1
|
Walker JK, Dillard CC, Gonzalez DE, Waldman HS, McAllister MJ. Impact of the menstrual cycle phases and time of day on markers of stress: salivary α-amylase and secretory immunoglobulin A. Stress 2025; 28:2449098. [PMID: 39757714 DOI: 10.1080/10253890.2024.2449098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 12/13/2024] [Indexed: 01/07/2025] Open
Abstract
Previous reports suggest that the menstrual cycle (MC) phases can impact cortisol concentrations. However, research is needed on whether the MC impacts other markers of stress and immune function. It has also been shown that some biomarkers are impacted by time of day, although differences between morning (AM) and afternoon (PM) biomarkers have not been studied over the course of the MC. This study assessed the effect of MC phases and time of day on salivary stress biomarkers [salivary α-amylase (sAA), secretory immunoglobulin A (SIgA)], progesterone, resting blood pressure and resting heart rate (RHR). A single-group repeated measure design was employed in which seventeen participants (n = 17) monitored their MC for two months while attending eight experimental sessions which included both AM and PM sessions during each predicted 1) menses, 2) follicular, 3) ovulatory and 4) luteal phases. Resting blood pressures, heart rates, body composition parameters (assessed via bioelectrical impedance analysis), sAA and SIgA concentrations were assessed. No time of day x MC phase interactions (p > 0.05) were noted for sAA or SIgA, resting blood pressure, heart rate, or body composition parameters. However, sAA and RHR were significantly higher in the PM, while SIgA was significantly higher in the AM. These data suggest that the MC phases do not impact sAA or SIgA, resting blood pressure, heart rates, or body composition parameters. However, time-of-day impacts RHR and concentrations of sAA and SIgA. These findings provide implications for female participants in research dealing with these biomarkers.
Collapse
Affiliation(s)
- Josey K Walker
- Metabolic & Applied Physiology Laboratory, Department of Health & Human Performance, Texas State University, San Marcos, TX, USA
| | - Courtney C Dillard
- Human Performance Research Laboratory, Department of Kinesiology, University of North Alabama, Florence, AL, USA
| | - Drew E Gonzalez
- Department of Kinesiology and Sport Management, Texas A&M University, TX, USA
| | - Hunter S Waldman
- Human Performance Research Laboratory, Department of Kinesiology, University of North Alabama, Florence, AL, USA
| | - Matthew J McAllister
- Metabolic & Applied Physiology Laboratory, Department of Health & Human Performance, Texas State University, San Marcos, TX, USA
- ALERRT Center, Texas State University, San Marcos, TX, USA
| |
Collapse
|
2
|
Yang W, Liu C, Li Z, Cui M. Exploring new drug treatment targets for immune related bone diseases using a multi omics joint analysis strategy. Sci Rep 2025; 15:10618. [PMID: 40148470 PMCID: PMC11950375 DOI: 10.1038/s41598-025-94053-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 03/11/2025] [Indexed: 03/29/2025] Open
Abstract
In the field of treatment and prevention of immune-related bone diseases, significant challenges persist, necessitating the urgent exploration of new and effective treatment methods. However, most existing Mendelian randomization (MR) studies are confined to a single analytical approach, which limits the comprehensive understanding of the pathogenesis and potential therapeutic targets of these diseases. In light of this, we propose the hypothesis that genetic variations in specific plasma proteins have a causal relationship with immune-related bone diseases through the MR mechanism, and that key therapeutic targets can be accurately identified using an integrated multi-omic analysis approach. This study comprehensively applied a variety of analytical methods. Firstly, the protein quantitative trait locus (pQTLs) data from two large plasma protein databases and the Genome-Wide Association Study (GWAS) data of nine immune-related bone diseases were used for Mendelian randomization (MR) analysis. At the same time, we employed the Summary-based Mendelian Randomization (SMR) method, combined with the Bayesian colocalization analysis method of coding genes, as well as the Linkage Disequilibrium Score Regression (LDSC) analysis method based on genetic correlation analysis, as methods to verify the genetic association between genes and complex diseases, thus comprehensively obtaining positive results. In addition, a Phenome-wide Association Study (PheWAS) was conducted on significantly positive genes, and their expression patterns in different tissues were also explored. Subsequently, we integrated Protein-Protein Interaction (PPI) network analysis, Gene Ontology (GO) analysis. Finally, based on the above analytical methods, drug prediction and molecular docking studies were carried out with the aim of accurately identifying key therapeutic targets. Through a comprehensive analysis using four methods, namely the Mendelian randomization (MR) analysis study, Summary-based Mendelian Randomization (SMR) analysis study, Bayesian colocalization analysis study, and Linkage Disequilibrium Score Regression (LDSC) analysis study. We found that through MR, SMR, and combined with Bayesian colocalization analysis, an association was found between rheumatoid arthritis (RA) and HDGF. Using the combination of MR and Bayesian colocalization analysis, as well as LDSC analysis, it was concluded that RA was related to CCL19 and TNFRSF14. Based on the methods of MR and Bayesian colocalization, an association was found between GPT and Crohn's disease-related arthritis, and associations were found between BTN1A1, EVI5, OGA, TNFRSF14 and multiple sclerosis (MS), and associations were found between ICAM5, CCDC50, IL17RD, UBLCP1 and psoriatic arthritis (PsA). Specifically, in the MR analysis of RA, HDGF (P_ivw = 0.0338, OR = 1.0373, 95%CI = 1.0028-1.0730), CCL19 (P_ivw = 0.0004, OR = 0.3885, 95%CI = 0.2299-0.6566), TNFRSF14 (P_ivw = 0.0007, OR = 0.6947, 95%CI = 0.5634-0.8566); in the MR analysis of MS, BTN1A1 (P_ivw = 0.0000, OR = 0.6101, 95%CI = 0.4813-0.7733), EVI5 (P_ivw = 0.0000, OR = 0.3032, 95%CI = 0.1981-0.4642), OGA (P_ivw = 0.0005, OR = 0.4599, 95%CI = 0.2966-0.7131), TNFRSF14 (P_ivw = 0.0002, OR = 0.4026, 95%CI = 0.2505-0.6471); in the MR analysis of PsA, ICAM5 (P_ivw = 0.0281, OR = 1.1742, 95%CI = 1.0174-1.3552), CCDC50 (P_ivw = 0.0092, OR = 0.7359, 95%CI = 0.5843-0.9269), IL17RD (P_ivw = 0.0006, OR = 0.7887, 95%CI = 0.6886-0.9034), UBLCP1 (P_ivw = 0.0021, OR = 0.6901, 95%CI = 0.5448-0.8741); in the MR analysis of Crohn's disease-related arthritis, GPT (P_ivw = 0.0006, OR = 0.0057, 95%CI = 0.0003-0.1111). In the Bayesian colocalization analysis of RA, HDGF (H4 = 0.8426), CCL19 (H4 = 0.9762), TNFRSF14 (H4 = 0.8016); in the Bayesian colocalization analysis of MS, BTN1A1 (H4 = 0.7660), EVI5 (H4 = 0.9800), OGA (H4 = 0.8569), TNFRSF14 (H4 = 0.8904); in the Bayesian colocalization analysis of PsA, ICAM5 (H4 = 0.9476), CCDC50 (H4 = 0.9091), IL17RD (H4 = 0.9301), UBLCP1 (H4 = 0.8862); in the Bayesian colocalization analysis of Crohn's disease-related arthritis, GPT (H4 = 0.8126). In the SMR analysis of RA, HDGF (p_SMR = 0.0338, p_HEIDI = 0.0628). In the LDSC analysis of RA, CCL19 (P = 0.0000), TNFRSF14 (P = 0.0258). By comprehensively analyzing plasma proteomic and transcriptomic data, we successfully identified key therapeutic targets for various clinical subtypes of immune-associated bone diseases. Our findings indicate that the significant positive genes associated with RA include HDGF, CCL19, and TNFRSF14; the positive gene linked to Crohn-related arthropathy is GPT; for MS, the positive genes are BTN1A1, EVI5, OGA, and TNFRSF14; and for PsA, the positive genes are ICAM5, CCDC50, IL17RD, and UBLCP1. Through this comprehensive analytical approach, we have screened potential therapeutic targets for different clinical subtypes of immune-related bone diseases. This research not only enhances our understanding of the pathogenesis of these conditions but also provides a solid theoretical foundation for subsequent drug development and clinical treatment, with the potential to yield significant advancements in the management of patients with immune-related bone diseases.
Collapse
Affiliation(s)
- Wei Yang
- School of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, China
| | - Chenglin Liu
- School of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, China
| | - Zhenhua Li
- Affiliated Hospital of Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun, 130117, Jilin, China.
| | - Miao Cui
- Capital Medical University, No.10, Xitoutiao, You'anmenwai, Beijing, 100069, Fengtai District, China.
| |
Collapse
|
3
|
Muñoz-Guzmán MA, Alba-Hurtado F. Role of sex hormones in the reactivation of Toxocara canis larvae in pregnant bitches. Vet Parasitol 2025; 334:110393. [PMID: 39818124 DOI: 10.1016/j.vetpar.2025.110393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 01/08/2025] [Accepted: 01/09/2025] [Indexed: 01/18/2025]
Abstract
Based on an exhaustive review, a theory was proposed with an immunoendocrine perspective on the reactivation of dormant Toxocara canis larvae in pregnant bitches and their transmission to fetuses through the placenta, or milk and colostrum to puppies. A historical review was carried out on the reactivation of dormant larvae of T. canis in pregnant bitches which suggested that larval reactivation of T. canis is not solely the effect of a single hormone (prolactin) but is the result of a series of events triggered by progesterone, prolactin, and estrogens. In the first third of gestation, progesterone is capable of directly stimulating the reactivation of larvae through hormonal receptors, indirectly downregulating the granulomatous proinflammatory response around dormant T. canis larvae, and directing the response to a Th2 profile with increased levels of antibodies and blood eosinophils. After a time, when progesterone levels decrease, prolactin and estrogen maintain larval stimulation through hormonal receptors and downregulation of Th1 and the granulomatous proinflammatory response. Collectively, these hormones play major roles in the reactivation of T. canis larvae in pregnant bitches. The series of complex events that occur during larval reactivation is a clear example of transregulation, in which host hormones regulate the vital functions of the parasite to positively influence its establishment and/or proliferation. Understanding larval reactivation from an immunoendocrine perspective helps us to comprehensively understand the complex parasite-host relationship of T. canis.
Collapse
Affiliation(s)
- Marco Antonio Muñoz-Guzmán
- Departamento de Ciencias Biológicas, Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Km. 2.5 Carretera Cuautitlán-Teoloyucan S/N, Cuautitlán Izcalli, MEX 54714, Mexico
| | - Fernando Alba-Hurtado
- Departamento de Ciencias Biológicas, Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Km. 2.5 Carretera Cuautitlán-Teoloyucan S/N, Cuautitlán Izcalli, MEX 54714, Mexico.
| |
Collapse
|
4
|
Chiu RG, Eldeirawi K, Dick AI, Nyenhuis SM, Vajaranant TS, Caskey R, Lee VS. Association of Menopause and Rhinitis Among Adult Women in the United States: Findings from the All of Us Research Program. Laryngoscope 2025. [PMID: 39853748 DOI: 10.1002/lary.32015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 12/26/2024] [Accepted: 01/07/2025] [Indexed: 01/26/2025]
Abstract
OBJECTIVE The inflammatory role of female hormones has been garnering increased attention in the literature. Studies suggest a link between estrogen and inflammatory conditions of the airways and nasal mucosa. However, there remains a paucity of literature regarding the associations of hormones with rhinitis. Given the profound hormonal changes that occur during menopause, we sought to better understand the association between menopause and rhinitis. METHODS Data from the All of Us Research Program regarding rhinitis diagnoses, menopause status, demographic variables, socioeconomic status, and comorbidities were extracted for female participants aged 40-60. Crude odds ratios (cORs) and 95% confidence intervals (CIs) were calculated for unadjusted associations between menopause and rhinitis. Variables were then included in multivariable logistic regression models, with separate models for allergic rhinitis (AR) and nonallergic rhinitis (NAR) as the outcome variables. Adjusted odds ratios (aOR) and 95% CI were calculated. RESULTS We identified 40,875 female participants aged 40-60 without any missing data. Compared with participants without rhinitis, a greater proportion of those with AR (51.6% vs. 55.6%; cOR: 1.18; 95% CI: 1.11-1.25) and NAR (51.6% vs. 58.9%; cOR: 1.34; 95% CI: 1.11-1.63) had experienced menopause. However, after controlling for covariates, menopause was associated with a decreased odds of AR (aOR: 0.89; 95% CI: 0.82-0.96) and not associated with NAR (aOR: 0.98; 95% CI: 0.77-1.24). CONCLUSION Menopause was independently associated with a decreased odds of AR but was not associated with NAR. Research should aim to further examine these relationships and hormonal mechanisms underlying the observed protective associations. LEVEL OF EVIDENCE 3 Laryngoscope, 2025.
Collapse
Affiliation(s)
- Richard G Chiu
- Department of Otolaryngology, College of Medicine, University of Illinois Chicago, Chicago, Illinois, U.S.A
| | - Kamal Eldeirawi
- Department of Population Health Nursing Science, College of Nursing, University of Illinois Chicago, Chicago, Illinois, U.S.A
| | - Anthony I Dick
- Department of Otolaryngology, College of Medicine, University of Illinois Chicago, Chicago, Illinois, U.S.A
| | - Sharmilee M Nyenhuis
- Department of Pediatrics, Section of Allergy and Immunology, University of Chicago, Chicago, Illinois, U.S.A
| | - Thasarat Sutabutr Vajaranant
- Department of Ophthalmology and Visual Sciences, College of Medicine, University of Illinois Chicago, Chicago, Illinois, U.S.A
| | - Rachel Caskey
- Department of Medicine, College of Medicine, University of Illinois Chicago, Chicago, Illinois, U.S.A
| | - Victoria S Lee
- Department of Otolaryngology, College of Medicine, University of Illinois Chicago, Chicago, Illinois, U.S.A
| |
Collapse
|
5
|
Marron K, Harrity C. Comparing surface immune markers in successful and non-viable ART pregnancies on the day of hCG measurement: a prospective pilot study. REPRODUCTION AND FERTILITY 2025; 6:e240034. [PMID: 39679612 PMCID: PMC11968024 DOI: 10.1530/raf-24-0034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 12/02/2024] [Accepted: 12/13/2024] [Indexed: 12/17/2024] Open
Abstract
Abstract Blood lymphocyte reference ranges in non-pregnant females are established, but changes in pregnancy are less well understood. The early identification of immunological markers that could suggest an increased risk of early pregnancy loss may allow for timely intervention to improve outcomes. A lymphocytic immunophenotype provides a broad assessment of important immune parameters and potential indicators, which may be of relevance to pregnancy outcome. Comparison of immunophenotype results on the day of a positive hCG after embryo transfer between successful and failed pregnancies allows for this assessment. Baseline non-pregnant lymphocyte percentage and cell/µL profiles were established with a comprehensive panel on 93 age-matched male factor controls. Sixty-five in-vitro fertilisation (IVF) patients had an immunophenotype assessment on the day of a positive hCG, followed by further hCG tests and ultrasound monitoring as required to ultimately evaluate success (live birth) or failure (miscarriage). Thirty-one pregnancies were viable, leading to a live birth, while 34 ended in miscarriage. Total CD56, pNK, NKT, CD4 and CD8 levels were equivalent between all groups. Regardless of the outcome, B lymphocytes increased in pregnancy compared to controls. Of interest, in the later miscarriage cohort, pNK-specific CD69 was reduced (1.6 vs 5.4%, P = 0.02), while CD57+ cells were increased (45.4 vs 38.9%, P = 0.025). Corresponding changes were observed in cell/µL concentrations. Low level CD69 activation and elevated CD56dim and CD57+ NK cells were identified as markers that could potentially identify a pregnancy at risk of miscarriage, with further study needed to explore whether these changes represent cause or effect. Lay summary Unexplained infertility remains a difficult issue for patients and physicians alike, but despite recent diagnostic strides and innovative methods, there are no clear solutions on the horizon. Pregnancies can still occur in these challenging populations, either spontaneously or by interventions such as IVF. The early identification of various immune markers by blood sampling that may correlate with the subsequent outcome could be beneficial in identifying pregnancies at increased risk of miscarriage and perhaps allowing for timely and effective interventions.
Collapse
Affiliation(s)
- Kevin Marron
- Enfer Medical, M7 Business Park, Naas, Kildare, Ireland
| | - Conor Harrity
- RCSI University of Medicine and Health Sciences, Dublin, Ireland
- Beaumont Hospital, Dublin, Ireland
| |
Collapse
|
6
|
Nesbitt C, Van Der Walt A, Butzkueven H, Cheung AS, Jokubaitis VG. Exploring the role of sex hormones and gender diversity in multiple sclerosis. Nat Rev Neurol 2025; 21:48-62. [PMID: 39658653 DOI: 10.1038/s41582-024-01042-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/30/2024] [Indexed: 12/12/2024]
Abstract
Sex and sex hormones are thought to influence multiple sclerosis (MS) through effects on inflammation, myelination and neurodegeneration, and exogenous hormones have been explored for their therapeutic potential. However, our understanding of how sex hormones influence MS disease processes and outcomes remains incomplete. Furthermore, our current knowledge is derived primarily from studies that focus exclusively on cisgender populations with exclusion of gender-diverse people. Gender-affirming hormone therapy comprising exogenous sex hormones or sex hormone blocking agents are commonly used by transgender and gender-diverse individuals, and it could influence MS risk and outcomes at various stages of disease. A better understanding of the impact and potential therapeutic effects of both endogenous and exogenous sex hormones in MS is needed to improve care and outcomes for cisgender individuals and, moreover, for gender-diverse populations wherein an evidence base does not exist. In this Perspective, we discuss the effects of endogenous and exogenous sex hormones in MS, including their potential therapeutic benefits, and examine both established sex-based dimorphisms and the potential for gender-diverse dimorphisms. We advocate for future research that includes gender-diverse people to enhance our knowledge of the interplay of sex and sex hormones in MS, leading to the development of more effective and inclusive treatment strategies and improvement of care for all individuals with MS.
Collapse
Affiliation(s)
- Cassie Nesbitt
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia.
- Multiple Sclerosis and Neuroimmunology Clinic, Alfred Health, Melbourne, Victoria, Australia.
- Department of Neurology, Alfred Health, Melbourne, Victoria, Australia.
| | - Anneke Van Der Walt
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
- Multiple Sclerosis and Neuroimmunology Clinic, Alfred Health, Melbourne, Victoria, Australia
- Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| | - Helmut Butzkueven
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
- Multiple Sclerosis and Neuroimmunology Clinic, Alfred Health, Melbourne, Victoria, Australia
- Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| | - Ada S Cheung
- Trans Health Research Group, Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia
- Department of Endocrinology, Austin Health, Heidelberg, Victoria, Australia
| | - Vilija G Jokubaitis
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia.
- Multiple Sclerosis and Neuroimmunology Clinic, Alfred Health, Melbourne, Victoria, Australia.
- Department of Neurology, Alfred Health, Melbourne, Victoria, Australia.
| |
Collapse
|
7
|
Alemán OR, Quintero JC, Camacho-Arroyo I. The language of glioblastoma: A tale of cytokines and sex hormones communication. Neurooncol Adv 2025; 7:vdaf017. [PMID: 40351835 PMCID: PMC12063100 DOI: 10.1093/noajnl/vdaf017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2025] Open
Abstract
Glioblastoma (GB) is the most aggressive and frequent tumor in the central nervous system and, in humans, represents the worst prognosis for cancer. GB develops a very complex microenvironment, recruiting and interacting with a variety of cells and soluble factors, including immune cells, cytokines, and sex hormones, that contribute to GB survival and progression. Recent evidence has shown a crosstalk between cytokine and sex hormone signaling in GB. This communication could provide GB resistance to treatments and malignancy. Then, how GB orchestrates this communication is a matter of interest. For instance, a critical interaction between tumor necrosis factor-beta (TGF-β) and estrogen receptor signaling has been reported in regulating epithelial-mesenchymal transition, an essential step in GB progression. Furthermore, an inhibition of TGF-β signaling by androgen receptor has been reported to promote GB tumorigenesis in men. Conversely, it has been described that cytokines regulate steroid hormone production in different organs, and this mechanism could be involved in GB development and progression. All these data suggest an intercommunication between the immune and endocrine systems in the tumor microenvironment. Thus, in this review, we focus on explaining the knowledge about this critical intercommunication system and its implication in GB progression.
Collapse
Affiliation(s)
- Omar Rafael Alemán
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 11000, México
| | - Juan Carlos Quintero
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 11000, México
| | - Ignacio Camacho-Arroyo
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 11000, México
| |
Collapse
|
8
|
Bowen MB, Melendez B, Zhang Q, Yang RK, Fellman BM, Lawson BC, Adjei NN, Celestino J, Wani KM, Singh B, Urbauer DL, Lazar AJ, Lu KH, Wargo JA, Westin SN, Yates MS. Long-Term Follow-up of Levonorgestrel Intrauterine Device for Atypical Hyperplasia and Early Endometrial Cancer Reveals Relapse Characterized by Immune Exhaustion. Clin Cancer Res 2024; 30:5073-5082. [PMID: 38922360 PMCID: PMC11567806 DOI: 10.1158/1078-0432.ccr-24-0362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/26/2024] [Accepted: 06/20/2024] [Indexed: 06/27/2024]
Abstract
PURPOSE Nonsurgical treatment options are increasingly needed for endometrial atypical hyperplasia (AH) and endometrioid endometrial cancer (EEC). Despite promising initial response rates, prospective long-term data and determinants for relapse are limited. MATERIALS AND METHODS Follow-up data from patients in our prospective phase II trial of levonorgestrel intrauterine device (LIUD) for AH/G1EEC were collected from medical records. Spatial transcriptomics (Nanostring GeoMX digital spatial profiling) with in silico cell type deconvolution and pathway analyses were employed on longitudinal biopsy samples from five patients across pre-treatment, on-treatment, and relapse. RESULTS Of 43 participants exhibiting initial response to LIUD, 41 had follow-up data. Sixteen (39%) experienced relapse. Clinical factors associated with shorter response duration included younger age, initial diagnosis of G1EEC, lack of response at 6 months, premenopausal status, and Hispanic ethnicity (P < 0.05), but only 6-month response status remained a significant predictor in a multivariate model (P = 0.023). LIUD increased abundance of NK cells (ΔMCP-counter score = 46.13, FDR = 0.004) and cytotoxic lymphocytes (ΔMCP-counter score = 277.67, FDR = 0.004), as well as lymphocyte cytotoxicity markers PRF1 (log2FC = 1.62, FDR = 0.025) and GZMA (log2FC = 2.47, FDR = 0.008). NK cells were reduced at relapse (ΔMCP-counter score = -55.96, FDR = 0.02). Immune-related pathways (IFNα response and TGFβ signaling) were enriched at relapse (FDR < 0.05). IDO1 expression, reflecting immune exhaustion, was upregulated at relapse (FDR < 0.05). CONCLUSIONS Upfront resistance and relapse after initial response to LIUD for AH/G1EEC impacts nearly half of patients, remaining a major hurdle for nonsurgical treatment of AH/G1EEC. Molecular studies evaluating longitudinal biopsies from a small cohort implicate immune mechanisms at relapse, including reversal of progestin-related immunomodulation and increased immune exhaustion. See related commentary by Johannet and Friedman, p. 5001.
Collapse
Affiliation(s)
- Mikayla Borthwick Bowen
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Brenda Melendez
- The Platform for Innovative Microbiome & Translational Research (PRIME-TR), Moon Shots Program, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Qian Zhang
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Richard K. Yang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Bryan M. Fellman
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Barrett C. Lawson
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Naomi N. Adjei
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Joseph Celestino
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Khalida M. Wani
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Bhavana Singh
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Diana L. Urbauer
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Alexander J. Lazar
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Karen H. Lu
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Jennifer A. Wargo
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Shannon N. Westin
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Melinda S. Yates
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| |
Collapse
|
9
|
Wang L, Xu S, Chen R, Ding Y, Liu M, Hou C, Wu Z, Men X, Bao M, He B, Li S. Exploring the causal association between epigenetic clocks and menopause age: insights from a bidirectional Mendelian randomization study. Front Endocrinol (Lausanne) 2024; 15:1429514. [PMID: 39247918 PMCID: PMC11377254 DOI: 10.3389/fendo.2024.1429514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 08/02/2024] [Indexed: 09/10/2024] Open
Abstract
Background Evidence suggests a connection between DNA methylation (DNAm) aging and reproductive aging. However, the causal relationship between DNAm and age at menopause remains uncertain. Methods Employing established DNAm epigenetic clocks, such as DNAm Hannum age acceleration (Hannum), Intrinsic epigenetic age acceleration (IEAA), DNAm-estimated granulocyte proportions (Gran), DNAm GrimAge acceleration (GrimAgeAccel), DNAm PhenoAge acceleration (PhenoAgeAccel), and DNAm-estimated plasminogen activator inhibitor-1 levels (DNAmPAIadjAge), a bidirectional Mendelian randomization (MR) study was carried out to explore the potential causality between DNAm and menopausal age. The primary analytical method used was the inverse variance weighted (IVW) estimation model, supplemented by various other estimation techniques. Results DNAm aging acceleration or deceleration, as indicated by Hannum, IEAA, Gran, GrimAgeAccel, PhenoAgeAccel, and DNAmPAIadjAge, did not exhibit a statistically significant causal effect on menopausal age according to forward MR analysis. However, there was a suggestive positive causal association between age at menopause and Gran (Beta = 0.0010; 95% confidence interval (CI): 0.0004, 0.0020) in reverse MR analysis. Conclusion The observed increase in granulocyte DNAm levels in relation to menopausal age could potentially serve as a valuable indicator for evaluating the physiological status at the onset of menopause.
Collapse
Affiliation(s)
- Ling Wang
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, School of Pharmaceutical Science, Changsha Medical University, Changsha, China
| | - Shuling Xu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Rumeng Chen
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Yining Ding
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Menghua Liu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Chunyan Hou
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Zhu Wu
- The Hunan Provincial Key Laboratory of the TCM Agricultural Biogenomics, Changsha Medical University, Changsha, China
| | - Xiaoju Men
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, School of Pharmaceutical Science, Changsha Medical University, Changsha, China
| | - Meihua Bao
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, School of Pharmaceutical Science, Changsha Medical University, Changsha, China
- The Hunan Provincial Key Laboratory of the TCM Agricultural Biogenomics, Changsha Medical University, Changsha, China
| | - Binsheng He
- The Hunan Provincial Key Laboratory of the TCM Agricultural Biogenomics, Changsha Medical University, Changsha, China
| | - Sen Li
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
10
|
Maham S, Yoon MS. Clinical Spectrum of Long COVID: Effects on Female Reproductive Health. Viruses 2024; 16:1142. [PMID: 39066303 PMCID: PMC11281454 DOI: 10.3390/v16071142] [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: 06/05/2024] [Revised: 07/15/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
The COVID-19 pandemic caused by SARS-CoV-2 has presented numerous health challenges, including long-term COVID, which affects female reproductive health. This review consolidates the current research on the impact of SARS-CoV-2 on the menstrual cycle, ovarian function, fertility, and overall gynecological health. This study emphasizes the role of angiotensin-converting enzyme receptors in viral entry and the subsequent tissue-specific pathological effects. It also explores the potential influence of long COVID on hormonal balance and immune responses, contributing to menstrual irregularities and impaired ovarian function. The findings indicate a higher prevalence of long-term COVID-19 among women, highlighting the substantial implications for reproductive health and the need for sex-sensitive longitudinal studies. Enhanced surveillance and targeted research are essential to develop effective interventions that prioritize women's reproductive well-being following SARS-CoV-2 infection. This review advocates for a sex-informed approach to ongoing COVID-19 research and healthcare strategies, aiming to provide up-to-date and pertinent data for healthcare providers and the general public, ultimately improving outcomes for females affected by long COVID.
Collapse
Affiliation(s)
- Syeda Maham
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health Sciences and Technology, Gachon University, Incheon 21999, Republic of Korea;
| | - Mee-Sup Yoon
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health Sciences and Technology, Gachon University, Incheon 21999, Republic of Korea;
- Department of Molecular Medicine, College of Medicine, Gachon University, Incheon 21999, Republic of Korea
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
| |
Collapse
|
11
|
Trivedi S, Deering-Rice CE, Aamodt SE, Huecksteadt TP, Myers EJ, Sanders KA, Paine R, Warren KJ. Progesterone amplifies allergic inflammation and airway pathology in association with higher lung ILC2 responses. Am J Physiol Lung Cell Mol Physiol 2024; 327:L65-L78. [PMID: 38651968 PMCID: PMC11380947 DOI: 10.1152/ajplung.00207.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 03/02/2024] [Accepted: 04/22/2024] [Indexed: 04/25/2024] Open
Abstract
Perimenstrual worsening of asthma occurs in up to 40% of women with asthma, leading to increased acute exacerbations requiring clinical care. The role of sex hormones during these times remains unclear. In the current study, we used a translational approach to determine whether progesterone exacerbates allergic inflammation in the traditional chicken egg ovalbumin (OVA) model in BALB/c mice. Simultaneously, we used peripheral blood mononuclear cells (PBMC) from healthy human donors to assess the effects of progesterone on circulating group 2 innate lymphoid cells (ILC2). Briefly, lungs of ovariectomized (OVX) or sham-operated female (F-Sham) controls were implanted with a progesterone (P4, 25 mg) (OVX-P4) or placebo pellet (OVX-Placebo), followed by sensitization and challenge with ovalbumin (OVA). Progesterone increased total inflammatory histologic scores, increased hyper-responsiveness to methacholine (MCh), increased select chemokines in the bronchoalveolar lavage (BAL) and serum, and increased ILC2 and neutrophil numbers, along the airways compared with F-Sham-OVA and OVX-Placebo-OVA animals. Lung ILC2 were sorted from F-Sham-OVA, OVX-Placebo-OVA and OVX-P4-OVA treated animals and stimulated with IL-33. OVX-P4-OVA lung ILC2 were more responsive to interleukin 33 (IL-33) compared with F-Sham-OVA treated, producing more IL-13 and chemokines following IL-33 stimulation. We confirmed the expression of the progesterone receptor (PR) on human ILC2, and showed that P4 + IL-33 stimulation also increased IL-13 and chemokine production from human ILC2. We establish that murine ILC2 are capable of responding to P4 and thereby contribute to allergic inflammation in the lung. We confirmed that human ILC2 are also hyper-responsive to P4 and IL-33 and likely contribute to airway exacerbations following allergen exposures in asthmatic women with increased symptoms around the time of menstruation.NEW & NOTEWORTHY There is a strong association between female biological sex and severe asthma. We investigated the allergic immune response, lung pathology, and airway mechanics in the well-described chicken egg ovalbumin (OVA) model with steady levels of progesterone delivered throughout the treatment period. We found that progesterone enhances the activation of mouse group 2 innate lymphoid cells (ILC2). Human ILC2 are also hyper-responsive to progesterone and interleukin 33 (IL-33), and likely contribute to airway exacerbations following allergen exposures in women with asthma.
Collapse
Affiliation(s)
- Shubhanshi Trivedi
- Division of Infectious Disease, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, United States
- George E. Whalen Department of Veterans Affairs Medical Center, Salt Lake City, Utah, United States
| | - Cassandra E Deering-Rice
- Department of Pharmacology and Toxicology, University of Utah College of Pharmacy, Salt Lake City, Utah, United States
| | - Samuel E Aamodt
- Division of Pulmonary Medicine, Department of Internal Medicine, University of Utah Health, Salt Lake City, Utah, United States
| | - Thomas P Huecksteadt
- George E. Whalen Department of Veterans Affairs Medical Center, Salt Lake City, Utah, United States
| | - Elizabeth J Myers
- Division of Neuroimmunology, Department of Neurology, University of Utah Health, Salt Lake City, Utah, United States
| | - Karl A Sanders
- Division of Pulmonary Medicine, Department of Internal Medicine, University of Utah Health, Salt Lake City, Utah, United States
- George E. Whalen Department of Veterans Affairs Medical Center, Salt Lake City, Utah, United States
| | - Robert Paine
- Division of Pulmonary Medicine, Department of Internal Medicine, University of Utah Health, Salt Lake City, Utah, United States
- George E. Whalen Department of Veterans Affairs Medical Center, Salt Lake City, Utah, United States
| | - Kristi J Warren
- Division of Pulmonary Medicine, Department of Internal Medicine, University of Utah Health, Salt Lake City, Utah, United States
- George E. Whalen Department of Veterans Affairs Medical Center, Salt Lake City, Utah, United States
| |
Collapse
|
12
|
Baskerville R, Castell L, Bermon S. Sports and Immunity, from the recreational to the elite athlete. Infect Dis Now 2024; 54:104893. [PMID: 38531477 DOI: 10.1016/j.idnow.2024.104893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 03/22/2024] [Indexed: 03/28/2024]
Abstract
The pivotal role of the immune system in physical activity is well-established. While interactions are complex, they tend to constitute discrete immune responses. Moderate intensity exercise causes leukocytosis with a mild anti-inflammatory cytokine profile and immunoenhancement. Above a threshold of intensity, lactate-mediated IL-6 release causes a proinflammatory state followed by a depressed inflammatory state, which stimulates immune adaptation and longer term cardiometabolic enhancement. Exercise-related immune responses are modulated by sex, age and immunonutrition. At all ability levels, these factors collectively affect the immune balance between enhancement or overload and dysfunction. Excessive training, mental stress or insufficient recovery risks immune cell exhaustion and hypothalamic pituitary axis (HPA) stress responses causing immunodepression with negative impacts on performance or general health. Participation in sport provides additional immune benefits in terms of ensuring regularity, social inclusion, mental well-being and healthier life choices in terms of diet and reduced smoking and alcohol, thereby consolidating healthy lifestyles and longer term health. Significant differences exist between recreational and professional athletes in terms of inherent characteristics, training resilience and additional stresses arising from competition schedules, travel-related infections and stress. Exercise immunology examines the central role of immunity in exercise physiology and straddles multiple disciplines ranging from neuroendocrinology to nutrition and genetics, with the aim of guiding athletes to train optimally and safely. This review provides a brief outline of the main interactions of immunity and exercise, some influencing factors, and current guidance on maintaining immune health.
Collapse
Affiliation(s)
| | - Linda Castell
- Green Templeton College, University of Oxford, Oxford, UK
| | - Stéphane Bermon
- World Athletics Health and Science Department, Monaco and LAMHESS, University Côte d'Azur, Nice, France
| |
Collapse
|
13
|
Barone JC, Wenzel E, Alluri V, Moriarity D, Pinna G, Walsh E, Rubinow DR, Morrow AL, Eisenlohr-Moul TA. Effects of estrogen and progesterone on neuroactive steroids and cytokines in patients with suicidality. Psychoneuroendocrinology 2023; 157:106359. [PMID: 37611527 PMCID: PMC10543480 DOI: 10.1016/j.psyneuen.2023.106359] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/02/2023] [Accepted: 08/07/2023] [Indexed: 08/25/2023]
Abstract
BACKGROUND In ovulating psychiatric patients experiencing suicidality, suicidal ideation (SI) often peaks perimenstrually. Our recent double-blind, placebo-controlled, crossover randomized clinical trial (RCT; NCT03720847) showed that perimenstrual administration of estradiol and progesterone (EP) can prevent this peak in SI and depressed mood. In this pre-registered follow-up analysis, we studied how the menstrual cycle and experimental manipulation affected two neurobiological systems associated with the menstrual cycle and suicide risk: GABAergic neuroactive steroids (NAS) and peripheral cytokines. METHODS In 26 psychiatric outpatients with natural menstrual cycles and past-month SI, we analyzed serum samples from three blood draws (midluteal, perimenstrual, midfollicular) per experimental condition (EP vs placebo) timed to a luteinizing hormone-surge ovulation test. Using gas chromatography/mass spectrometry (GC/MS), we measured the progesterone (P4)-derived pregnane NAS (3α,5α)- 3-hydroxypregnan20-one (3α,5α-THP), (3α,5β)- 3-hydroxypregnan-20-one (3α,5β-THP), (3α,5α)- 3,21-dihydroxypregnan-20-one (3α,5α-THDOC), (3α,5α)- 3-hydroxyandrostan-17-one (3α,5α-A), the androstane NAS (3α,5β)- 3-hydroxyandrostan-17-one (3α,5β-A), (3α,5α,17β)-androstane-3,17-diol (3α,5α-A-diol), (3α,5β,17β)-androstane-3,17-diol (3α,5β-A-diol), and their precursor pregnenolone. High sensitivity multiplex assay kits quantified peripheral cytokines IL-1β, IL-6, and TNF-α. RESULTS P4-derived NAS fluctuated in parallel with P4 and increased with exogenous perimenstrual administration of EP. Conversely, androstane NAS either did not fluctuate or fluctuated inversely from P4, and these NAS decreased with exogenous EP. Peripheral cytokines did not show cyclical patterns, but each significantly predicted SI, depressed mood, or anxiousness. Concomitant SSRI medication use predicted lower androstane NAS. CONCLUSIONS While preliminary and exploratory, our findings provide critical descriptive context for future studies. Further, our work presents menstrual cycle-related patterns for ten frequently-studied biomarkers, allowing for improved quality of comparisons involving naturally-cycling populations in research.
Collapse
Affiliation(s)
- Jordan C Barone
- University of Illinois at Chicago, Dept of Psychiatry, 912 S Wood St, Chicago, IL 60612, USA.
| | - Elizabeth Wenzel
- University of Illinois at Chicago, Dept of Psychiatry, 912 S Wood St, Chicago, IL 60612, USA
| | - Viraja Alluri
- University of Illinois at Chicago, Dept of Psychiatry, 912 S Wood St, Chicago, IL 60612, USA
| | - Daniel Moriarity
- University of California, Los Angeles, Department of Psychiatry and Biobehavioral Sciences, 757 Westwood Plaza #4, Los Angeles, CA 90095, USA; Stanford University, Department of Genetics, 291 Campus Drive, Stanford, CA 94305, USA
| | - Graziano Pinna
- University of Illinois at Chicago, Dept of Psychiatry, 912 S Wood St, Chicago, IL 60612, USA
| | - Erin Walsh
- University of North Carolina-Chapel Hill, Department of Psychiatry, 101 Manning Dr. #1, Chapel Hill, NC 27514, USA
| | - David R Rubinow
- University of North Carolina-Chapel Hill, Department of Psychiatry, 101 Manning Dr. #1, Chapel Hill, NC 27514, USA
| | - A Leslie Morrow
- University of North Carolina-Chapel Hill, Department of Psychiatry, 101 Manning Dr. #1, Chapel Hill, NC 27514, USA
| | - Tory A Eisenlohr-Moul
- University of Illinois at Chicago, Dept of Psychiatry, 912 S Wood St, Chicago, IL 60612, USA
| |
Collapse
|
14
|
Mamat @ Yusof MN, Chew KT, Kampan NC, Shafiee MN. Expression of PD-1 and PD-L1 in Endometrial Cancer: Molecular and Clinical Significance. Int J Mol Sci 2023; 24:15233. [PMID: 37894913 PMCID: PMC10607163 DOI: 10.3390/ijms242015233] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/07/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
The landscape of diagnosing and treating endometrial cancer is undergoing a profound transformation due to the integration of molecular analysis and innovative therapeutic approaches. For several decades, the cornerstone treatments for endometrial cancer have included surgical resection, cytotoxic chemotherapy, hormonal therapy, and radiation therapy. However, in recent years, the concept of personalised medicine has gained momentum, reshaping the way clinicians approach cancer treatment. Tailoring treatments based on specific biomarkers has evolved into a standard practice in both initial and recurrent therapy protocols. This review aims to provide an in-depth exploration of the current state of molecular analysis and treatment strategies in the context of endometrial cancer, focusing on the immunological aspect of the PD-1/PD-L1 axis. Furthermore, it seeks to shed light on emerging and innovative approaches that hold promise for the future modulation of endometrial cancer treatments. In essence, as researchers delve into the complex molecular landscape of endometrial cancer and harness the understanding of the PD-1/PD-L1 axis, we are paving the way for more targeted, effective, and personalised therapies that have the potential to significantly improve the outcomes and quality of life for patients with this challenging disease.
Collapse
Affiliation(s)
| | | | | | - Mohamad Nasir Shafiee
- Gynaecologic-Oncology Unit, Department of Obstetrics and Gynaecology, Hospital Canselor Tuanku Muhriz, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
15
|
Doshi B, Athans SR, Woloszynska A. Biological differences underlying sex and gender disparities in bladder cancer: current synopsis and future directions. Oncogenesis 2023; 12:44. [PMID: 37666817 PMCID: PMC10477245 DOI: 10.1038/s41389-023-00489-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 08/21/2023] [Accepted: 08/23/2023] [Indexed: 09/06/2023] Open
Abstract
Sex and gender disparities in bladder cancer have long been a subject of interest to the cancer research community, wherein men have a 4 times higher incidence rate than women, and female patients often present with higher-grade disease and experience worse outcomes. Despite the known differences in disease incidence and clinical outcomes between male and female bladder cancer patients, clinical management remains the same. In this review, we critically analyze studies that report on the biological differences between men and women and evaluate how these differences contribute to sex and gender disparities in bladder cancer. Distinct characteristics of the male and female immune systems, differences in circulating hormone levels and hormone receptor expression, and different genetic and epigenetic alterations are major biological factors that all likely contribute to disparate incidence rates and outcomes for male and female bladder cancer patients. Future preclinical and clinical studies in this area should employ experimental approaches that account for and consider sex and gender disparities in bladder cancer, thereby facilitating the development of precision medicine for the effective treatment of bladder cancer in all patients.
Collapse
Affiliation(s)
- Bhavisha Doshi
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14203, USA
| | - Sarah R Athans
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14203, USA
| | - Anna Woloszynska
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14203, USA.
| |
Collapse
|