (G.C.); (V.P.); (M.M.S.)
| | | | - Vincenzo Giambra
- Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT) Fondazione “Casa Sollievo della Sofferenza”, 71013 San Giovanni Rotondo (Fg), Italy;
| | - Francesco Giuliani
- ICT Innovation and Research Unit, Fondazione IRCCS “Casa Sollievo della Sofferenza”,
71013 San Giovanni Rotondo (Fg), Italy;
| | - Alberto Maiorana
- GSSL Unit, Fondazione “Casa Sollievo della Sofferenza”, 71013 San Giovanni Rotondo (Fg), Italy;
| | - Nicola Serra
- Department of Public Health, University “Federico II”, 80131 Naples, Italy;
| | - Alessandra Mangia
- Liver Unit, Fondazione IRCCS “Casa Sollievo della Sofferenza”, 71013 San Giovanni Rotondo (Fg), Italy; (G.C.); (V.P.); (M.M.S.)
| |
Collapse
|
25
|
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: 3.5] [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
|
26
|
Duffy D, Yourkavitch J, Bruinvels G, Rinaldi NJ, Wideman L. The development and initial validation of the Health and Reproductive Survey (HeRS). ACTA ACUST UNITED AC 2021; 17:17455065211004814. [PMID: 34348519 PMCID: PMC8358484 DOI: 10.1177/17455065211004814] [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] [Indexed: 11/06/2022]
Abstract
Background: Due to the diversity in profiles associated with the female reproductive cycle and their potential physiological and psychological effects, monitoring the reproductive status of exercising females is important from a practical and research perspective. Moreover, as physical activity can influence menstrual function, the effects of physical activity energy expenditure on reproductive function should also be considered. Aim: The aim of this study was to develop and establish initial face and content validity of the Health and Reproductive Survey (HeRS) for physically active females, which is a retrospective assessment of menstrual function from menarche (first menstruation) to menopause (cessation of menstruation). Methods: Face validity was evaluated qualitatively, and the initial content validity was established through a principal component analysis. The face validity process was completed by 26 females aged 19–67 years and the content validity was established through a survey sent to a convenience sample of 392 females, of which 230 females (57.9% and aged 18–49 years) completed the survey. Results: The revisions made following the face validation improved the understanding, flow, and coherence of the survey. The principal component analysis indicated that, at a minimum, the survey measures these constructs: menstrual cessation and associated moderators, athletic participation and performance levels (as associated with menstruation change and the menstrual cycle), age and menstrual cessation, hormonal contraception (“birth control”), and menarche and associated moderators. Conclusion: The Health and Reproductive Survey (HeRS) is a partially validated tool that can be used by researchers to characterize the menstrual status of physically active females relative to their physical activity status.
Collapse
Affiliation(s)
- Donna Duffy
- Center for Women's Health and Wellness, University of North Carolina at Greensboro, Greensboro, NC, USA.,Department of Kinesiology, University of North Carolina at Greensboro, Greensboro, NC, USA
| | - Jennifer Yourkavitch
- Department of Public Health Education, University of North Carolina, at Greensboro, Greensboro, NC, USA
| | - Georgie Bruinvels
- Faculty of Sport, Health and Applied Science, St. Mary's University, Twickenham, UK.,Orreco, Business Innovation Unit, National University of Ireland, Galway, Ireland
| | - Nicola J Rinaldi
- NPNW Consulting, Lexington, MA, USA.,Antica Press LLC, Waltham, MA, USA
| | - Laurie Wideman
- Department of Kinesiology, University of North Carolina at Greensboro, Greensboro, NC, USA
| |
Collapse
|
27
|
Cardiac Immunology: A New Era for Immune Cells in the Heart. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021. [PMID: 32910424 DOI: 10.1007/5584_2020_576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/10/2023]
Abstract
The immune system is essential for the development and homeostasis of the human body. Our current understanding of the immune system on disease pathogenesis has drastically expanded over the last decade with the definition of additional non-canonical roles in various tissues. Recently, tissue-resident immune cells have become an important research topic for understanding their roles in the prevention, pathogenesis, and recovery from the diseases. Heart resident immune cells, particularly macrophage subtypes, and their characteristic morphology, distribution in the cardiac tissue, and transcriptional profile have been recently reported in the experimental animal models, unrevealing novel and unexpected roles in electrophysiological regulation of the heart both at the steady-state and diseased state. Immunological processes have been widely studied in both sterile cardiac disorders, such as myocardial infarction, autoimmune cardiac diseases, or infectious cardiac diseases, such as myocarditis, endocarditis, and acute rheumatic carditis. Following cardiac injury, innate and adaptive immunity have critical roles in pro- and anti-inflammatory processes. Heart resident immune cells not only provide defense against infectious diseases but also contribute to the homeostasis. In recent years, physiological changes and pathological processes were demonstrated to alter the abundance, distribution, polarization, and diversity of immune cells in the heart. Accumulating evidence indicates that cardiac remodeling is controlled by the complex crosstalk between cardiomyocytes and cardiac immune cells through the gap junctions, providing the ion flow to achieve synchronization and modulation of contractility. This review article aims to review the well-documented roles of both resident and recruited immune cell in the heart, as well as their recently uncovered unconventional roles in both cardiac homeostasis and cardiovascular diseases. We have mostly focused on studies on animal models used in preclinical research, underlying the need for further investigations in humans or in vitro human models. It may be foreseen that the further comprehensive investigations of cardiac immunology might harbor new therapeutic options for cardiac disorders that have tremendous medical potential.
Collapse
|
28
|
Ye L, Huang W, Liu S, Cai S, Hong L, Xiao W, Thiele K, Zeng Y, Song M, Diao L. Impacts of Immunometabolism on Male Reproduction. Front Immunol 2021; 12:658432. [PMID: 34367130 PMCID: PMC8334851 DOI: 10.3389/fimmu.2021.658432] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 06/29/2021] [Indexed: 12/24/2022] Open
Abstract
The physiological process of male reproduction relies on the orchestration of neuroendocrine, immune, and energy metabolism. Spermatogenesis is controlled by the hypothalamic-pituitary-testicular (HPT) axis, which modulates the production of gonadal steroid hormones in the testes. The immune cells and cytokines in testes provide a protective microenvironment for the development and maturation of germ cells. The metabolic cellular responses and processes in testes provide energy production and biosynthetic precursors to regulate germ cell development and control testicular immunity and inflammation. The metabolism of immune cells is crucial for both inflammatory and anti-inflammatory responses, which supposes to affect the spermatogenesis in testes. In this review, the role of immunometabolism in male reproduction will be highlighted. Obesity, metabolic dysfunction, such as type 2 diabetes mellitus, are well documented to impact male fertility; thus, their impacts on the immune cells distributed in testes will also be discussed. Finally, the potential significance of the medicine targeting the specific metabolic intermediates or immune metabolism checkpoints to improve male reproduction will also be reassessed.
Collapse
Affiliation(s)
- Lijun Ye
- Shenzhen Key Laboratory for Reproductive Immunology of Peri-implantation, Clinical Research Center for Reproductive Medicine, Shenzhen Zhongshan Urology Hospital, Shenzhen, China
| | - Wensi Huang
- Shenzhen Key Laboratory for Reproductive Immunology of Peri-implantation, Clinical Research Center for Reproductive Medicine, Shenzhen Zhongshan Urology Hospital, Shenzhen, China
| | - Su Liu
- Shenzhen Key Laboratory for Reproductive Immunology of Peri-implantation, Clinical Research Center for Reproductive Medicine, Shenzhen Zhongshan Urology Hospital, Shenzhen, China
| | - Songchen Cai
- Shenzhen Key Laboratory for Reproductive Immunology of Peri-implantation, Clinical Research Center for Reproductive Medicine, Shenzhen Zhongshan Urology Hospital, Shenzhen, China
| | - Ling Hong
- Shenzhen Key Laboratory for Reproductive Immunology of Peri-implantation, Clinical Research Center for Reproductive Medicine, Shenzhen Zhongshan Urology Hospital, Shenzhen, China
| | - Weiqiang Xiao
- Shenzhen Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen, China
| | - Kristin Thiele
- Division of Experimental Feto-Maternal Medicine, Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Yong Zeng
- Shenzhen Key Laboratory for Reproductive Immunology of Peri-implantation, Clinical Research Center for Reproductive Medicine, Shenzhen Zhongshan Urology Hospital, Shenzhen, China
| | - Mingzhe Song
- Shenzhen Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen, China
| | - Lianghui Diao
- Shenzhen Key Laboratory for Reproductive Immunology of Peri-implantation, Clinical Research Center for Reproductive Medicine, Shenzhen Zhongshan Urology Hospital, Shenzhen, China
| |
Collapse
|
29
|
Ghare Naz MS, Banaei M, Dashti S, Tehrani FR. An overview of sex hormones in relation to SARS-CoV-2 infection. Future Virol 2021. [PMID: 34306167 PMCID: PMC8293688 DOI: 10.2217/fvl-2021-0058] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 07/01/2021] [Indexed: 01/03/2023]
Abstract
Aim: Sex differences in COVID-19 outcomes might be explained from a sex hormones (SexHs) perspective. Materials & methods: PubMed, Scopus, Web of Science, EMBASE and Google Scholar were searched up to March 2021. Results: Based on the literature review, the crosstalk between SexHs (estrogens, progesterone and testosterone), their receptors (estrogen α and β, androgen, and progesterone) and the immune system shaped the sex-related differences in immune responses against COVID-19. Differential production of SexHs over the lifespan (during pregnancy, reproductive years, menopause and andropause) and over different seasons may result in disparities in body response toward COVID-19. Moreover, SexHs-specific differences might affect vaccine efficacy and response to treatment. Conclusion: The roles of SexHs need to be considered in vaccine development and even treatment of COVID-19.
Collapse
Affiliation(s)
- Marzieh Saei Ghare Naz
- Reproductive Endocrinology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, 1985717413, Iran
| | - Mojdeh Banaei
- Mother & Child Welfare Research Center, Hormozgan University of Medical Sciences, Bandar Abbas, 7916613885, Iran
| | - Sareh Dashti
- Department of Midwifery, Mashhad Branch, Islamic Azad University, Mashhad, 9187147578, Iran
| | - Fahimeh Ramezani Tehrani
- Reproductive Endocrinology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, 1985717413, Iran
| |
Collapse
|
30
|
Campesi I, Montella A, Franconi F. Human monocytes respond to lipopolysaccharide (LPS) stimulation in a sex-dependent manner. J Cell Physiol 2021; 237:580-588. [PMID: 34252202 PMCID: PMC9292909 DOI: 10.1002/jcp.30503] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 06/28/2021] [Accepted: 06/29/2021] [Indexed: 01/08/2023]
Abstract
Monocytes play a critical role in inflammation and immune response, their activity being sex‐dependent. However, the basis of sex differences is not well understood. Therefore, we investigated the lipopolysaccharide (LPS) effects on tumor necrosis factor‐α (TNF‐α) release, autophagy, and chemotaxis in freshly isolated monocytes from healthy young men and women. In basal conditions, male and female monocytes had similar TNF‐α release, chemotaxis, and estrogen receptors (ER‐α) and ER‐β expression, while the LC3II/I ratio was significantly higher in males. LPS treatment induced qualitative and quantitative sex differences. It reduced autophagy and increased TNF‐α release only in male monocytes, while, chemotaxis was significantly influenced only in female cells. Moreover, it reduced the expression of ER‐α only in female cells, while ER‐β expression was reduced in both sexes, but more markedly in female cells. Finally, the interplay between LPS treatment and 17‐β‐estradiol (E2) was present only in female cells. Globally, these findings expand the concept that sex plays a role in regulating monocytes' functions, being sex differences cell‐ and parameter‐specific.
Collapse
Affiliation(s)
- Ilaria Campesi
- Dipartimento di Scienze Biomediche, Università Degli Studi di Sassari, Sassari, Italy.,Laboratorio Nazionale di Medicina e Farmacologia di Genere, Istituto Nazionale Biostrutture Biosistemi, Sassari, Italy
| | - Andrea Montella
- Dipartimento di Scienze Biomediche, Università Degli Studi di Sassari, Sassari, Italy
| | - Flavia Franconi
- Laboratorio Nazionale di Medicina e Farmacologia di Genere, Istituto Nazionale Biostrutture Biosistemi, Sassari, Italy
| |
Collapse
|
31
|
Pillerová M, Borbélyová V, Hodosy J, Riljak V, Renczés E, Frick KM, Tóthová Ľ. On the role of sex steroids in biological functions by classical and non-classical pathways. An update. Front Neuroendocrinol 2021; 62:100926. [PMID: 34089761 PMCID: PMC8523217 DOI: 10.1016/j.yfrne.2021.100926] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 05/29/2021] [Accepted: 05/31/2021] [Indexed: 12/02/2022]
Abstract
The sex steroid hormones (SSHs) play several roles in regulation of various processes in the cardiovascular, immune, muscular and neural systems. SSHs affect prenatal and postnatal development of various brain structures, including regions associated with important physiological, behavioral, cognitive, and emotional functions. This action can be mediated by either intracellular or transmembrane receptors. While the classical mechanisms of SSHs action are relatively well examined, the physiological importance of non-classical mechanism of SSHs action through membrane-associated and transmembrane receptors in the brain remains unclear. The most recent summary describing the role of SSHs in different body systems is lacking. Therefore, the aim of this review is to discuss classical and non-classical signaling pathways of testosterone and estradiol action via their receptors at functional, cellular, tissue level and to describe the effects on various body systems and behavior. Particular emphasis will be on brain regions including the hippocampus, hypothalamus, frontal cortex and cerebellum.
Collapse
Affiliation(s)
- Miriam Pillerová
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| | - Veronika Borbélyová
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| | - Július Hodosy
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| | - Vladimír Riljak
- Institute of Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Emese Renczés
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| | - Karyn M Frick
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Ľubomíra Tóthová
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia.
| |
Collapse
|
32
|
Abstract
Animal models with high translational validity are essential tools in understanding disease pathogenesis and in the development of therapeutic strategies. Multiple sclerosis (MS) is an autoimmune demyelinating disease of the central nervous system characterized by progressive neurological deficits and socioeconomic burden. Experimental autoimmune encephalomyelitis (EAE) is the most extensively utilized animal model of MS, with well-characterized rodent and non-human primate variants. The EAE model is typically induced by either active immunization with myelin-derived proteins or peptides in adjuvant or by passive transfer of activated myelin-specific CD4+ T lymphocytes. To date, the EAE model has been an essential tool in the development of at least seven U.S. Food and Drug Administration (FDA)-approved immunomodulatory drugs for the treatment of MS, including glatiramer acetate, fingolimod, and natalizumab. However, the translational validity of the EAE model is frequently compromised due to poor study design, inconsistent clinical scoring endpoints, and inappropriate statistical calculations. No single animal model accurately reflects the complexity of human MS pathogenesis. Beyond EAE, multiple additional animal models are described, including Theiler's murine encephalomyelitis virus and cuprizone-induced demyelination, which facilitate the study of pathogen-induced CNS autoimmunity and remyelination, respectively. This overview summarizes several of the most frequently used animal models of MS and highlights key factors that significantly influence the experimental outcome and affect translational validity. © 2021 Wiley Periodicals LLC.
Collapse
Affiliation(s)
- Paul Smith
- Incyte Research Institute, Wilmington, Delaware
| |
Collapse
|
33
|
Wehbe Z, Hammoud SH, Yassine HM, Fardoun M, El-Yazbi AF, Eid AH. Molecular and Biological Mechanisms Underlying Gender Differences in COVID-19 Severity and Mortality. Front Immunol 2021; 12:659339. [PMID: 34025658 PMCID: PMC8138433 DOI: 10.3389/fimmu.2021.659339] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 04/20/2021] [Indexed: 12/13/2022] Open
Abstract
Globally, over two million people have perished due to the recent pandemic caused by SARS-CoV-2. The available epidemiological global data for SARS-CoV-2 portrays a higher rate of severity and mortality in males. Analyzing gender differences in the host mechanisms involved in SARS-CoV-2 infection and progression may offer insight into the more detrimental disease prognosis and clinical outcome in males. Therefore, we outline sexual dimorphisms which exist in particular host factors and elaborate on how they may contribute to the pronounced severity in male COVID-19 patients. This includes disparities detected in comorbidities, the ACE2 receptor, renin-angiotensin system (RAS), signaling molecules involved in SARS-CoV-2 replication, proteases which prime viral S protein, the immune response, and behavioral considerations. Moreover, we discuss sexual disparities associated with other viruses and a possible gender-dependent response to SARS-CoV-2 vaccines. By specifically highlighting these immune-endocrine processes as well as behavioral factors that differentially exist between the genders, we aim to offer a better understanding in the variations of SARS-CoV-2 pathogenicity.
Collapse
Affiliation(s)
- Zena Wehbe
- Department of Biology, American University of Beirut, Beirut, Lebanon
| | - Safaa Hisham Hammoud
- Department of Pharmacology and Therapeutics, Beirut Arab University, Beirut, Lebanon
| | | | - Manal Fardoun
- Department of Biology, American University of Beirut, Beirut, Lebanon
| | - Ahmed F. El-Yazbi
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut, Lebanon
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Ali H. Eid
- Department of Basic Medical Sciences, College of Medicine, Qatar University Health, Qatar University, Doha, Qatar
- Biomedical and Pharmaceutical Research Unit, Qatar University Health, Qatar University, Doha, Qatar
| |
Collapse
|
34
|
Lee S, Park SK, Park H, Lee W, Kwon JH, Hong YC, Ha M, Kim Y, Lee B, Ha E. Prenatal heavy metal exposures and atopic dermatitis with gender difference in 6-month-old infants using multipollutant analysis. ENVIRONMENTAL RESEARCH 2021; 195:110865. [PMID: 33600821 DOI: 10.1016/j.envres.2021.110865] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 01/28/2021] [Accepted: 02/05/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND Prenatal exposure to heavy metals during critical developmental phases has been implicated in allergic phenotypes. However, few studies have been conducted on the gender-specific association of prenatal heavy metal exposure with atopic dermatitis (AD) in infants. OBJECTIVE To examine the gender-specific association of prenatal exposure to multiple heavy metals with AD incidence in 6-month-old infants using data from the Mothers and Children's Environmental Health (MOCEH). METHODS We evaluated 738 mother-child pairs from the MOCEH study, an ongoing prospective birth cohort. The concentrations of three heavy metals (lead, mercury and cadmium) in maternal blood samples were measured during early and late pregnancy. Each quartile of heavy metal concentration was used to consider the possible nonlinear association with AD. For assessing the multi-pollutant model, we constructed the multivariate regression model including all three heavy metals at both early and late pregnancy. Further, the group Lasso model was used to perform the variable selection with categorized exposures and assess the effect of multiple pollutants including their pairwise interactions. RESULTS A total of 200 incident cases of AD were diagnosed in 6-month-old infants. In the multivariate regression model of the boy group, adjusted odds ratios comparing the second, third and fourth quartile of lead exposure in boys with the first quartile were 1.83 (95% CI: 1.00, 3.38), 1.04 (0.91, 3.32) and 2.40 (1.18, 4.90), respectively. However, the only second quartile of lead exposure compared to first quartile was significantly associated with AD in girls. In addition, the results of the group Lasso model were similar with the results of multivariate regression model. CONCLUSION The results suggest that lead exposure in late pregnancy increases risk of AD in 6-month-old boys although the strength of association is weak. Further studies are needed to confirm the susceptibility window and gender differences in lead-induced AD.
Collapse
Affiliation(s)
- Seulbi Lee
- Occupational and Environmental Medicine, College of Medicine, Ewha Womans University, Seoul, Republic of Korea; Department of Epidemiology, School of Public Health, University of Michigan, Ann arbor, MI, United States
| | - Sung Kyun Park
- Department of Epidemiology, School of Public Health, University of Michigan, Ann arbor, MI, United States; Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, United States
| | - Hyesook Park
- Department of Preventive Medicine, College of Medicine, Ewha Womans University, Seoul, Republic of Korea; Graduate Program in System Health Science and Engineering, Ewha Womans University, Seoul, Republic of Korea
| | - Woojoo Lee
- Department of Public Health Sciences, Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
| | - Jung Hyun Kwon
- Department of Pediatrics, Korea University College of Medicine, Seoul, South Korea
| | - Yun-Chul Hong
- Department of Preventive Medicine, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Mina Ha
- Department of Preventive Medicine, College of Medicine, Dankook University, Cheonan, Republic of Korea
| | - Yangho Kim
- Department of Occupational and Environmental Medicine, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Republic of Korea
| | - Boeun Lee
- Environmental Health Research Division, National Institute of Environmental Research, Incheon, Republic of Korea
| | - Eunhee Ha
- Occupational and Environmental Medicine, College of Medicine, Ewha Womans University, Seoul, Republic of Korea; Graduate Program in System Health Science and Engineering, Ewha Womans University, Seoul, Republic of Korea; Ewha Medical Research Institute, College of Medicine, Ewha Womans University, Seoul, Republic of Korea.
| |
Collapse
|
35
|
Bello-Alvarez C, Camacho-Arroyo I. Impact of sex in the prevalence and progression of glioblastomas: the role of gonadal steroid hormones. Biol Sex Differ 2021; 12:28. [PMID: 33752729 PMCID: PMC7986260 DOI: 10.1186/s13293-021-00372-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 03/04/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND As in other types of cancers, sex is an essential factor in the origin and progression of glioblastomas. Research in the field of endocrinology and cancer suggests that gonadal steroid hormones play an important role in the progression and prevalence of glioblastomas. In the present review, we aim to discuss the actions and mechanism triggered by gonadal steroid hormones in glioblastomas. MAIN BODY Glioblastoma is the most common malignant primary brain tumor. According to the epidemiological data, glioblastomas are more frequent in men than in women in a 1.6/1 proportion both in children and adults. This evidence, and the knowledge about sex influence over the prevalence of countless diseases, suggest that male gonadal steroid hormones, such as testosterone, promote glioblastomas growth. In contrast, a protective role of female gonadal steroid hormones (estradiol and progesterone) against glioblastomas has been questioned. Several pieces of evidence demonstrate a variety of effects induced by female and male gonadal steroid hormones in glioblastomas. Several studies indicate that pregnancy, a physiological state with the highest progesterone and estradiol levels, accelerates the progression of low-grade astrocytomas to glioblastomas and increases the symptoms associated with these tumors. In vitro studies have demonstrated that progesterone has a dual role in glioblastoma cells: physiological concentrations promote cell proliferation, migration, and invasion while very high doses (out physiological range) reduce cell proliferation and increases cell death. CONCLUSION Gonadal steroid hormones can stimulate the progression of glioblastomas through the increase in proliferation, migration, and invasion. However, the effects mentioned above depend on the concentrations of these hormones and the receptor involved in hormone actions. Estradiol and progesterone can exert promoter or protective effects while the role of testosterone has been always associated to glioblastomas progression.
Collapse
Affiliation(s)
- Claudia Bello-Alvarez
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México (UNAM), 04510, Ciudad de México, México
| | - Ignacio Camacho-Arroyo
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México (UNAM), 04510, Ciudad de México, México.
| |
Collapse
|
36
|
Pain in Women: A Perspective Review on a Relevant Clinical Issue that Deserves Prioritization. Pain Ther 2021; 10:287-314. [PMID: 33723717 PMCID: PMC8119594 DOI: 10.1007/s40122-021-00244-1] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 02/08/2021] [Indexed: 02/07/2023] Open
Abstract
Introduction Gender equity and gender medicine are opportunities not to be missed, and this Expert Group Opinion Paper on pain in women aims to review the treatment of pain conditions mainly affecting women, as well as the fundamental aspects of the different clinical response to drug treatment between the genders, and what can be done for gender-specific rehabilitation. Methods Perspective review. Results Genotypic and phenotypic differences in pain between the sexes are conditioned by anatomical, physiological, neural, hormonal, psychological, social, and cultural factors, such as the response to pharmacological treatment to control pain. The examination of these factors shows that women are affected by pain diseases more frequently and severely than men and that they report pain more frequently and with a lower pain threshold than men. Some forms of pain are inherently related to gender differences, such as pain related to the genitourinary system. However, other forms of chronic pain are seen more frequently in women than men, such as migraine, rheumatological, and musculoskeletal pain, in particular fibromyalgia. Discussion Research is needed into the pathophysiological basis for gender differences in the generation of acute pain and maintenance of chronic pain, including the factors that put women at higher risk for developing chronic pain. In addition, different specialties need to collaborate to develop gender-related diagnostic and therapeutic guidelines, and healthcare professionals need to upskill themselves in the appropriate management of pain using existing diagnostic tools and therapeutic options.
Collapse
|
37
|
Sexual dimorphism in immunometabolism and autoimmunity: Impact on personalized medicine. Autoimmun Rev 2021; 20:102775. [PMID: 33609790 DOI: 10.1016/j.autrev.2021.102775] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 12/20/2020] [Indexed: 02/06/2023]
Abstract
Immune cells play essential roles in metabolic homeostasis and thus, undergo analogous changes in normal physiology (e.g., puberty and pregnancy) and in various metabolic and immune diseases. An essential component of this close relationship between the two is sex differences. Many autoimmune diseases, such as systemic lupus erythematous and multiple sclerosis, feature strikingly increased prevalence in females, whereas in contrast, infectious diseases, such as Ebola and Middle East Respiratory Syndrome, affect more men than women. Therefore, there are fundamental aspects of metabolic homeostasis and immune functions that are regulated differently in males and females. This can be observed in sex hormone-immune interaction where androgens, such as testosterone, have shown immunosuppressive effects whilst estrogen is on the opposite side of the spectrum with immunoenhancing facilitation of mechanisms. In addition, the two sexes exhibit significant differences in metabolic regulation, with estrous cycles in females known to induce variability in traits and more pronounced metabolic disease phenotype exhibited by males. It is likely that these differences underlie both the development of metabolic and autoimmune diseases and the response to current treatment options. Sexual dimorphism in immunometabolism has emerged to become an area of intense research, aiming to uncover sex-biased effector molecules in the various metabolic tissues and immune cell types, identify sex-biased cell-type-specific functions of common effector molecules, and understand whether the sex differences in metabolic and immune functions influence each other during autoimmune pathogenesis. In this review, we will summarize recent findings that address these critical questions of sexual dimorphism in immunometabolism as well as their translational implications for the clinical management of autoimmune diseases.
Collapse
|
38
|
Kumar RS, Goyal N. Estrogens as regulator of hematopoietic stem cell, immune cells and bone biology. Life Sci 2021; 269:119091. [PMID: 33476629 DOI: 10.1016/j.lfs.2021.119091] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 01/03/2021] [Accepted: 01/11/2021] [Indexed: 12/11/2022]
Abstract
Hematopoietic stem cells provide continuous supply of all the immune cells, through proliferation and differentiation decisions. These decisions are controlled by local bone marrow environment as well as by long-range signals for example endocrine system. Sex dependent differential immunological responses have been described under homeostasis and disease conditions. Females show higher longevity than male counterpart that seems to depend on major female sex hormone, estrogen. There are four estrogens - Estrone (E1), estradiol (E2), Estriol (E3) and Estetrol (E4) that spatially and temporarily present during different female reproductive phases. In this review, we discussed recent updates describing the effects of estrogen on HSC, immune cells and in bone biology. Estradiol (E2) being a major/abundant estrogen is extensively investigated, while effects of other estrogens E1, E3 and E4 are started to unravel recently. Furthermore, clinical effect of estrogen as hormone therapy is discussed in HSC and immune cells perspectives. The data presented in this review is compiled by searches of PubMed, database of American Cancer Society (ACS). We have included article from September 1994 to March 2020 as covering all article in chronological order is not fissile so we included relevant article with substantial information in this specific area of research by using the search term (alone or in combination) estrogen, hematopoietic stem cell, immune cells, gender difference, estrone, estriol, estetrol, therapeutic application, pregnancy, effect on bone.
Collapse
Affiliation(s)
- Rupali Sani Kumar
- CSIR-Central Drug Research Institute, Sector-10, Jankipuram Extension, Sitapur Road, Lucknow 226031, Uttar Pradesh, India.
| | - Neena Goyal
- CSIR-Central Drug Research Institute, Sector-10, Jankipuram Extension, Sitapur Road, Lucknow 226031, Uttar Pradesh, India
| |
Collapse
|
39
|
Islam MS, Chaudhuri I, Mobin MA, Islam M, Mahmud MS, KutubUddin M, Kabir KMA, Kamrujjaman M. The Perspective of Acquired Immunity to Combat against Infectious Diseases: An Overview. Health (London) 2021. [DOI: 10.4236/health.2021.139077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
40
|
Campesi I, Montella A, Sotgiu G, Dore S, Carru C, Zinellu A, Palermo M, Franconi F. Combined oral contraceptives modify the effect of smoking on inflammatory cellular indexes and endothelial function in healthy subjects. Eur J Pharmacol 2020; 891:173762. [PMID: 33253680 DOI: 10.1016/j.ejphar.2020.173762] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 11/04/2020] [Accepted: 11/06/2020] [Indexed: 12/12/2022]
Abstract
Little information is available on the influence of sex in combination with smoking habits and combined oral contraceptives (COC) use on cellular inflammatory indexes such as neutrophil/lymphocyte ratio (NLR), derived NRL (dNLR), platelet/lymphocyte ratio (PLR), monocyte/lymphocyte ratio (MLR), mean platelet volume/platelet count (MPV/PLT), aggregate inflammation systemic index (AISI), and systemic inflammation response index (SIRI), which are cost-effective biomarkers to assessing inflammation. Therefore, the effect of COC was studied alone or in association with smoking and compared with results from healthy COC-free women and men. Furthermore, the association of cellular inflammatory indexes with endothelial function (arginine (Arg), asymmetric dimethylarginine (ADMA), symmetric dimethylarginine (SDMA) and lipid peroxidation (malondialdehyde MDA) biomarkers was evaluated. Blood was collected for hematological and biochemical analysis, which were used to calculate PLR, NLR, dNLR, MLR, MPV/PLT, AISI, and SIRI. Serum samples were assayed for Arg, ADMA, SDMA, and MDA. Monocytes, MLR, SIRI, and MPV/PLT were higher in men, while PLT count was higher in women. COC use increased lymphocytes and lowered PLR and MLR. Smoking reduced sexually divergent parameters, especially in COC users: smoking and non-smoking COC-free women displayed six divergent parameters, while COC users displayed only two (monocytes and MPV). In addition, COC affected endothelial function, reducing ADMA and Arg. Moreover, COC-free women had lower Arg levels than men. In conclusion, COC use strongly influence the effects of tobacco smoking, which are sex and parameter specific. Further, these data stress that COC use and smoking attitude select different cohorts indicating that sex and gender studies need intersectionality.
Collapse
Affiliation(s)
- Ilaria Campesi
- Dipartimento di Scienze Biomediche, Università Degli Studi di Sassari, 07100, Sassari, Italy; Laboratorio Nazionale di Farmacologia e Medicina di Genere, Istituto Nazionale Biostrutture Biosistemi, 07100, Sassari, Italy.
| | - Andrea Montella
- Dipartimento di Scienze Biomediche, Università Degli Studi di Sassari, 07100, Sassari, Italy; Unità Operativa di Genetica e Biologia Dello Sviluppo, Azienda Ospedaliero Universitaria di Sassari, 07100, Sassari, Italy
| | - Giovanni Sotgiu
- Dipartimento di Scienze Mediche Chirurgiche e Sperimentali, Università Degli Studi di Sassari, 07100, Sassari, Italy
| | - Simone Dore
- Dipartimento di Scienze Mediche Chirurgiche e Sperimentali, Università Degli Studi di Sassari, 07100, Sassari, Italy
| | - Ciriaco Carru
- Dipartimento di Scienze Biomediche, Università Degli Studi di Sassari, 07100, Sassari, Italy
| | - Angelo Zinellu
- Dipartimento di Scienze Biomediche, Università Degli Studi di Sassari, 07100, Sassari, Italy
| | - Mario Palermo
- Unità Operativa di Endocrinologia, Azienda Ospedaliero Universitaria di Sassari, 07100, Sassari, Italy
| | - Flavia Franconi
- Laboratorio Nazionale di Farmacologia e Medicina di Genere, Istituto Nazionale Biostrutture Biosistemi, 07100, Sassari, Italy
| |
Collapse
|
41
|
Alhussien MN, Dang AK. Interaction between stress hormones and phagocytic cells and its effect on the health status of dairy cows: A review. Vet World 2020; 13:1837-1848. [PMID: 33132594 PMCID: PMC7566244 DOI: 10.14202/vetworld.2020.1837-1848] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 07/22/2020] [Indexed: 12/14/2022] Open
Abstract
Dairy cows are exposed to various stressors during their production cycle that makes them more susceptible to various diseases. Phagocytes (neutrophils and macrophages) are important soldiers of the innate immune system. Neutrophils are the first responders to an inflammatory response and stress and kill pathogens by generating reactive oxygen species and by the release of various antimicrobial peptides, enzymes, neutrophil extracellular trap formation, etc. Macrophages, the other phagocytes, are also the cleanup crew for the innate immune system that removes debris, pathogens, and dead neutrophils later on after an inflammatory response. The neuroendocrine system along with phagocytes exhibits an immunomodulatory potential during stressful conditions. Neuroendocrine system directly affects the activity of phagocytes by communicating bidirectionally through shared receptors and messenger molecules such as hormones, neurotransmitters, or cytokines. Different immune cells may show variable responses to each hormone. Short time exposure to stress can be beneficial, but repeated or extended exposure to stress may be detrimental to the overall health and well-being of an animal. Although some stresses associated with farming practices in dairy cows are unavoidable, better understanding of the interactions occurring between various stress hormones and phagocytic cells can help to reduce stress, improve productivity and animal welfare. This review highlights the role played by various stress hormones in modulating phagocytic cell performance of dairy cattle under inflammatory conditions.
Collapse
Affiliation(s)
- Mohanned Naif Alhussien
- Lactation and Immuno-Physiology Laboratory, ICAR-National Dairy Research Institute, Karnal, Haryana, India
| | - Ajay Kumar Dang
- Lactation and Immuno-Physiology Laboratory, ICAR-National Dairy Research Institute, Karnal, Haryana, India
| |
Collapse
|
42
|
Lasrado N, Jia T, Massilamany C, Franco R, Illes Z, Reddy J. Mechanisms of sex hormones in autoimmunity: focus on EAE. Biol Sex Differ 2020; 11:50. [PMID: 32894183 PMCID: PMC7475723 DOI: 10.1186/s13293-020-00325-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 08/11/2020] [Indexed: 12/17/2022] Open
Abstract
Sex-related differences in the occurrence of autoimmune diseases is well documented, with females showing a greater propensity to develop these diseases than their male counterparts. Sex hormones, namely dihydrotestosterone and estrogens, have been shown to ameliorate the severity of inflammatory diseases. Immunologically, the beneficial effects of sex hormones have been ascribed to the suppression of effector lymphocyte responses accompanied by immune deviation from pro-inflammatory to anti-inflammatory cytokine production. In this review, we present our view of the mechanisms of sex hormones that contribute to their ability to suppress autoimmune responses with an emphasis on the pathogenesis of experimental autoimmune encephalomyelitis.
Collapse
Affiliation(s)
- Ninaad Lasrado
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE, 68583, USA
| | - Ting Jia
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE, 68583, USA
| | | | - Rodrigo Franco
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE, 68583, USA
| | - Zsolt Illes
- Department of Neurology, Odense University Hospital, University of Southern Denmark, Odense, Denmark
| | - Jay Reddy
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE, 68583, USA.
| |
Collapse
|
43
|
Chanana N, Palmo T, Sharma K, Kumar R, Graham BB, Pasha Q. Sex-derived attributes contributing to SARS-CoV-2 mortality. Am J Physiol Endocrinol Metab 2020; 319:E562-E567. [PMID: 32726128 PMCID: PMC7473885 DOI: 10.1152/ajpendo.00295.2020] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Epidemiological data in COVID-19 mortality indicate that men are more prone to die of SARS-CoV-2 infection than women, but biological causes for this sexual dimorphism are unknown. We discuss the prospective behavioral and biological differences between the sexes that could be attributed to this sex-based differentiation. The female sex hormones and the immune stimulatory genes, including Toll-like receptors, interleukins, and micro-RNAs present on X-chromosome, may impart lesser infectivity and mortality of the SARS-CoV-2 in females over males. The sex hormone estrogen interacts with the renin-angiotensin-aldosterone system, one of the most critical pathways in COVID-19 infectivity, and modulates the vasomotor homeostasis. Testosterone on the contrary enhances the levels of the two most critical molecules, angiotensin-converting enzyme 2 (ACE2) and the transmembrane protease serine-type 2 (TMPRSS2), transcriptionally and posttranslationally, thereby increasing viral load and delaying viral clearance in men as compared with women. We propose that modulating sex hormones, either by increasing estrogen or antiandrogen, may be a therapeutic option to reduce mortality from SARS-CoV-2.
Collapse
Affiliation(s)
- Neha Chanana
- CSIR-Institute of Genomics and Integrative Biology, Delhi, India
| | - Tsering Palmo
- CSIR-Institute of Genomics and Integrative Biology, Delhi, India
| | - Kavita Sharma
- CSIR-Institute of Genomics and Integrative Biology, Delhi, India
| | - Rahul Kumar
- Department of Medicine, University of California San Francisco, San Francisco, California
| | - Brian B Graham
- Department of Medicine, University of California San Francisco, San Francisco, California
| | - Qadar Pasha
- CSIR-Institute of Genomics and Integrative Biology, Delhi, India
| |
Collapse
|
44
|
Ben-Batalla I, Vargas-Delgado ME, von Amsberg G, Janning M, Loges S. Influence of Androgens on Immunity to Self and Foreign: Effects on Immunity and Cancer. Front Immunol 2020; 11:1184. [PMID: 32714315 PMCID: PMC7346249 DOI: 10.3389/fimmu.2020.01184] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 05/13/2020] [Indexed: 12/17/2022] Open
Abstract
It is well-known that sex hormones can directly and indirectly influence immune cell function. Different studies support a suppressive role of androgens on different components of the immune system by decreasing antibody production, T cell proliferation, NK cytotoxicity, and stimulating the production of anti-inflammatory cytokines. Androgen receptors have also been detected in many different cells of hematopoietic origin leading to direct effects of their ligands on the development and function of the immune system. The immunosuppressive properties of androgens could contribute to gender dimorphisms in autoimmune and infectious disease and thereby also hamper immune surveillance of tumors. Consistently, females generally are more prone to autoimmunity, while relatively less susceptible to infections, and have lower incidence and mortality of the majority of cancers compared to males. Some studies show that androgen deprivation therapy (ADT) can induce expansion of naïve T cells and increase T-cell responses. Emerging clinical data also reveal that ADT might enhance the efficacy of various immunotherapies including immune checkpoint blockade. In this review, we will discuss the potential role of androgens and their receptors in the immune responses in the context of different diseases. A particular focus will be on cancer, highlighting the effect of androgens on immune surveillance, tumor biology and on the efficacy of anti-cancer therapies including emerging immune therapies.
Collapse
Affiliation(s)
- Isabel Ben-Batalla
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald Comprehensive Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - María Elena Vargas-Delgado
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald Comprehensive Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gunhild von Amsberg
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald Comprehensive Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Martini-Clinic, Prostate Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Melanie Janning
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald Comprehensive Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Division of Personalized Medical Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Personalized Oncology, University Hospital Mannheim, Mannheim, Germany
| | - Sonja Loges
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald Comprehensive Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Division of Personalized Medical Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Personalized Oncology, University Hospital Mannheim, Mannheim, Germany
| |
Collapse
|
45
|
Chatterjee S, Ghosh R, Biswas P, Dubey S, Guria RT, Sharma CB, Kalra S. COVID-19: the endocrine opportunity in a pandemic. MINERVA ENDOCRINOL 2020; 45:204-227. [PMID: 32548995 DOI: 10.23736/s0391-1977.20.03216-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The 2019 Coronavirus disease (COVID-19) pandemic has disrupted the social, economical and medical system worldwide. Although it is strictly an infectious disease, its intricate bidirectional relationship with various non-communicable metabolic diseases and endocrinological factors has been observed. While diabetes, hypertension, obesity have been found to be independent risk factors for COVID-19 disease severity and mortality, more inclination towards sedentary lifestyle, psychosocial stress at this critical time may be the harbingers of metabolic syndrome. Thus, endocrinologists have a great opportunity to play their role to combat this pandemic. This paper examines how various endocrinological disorders influence the dynamics of COVID-19 and vice versa. Moreover, it also intends to review the clinical guidelines to be adopted in practice of endocrinology in this trying time.
Collapse
Affiliation(s)
- Subhankar Chatterjee
- Department of General Medicine, Rajendra Institute of Medical Sciences, Ranchi, India -
| | - Ritwik Ghosh
- Department of General Medicine, Burdwan Medical College and Hospital, Burdwan, India
| | - Payel Biswas
- Department of Radiodiagnosis, Care and Cure Hospital, Barasat, India
| | - Souvik Dubey
- Department of Neuromedicine, Bangur Institute of Neurosciences, Institute of Post Graduate Medical Education and Research and SSKM Hospital, Kolkata, India
| | - Rishi T Guria
- Department of General Medicine, Rajendra Institute of Medical Sciences, Ranchi, India
| | - Chandra B Sharma
- Department of General Medicine, Rajendra Institute of Medical Sciences, Ranchi, India
| | - Sanjay Kalra
- Department of Endocrinology, Bharti Hospital, Karnal, India
| |
Collapse
|
46
|
Sex-Specific Effects of the Nlrp3 Inflammasome on Atherogenesis in LDL Receptor-Deficient Mice. JACC Basic Transl Sci 2020; 5:582-598. [PMID: 32613145 PMCID: PMC7315187 DOI: 10.1016/j.jacbts.2020.03.016] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/11/2020] [Accepted: 03/11/2020] [Indexed: 12/27/2022]
Abstract
In this study we observed sex-specific effects of the NLRP3 inflammasome on atherogenesis in LDLR-deficient mice, with NLRP3 inflammasome playing a more prominent role in atherosclerosis in female mice than in males. Sex hormones may be involved in NLRP3 inflammasome–mediated atherogenesis and may underlie differential responses to anti-NLRP3 therapy between males and females. Testosterone may play an inhibitory role by blocking NLRP3 inflammasome and inflammation in atherogenesis, whereas female sex hormones may promote NLRP3 inflammasome–mediated atherosclerosis. The results of the present study may help design future clinical trials, with the objective to personalize cardiovascular care for men and women.
In the Ldlr-/- mouse model of atherosclerosis, female Nlrp3-/- bone marrow chimera and Nlrp3-/- mice developed significantly smaller lesions in the aortic sinus and decreased lipid content in aorta en face, but a similar protection was not observed in males. Ovariectomized female mice lost protection from atherosclerosis in the setting of NLRP3 deficiency, whereas atherosclerosis showed a greater dependency on NLRP3 in castrated males. Thus, castration increased the dependency of atherosclerosis on the NLRP3 inflammasome, suggesting that testosterone may block inflammation in atherogenesis. Conversely, ovariectomy reduced the dependency on NLRP3 inflammasome components for atherogenesis, suggesting that estrogen may promote inflammasome-mediated atherosclerosis.
Collapse
|
47
|
Santinelli L, Ceccarelli G, Borrazzo C, Innocenti GP, Frasca F, Cavallari EN, Celani L, Nonne C, Mastroianni CM, d'Ettorre G. Sex-related differences in markers of immune activation in virologically suppressed HIV-infected patients. Biol Sex Differ 2020; 11:23. [PMID: 32357901 PMCID: PMC7195770 DOI: 10.1186/s13293-020-00302-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Accepted: 04/16/2020] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVES Gender-specific studies remain a neglected area of biomedical research. Recent reports have emphasized that sex-related biological factors may affect disease progression during HIV-1 infection. The aim of this study was to investigate the influence of sex on the levels of immune activation in the gut and in peripheral blood of individuals with HIV treated with fully suppressive antiretroviral therapy (ART). METHODS Thirty individuals with HIV undergoing long-term fully suppressive ART were enrolled in this study. Lamina propria lymphocytes (LPL) and peripheral blood mononuclear cells (PBMCs) were isolated from gut biopsies collected by pancolonoscopy and peripheral blood samples. The expression of markers of immune activation was evaluated by multi-parametric flow cytometry. This is a sub analysis of ClinicalTrials.gov Identifier: NCT02276326 RESULTS: We observed differences in the levels of immune activation in the gut and in PBMCs, with values higher in the gut compartment compared to PBMCs. In addition, we found that the mean value of the levels of immune activation was higher in the women than in the men. Finally, we measured the markers of immune activation by mean relative difference (MRD) and confirmed the higher value in the women. CONCLUSION A significant sex-related difference in the level of immune activation was observed in a population of individuals with HIV on long-term ART. A more complete characterization of these differences may support the introduction of sex-specific approaches in the clinical management of individuals with HIV.
Collapse
Affiliation(s)
- Letizia Santinelli
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Giancarlo Ceccarelli
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy.
| | - Cristian Borrazzo
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | | | - Federica Frasca
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | | | - Luigi Celani
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Chiara Nonne
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | | | - Gabriella d'Ettorre
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
48
|
Wu B, Sun X, Yuan B, Ge F, Gupta HB, Chiang HC, Li J, Hu Y, Curiel TJ, Li R. PPARγ inhibition boosts efficacy of PD-L1 Checkpoint Blockade Immunotherapy against Murine Melanoma in a sexually dimorphic manner. Int J Biol Sci 2020; 16:1526-1535. [PMID: 32226299 PMCID: PMC7097912 DOI: 10.7150/ijbs.42966] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 02/19/2020] [Indexed: 01/08/2023] Open
Abstract
Immune checkpoint blockade-based immunotherapy has become standard of care for multiple cancer types. However, the overall response rates among various cancer types still remain unsatisfactory. There is a pressing clinical need to identify combination therapies to improve efficacy of anticancer immunotherapy. We previously showed that pharmacologic inhibition of PPARγ by GW9662 boosts αPD-L1 and αPD-1 antibody efficacy in treating murine mammary tumors. In addition, we defined sexually dimorphic αPD-L1 efficacy in B16 melanoma. Here, we show a sexually dimorphic response to the combination of GW9662 and αPD-L1 immunotherapy in B16 melanoma. Combination effects were observed in female, but not male hosts. Neither female oöphorectomy impairs, nor does male castration rescue the combination effects, suggesting a sex hormone-independent response to this combination therapy. In diet-induced obese females, melanoma growth remained responsive to the combination treatment, albeit less robustly than lean females. These findings are informative for future design and application of immunotherapy-related combination therapy for treating human melanoma patients by taking gender and obesity status into consideration.
Collapse
Affiliation(s)
- Bogang Wu
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - Xiujie Sun
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - Bin Yuan
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - Fei Ge
- Department of Molecular Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Harshita B Gupta
- Department of Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Huai-Chin Chiang
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - Jingwei Li
- Department of Molecular Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Yanfen Hu
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - Tyler J Curiel
- Department of Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Rong Li
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| |
Collapse
|
49
|
Trevisan A, Giuliani A, Scapellato ML, Anticoli S, Carsetti R, Zaffina S, Brugaletta R, Vonesch N, Tomao P, Ruggieri A. Sex Disparity in Response to Hepatitis B Vaccine Related to the Age of Vaccination. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17010327. [PMID: 31906550 PMCID: PMC6981715 DOI: 10.3390/ijerph17010327] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 12/24/2019] [Accepted: 12/31/2019] [Indexed: 12/27/2022]
Abstract
Hepatitis B virus (HBV) infection is one of the major infectious hazards for health-care workers (HCWs) because of the frequency of percutaneous exposures to blood or body fluids. For this reason, all HCWs should be vaccinated, including students in medicine and health professional degree programs. The aim of this study was to assess the immune coverage to anti-HBV vaccine and long-lasting protective titres of anti-HBs antibodies in female and male students to evaluate gender-related differences in response to HBV vaccination. Data relative to anti-HBs antibody titre, sex, age, and age at vaccination were collected and analyzed from 5291 Italian students (1812 males and 3479 females) of the graduate courses at the School of Medicine, who underwent the mandatory health surveillance of workers exposed to biological risk. The results indicated that gender affects the immune response to HBV vaccine, particularly evident in the case of females vaccinated after one year of age who exhibited a statistically significant (p = 0.0023) 1.21-fold increase in median antibody titre with respect to males. Our findings could contribute to the optimization of HBV vaccination schedules in health surveillance of HCWs.
Collapse
Affiliation(s)
- Andrea Trevisan
- Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy;
- Correspondence: (A.T.); (A.R.)
| | - Alessandro Giuliani
- Environment and Health Department, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Maria Luisa Scapellato
- Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy;
| | - Simona Anticoli
- Istituto Superiore di Sanità, Center for Gender Specific Medicine, 00161 Rome, Italy;
| | - Rita Carsetti
- Diagnostic Immunology Unit, Department of Laboratories and B cell Pathophysiology Unit, Immunology Research Area, Bambino Gesù Children Hospital, 00165 Rome, Italy;
| | - Salvatore Zaffina
- Occupational Medicine, Health Directorate, Bambino Gesù Children’s Hospital, 00165 Rome, Italy; (S.Z.); (R.B.)
| | - Rita Brugaletta
- Occupational Medicine, Health Directorate, Bambino Gesù Children’s Hospital, 00165 Rome, Italy; (S.Z.); (R.B.)
| | - Nicoletta Vonesch
- Department of Occupational and Environmental Medicine, Epidemiology and Hygiene, Italian National Workers Compensation Authority, Monte Porzio Catone, 00078 Rome, Italy; (N.V.); (P.T.)
| | - Paola Tomao
- Department of Occupational and Environmental Medicine, Epidemiology and Hygiene, Italian National Workers Compensation Authority, Monte Porzio Catone, 00078 Rome, Italy; (N.V.); (P.T.)
| | - Anna Ruggieri
- Istituto Superiore di Sanità, Center for Gender Specific Medicine, 00161 Rome, Italy;
- Correspondence: (A.T.); (A.R.)
| |
Collapse
|
50
|
Talarowska ME, Szemraj J, Kuan-Pin S. Expression of ESR1 and ESR2 oestrogen receptor encoding gene and personality traits - preliminary study. PRZEGLAD MENOPAUZALNY = MENOPAUSE REVIEW 2019; 18:133-140. [PMID: 31975979 PMCID: PMC6970415 DOI: 10.5114/pm.2019.90804] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 11/03/2019] [Indexed: 12/24/2022]
Abstract
INTRODUCTION The main objective of the study is to examine the hypothesis claiming a correlation between personality traits measured with the use of the Minnesota Multiphasic Personality Inventory (MMPI-2) personality questionnaire and the expression of the ERα (ESR1) and ERβ (ESR2) encoding gene in patients suffering from depression. MATERIAL AND METHODS The experiment was carried out on a group of 44 individuals with depression. The Polish variant of the MMPI-2 was applied with the aim of assessing personality traits. Furthermore, the authors evaluated the expression of the genes encoding the oestrogen receptors (ERα and ERβ) at the mRNA level and protein level in the studied population. RESULTS No significant differences in the expression of ERα and ERβ encoding genes were found and confirmed in the patients with the first episode of depression and those suffering from subsequent episodes of the disease. No differences were found between women and men, either. In women a positive relationship was found between the scale of psychopathy (p = 0.04), paranoia (p = 0.01), and mania (p = 0.03) and expression for the ERα encoding gene at the mRNA level. A negative relationship was found between the mania scale and ERβ encoding gene expression at mRNA (p = 0.03) and protein (p = 0.04) levels. In males a positive relationship between anxiety as a personality trait and expression of the ERβ receptor encoding gene at mRNA level (p = 0.03) and protein level (p = 0.03) was found. CONCLUSIONS Personality traits may be linked with the expression of genes encoding oestrogen receptors (ERα and ERβ) among patients with depressive disorders.
Collapse
Affiliation(s)
- Monika E. Talarowska
- Department of Personality and Individual Differences, Institute of Psychology, University of Lodz, Poland
| | - Janusz Szemraj
- Department of Medical Biochemistry, Medical University of Lodz, Poland
| | - Su Kuan-Pin
- Department of General Psychiatry, China Medical University Hospital, Taichung City, Taiwan
| |
Collapse
|