bims-simsho Biomed News
on Systems immunology and sex hormones
Issue of 2025–09–07
24 papers selected by
Chun-Chi Chang, Lunds universitet



  1. Sci Rep. 2025 Aug 31. 15(1): 32032
      A strong sex-bias characterizes many respiratory immune diseases and has been attributed to sexually dimorphic immune responses. However, the role of lung-resident immunity in this context remains elusive. Here, we thoroughly characterized the lung-resident immune landscape in male and female mice, with a special focus on sex hormone effects in this context. Androgens were found to exert the strongest effects, markedly impacting B cells and neutrophils in both male and female lungs. Castrated males exhibited increased, while testosterone-treated females and males decreased lung-residing B cells. Testosterone supplementation of castrated males and females resulted in increased lung-residing neutrophils. Sex-mismatched orthotopic lung transplantation further supported these findings, since lungs isolated from female donors exhibited reduced tissue-residing B cells after their transplantation into male recipients. For the remaining lung-resident immune cell populations, sex differences were observed at the level of cell frequencies, with male lungs exhibiting higher frequencies of alveolar macrophages and lower frequencies of lung-resident dendritic cells and CD4⁺ tissue-resident memory T cells. Castration reversed some of these findings. Our findings highlight that the sexual dimorphism of the lung-resident immune landscape is modulated by sex hormones and especially androgens, thereby providing insights into the sex-specific manifestation of respiratory immune diseases.
    Keywords:  Lung; Sexual dimorphism; Testosterone; Tissue-resident immunity
    DOI:  https://doi.org/10.1038/s41598-025-15941-6
  2. Eur J Immunol. 2025 Sep;55(9): e70051
      Dysbiosis of the vaginal microbiome is associated with increased inflammation in the female genital tract. Microbiota associated with bacterial vaginosis (BV), such as Gardnerella vaginalis, Megasphaera elsdenii, and Prevotella timonensis, replace the health-associated bacterium Lactobacillus crispatus and cause inflammation affecting mucosal integrity and immunity. However, it remains unclear how these BV-associated bacteria modulate immune cells and enhance inflammation. Here, we investigated whether BV-associated bacteria directly affected dendritic cell (DC) function. Notably, P. timonensis but not M. elsdenii induced cell-cell clustering between monocytic cell lines and, importantly, between primary DCs and primary CD4 T cells. Our data indicate that this increased clustering is independent of LFA-1. Moreover, P. timonensis enhanced DC-mediated CD4 T cell proliferation. Altogether, these results suggest that P. timonensis-induced cell-cell clustering contributes to the elevated mucosal inflammation observed during bacterial vaginosis.
    Keywords:  CD4 T cells; Prevotella timonensis; adhesion; bacterial infections; cellular proliferation; cell–cell clustering; dendritic cells (DCs); immune regulation; vaginal dysbiosis; vaginal microbiome
    DOI:  https://doi.org/10.1002/eji.70051
  3. Front Neurol. 2025 ;16 1648017
       Objective: Sex hormones play a key role in migraine pathophysiology, yet their impact in men remains unclear. This study investigates sex hormone profiles and their potential relationship with Calcitonin Gene-Related Peptide (CGRP) in men with episodic migraine.
    Methods: We analyzed serum blood levels of sex hormones testosterone, estradiol (E2), progesterone, follicle-stimulating hormone (FSH), luteinizing hormone (LH) and CGRP in age and body mass index (BMI)-matched men with and without migraine.
    Results: A total of 120 male participants (n = 60 with migraine and n = 60 without migraine) completed the study. The mean age was 44.4 ± 14.4 years in migraine group and 44.5 ± 16.2 in the control group. Men with migraine had lower progesterone levels (0.2 nmoL/L, IQR 0.2) and a higher E2 to progesterone (E2/P) ratio (0.33, IQR 0.26) compared to healthy controls (0.5 nmoL/L, IQR 0.2, p < 0.001; 0.25, IQR 0.19, p < 0.02). Median E2 was 0.09 nmoL/L (IQR 0.03) in migraine patients and 0.12 nmoL/L (IQR 0.04) in controls (p = 0.07). There were no significant differences in testosterone, testosterone to E2 (T/E2) ratio, LH and FSH levels. CGRP serum levels did not differ between groups and showed no correlation with sex hormone levels. Subgroup analysis revealed no differences in hormone or CGRP levels between migraine patients with and without aura.
    Discussion: Our findings indicate higher progesterone levels and lower E2/P ratios in healthy men compared to those with migraine, suggesting a potential association between sex hormone profiles and migraine in men. These results warrant further investigation into the hormonal modulation of migraine beyond the female population.
    Keywords:  CGRP; estrogen; men; migraine; progesterone; sex hormones; testosterone
    DOI:  https://doi.org/10.3389/fneur.2025.1648017
  4. J Leukoc Biol. 2025 Aug 29. pii: qiaf123. [Epub ahead of print]
      Trained immunity amplifies innate immune responses in an antigen-independent manner. This study explored the ability of trained human primary macrophages to modulate the phenotype and function of T cells. Macrophages play an important role in antigen presentation, resulting in T-cell activation and antigen-specific clonal expansion; however, few studies have investigated whether trained immunity induction in macrophages modulates T cell activation. Here, through surface marker analysis of naive, β-glucan-, and BCG-trained macrophages, we identified eight distinct macrophage clusters following trained immunity induction. One of these populations showed an increase in surface activation markers CD40 and CD86, as well as MHC molecules. In vitro co-culture of T cells with autologous BCG-trained macrophages resulted in a skewing towards TH17 cells. We also observed an increase in TH17 percentage after BCG vaccination of human subjects. The bias towards TH17 triggered by trained macrophages required direct T cell to macrophage contact. Trained macrophages potentiated TH17 skewing independently of the antigen presented. While co-cultures of T cells and BCG-trained macrophages responded with higher production of interferon (IFN)-γ and interleukin (IL)-17 after stimulation, no clear shifts towards effector or memory T cells were observed. In conclusion, this study provides evidence that BCG-trained macrophages can modulate T cell function towards a TH17 phenotype, suggesting that BCG-induced trained immunity has the potential to enhance not only innate immune responses but also to modify adaptive T cell immunity.
    Keywords:  BCG; T cells; Trained Immunity; macrophages; vaccination
    DOI:  https://doi.org/10.1093/jleuko/qiaf123
  5. Fertil Steril. 2025 Aug 26. pii: S0015-0282(25)01847-3. [Epub ahead of print]
      Polycystic Ovary Syndrome (PCOS) is the most common endocrine disorder among women of reproductive age, with a prevalence estimated between 10-13%. It is characterized by ovulatory dysfunction, hyperandrogenism, and polycystic ovarian morphology. Anti-Müllerian hormone (AMH) plays a key role in regulating normal reproductive function in both males and females. Over the past few decades, significant progress has been made in understanding AMH, with numerous studies revealing its unexpected roles throughout the Hypothalamic-Pituitary-Gonadal (HPG) axis. AMH and its receptor are also produced in the forebrain, where they contribute to the proper migration of gonadotropin-releasing hormone (GnRH) neurons during development. At the hypothalamic and pituitary levels, AMH has been shown to increase the frequency of GnRH pulses, which in turn leads to increased luteinizing hormone (LH) secretion. In PCOS, this regulatory mechanism appears to be disrupted. The LH/follicle-stimulating hormone (FSH) ratio is often elevated due to altered GnRH pulse frequency. Elevated AMH levels contribute to this dysregulation by increasing GnRH pulsatility, which leads to enhanced LH secretion. This, in turn, stimulates theca cells to produce excessive androgens, resulting in hyperandrogenism, follicular arrest, and anovulation. AMH levels are strongly correlated with these clinical features of PCOS and are predictive of outcomes in assisted reproductive treatments (ART), including a higher risk of adverse pregnancy outcomes. Additionally, offspring born to mothers with PCOS tend to have elevated levels of AMH and androgens at birth, which may alter the HPG axis and contribute to the development of PCOS later in life.
    Keywords:  AMH; AMHR2; GnRH; MIS; Neuroendocrine; PCOS and PCOM
    DOI:  https://doi.org/10.1016/j.fertnstert.2025.08.023
  6. J Obstet Gynaecol Res. 2025 Sep;51(9): e70056
       INTRODUCTION: Polycystic ovary syndrome (PCOS) is a complex endocrine disorder affecting approximately 20% of women of reproductive age. The impact of high androgen levels on pregnancy and fetal outcomes is complex and multifaceted, largely due to the heterogeneous nature of PCOS.
    MATERIALS AND METHODS: This prospective cohort study included 65 pregnant women diagnosed with PCOS (using the Rotterdam criteria) and 65 age-matched healthy pregnant controls at Harran University Hospital. Blood samples were collected at 18-19 weeks of gestation to quantify testosterone and sex hormone-binding globulin (SHBG) levels, and the free androgen index (FAI) was calculated. Demographic and clinical data were recorded, and obstetric complications and birth outcomes were analyzed, adjusting for potential confounding factors using multivariate analysis.
    RESULTS: Pregnant women with PCOS exhibited lower parity (1.8 ± 1.0 vs. 2.2 ± 1.3, p = 0.033), lower birth weight (2827.0 ± 579.3 g vs. 3059.1 ± 561.2 g, p = 0.022), and lower SHBG levels (67.8 ± 58.8 nmol/L vs. 125.3 ± 36.8 nmol/L, p <0.001). Conversely, they demonstrated higher BMI (31.1 ± 6.1 vs. 27.6 ± 5.1, p = 0.001), total testosterone (32.6 ± 14.1 vs. 26.1 ± 8.1, p = 0.009), and FAI levels (136.9 ± 128.2 vs. 31.2 ± 39.4, p <0.001). The incidence of complications such as intrauterine growth retardation (IUGR) and small for gestational age (SGA) was higher in the PCOS group, while the healthy birth rate was lower. The incidence of preeclampsia was also higher in the PCOS group. In the PCOS group, the FAI demonstrated a negative correlation with gestational week and birth weight (r = -0.376, p = 0.002), indicating that as these values increased, the FAI exhibited a corresponding decrease.
    CONCLUSIONS: This study demonstrates the adverse effects of hyperandrogenism on pregnancy and fetal development in women with PCOS, highlighting the clinical significance of this condition. Further research is required to elucidate the underlying mechanisms of hyperandrogenism and to identify effective strategies to improve pregnancy outcomes in women with PCOS.
    Keywords:  PCOS; SGA; SHBG; androgen; free androgen index; premature
    DOI:  https://doi.org/10.1111/jog.70056
  7. Elife. 2025 Sep 02. pii: e106443. [Epub ahead of print]14
      The long-term functional adaptation of innate immune cells following an initial stimulation, referred to as trained immunity or innate immune memory, enhances responsiveness and protection against secondary infections. Toll-like receptors (TLRs), an evolutionarily conserved family, recognize microbial-associated molecular patterns, initiating innate and adaptive immune responses. TLR signaling cascades induce the production of pro-inflammatory cytokines, antimicrobial peptides, and interferons, promoting pathogen clearance, while also driving epigenetic and metabolic reprogramming that enhances immune responses and protection to subsequent challenges. However, TLRs also recognize endogenous ligands contributing to chronic inflammation and autoimmune diseases. This review examines the role of TLRs and their various agonists in mediating trained immunity across diverse immune cell types, with an emphasis on their dual role in protecting against infections and chronic inflammation. It highlights recent clinical trials of TLR agonists as immunomodulatory agents and their therapeutic potential in infectious diseases and cancer. By providing an in-depth analysis of TLR-driven trained immunity, this review highlights the extensive influence of TLRs on immune cell populations and their implications for the development of novel, broad-spectrum immunotherapies.
    Keywords:  Toll-like receptors; chronic inflammation; immunology; inflammation; innate immunity; microbial infection; trained immunity
    DOI:  https://doi.org/10.7554/eLife.106443
  8. Reproduction. 2025 Aug 22. pii: REP-25-0126. [Epub ahead of print]
      Polycystic ovary syndrome (PCOS) is a common and heterogeneous disorder currently diagnosed only in reproductive-age women. Familial clustering and twin studies have provided strong evidence for a genetic contribution to PCOS pathogenesis. First-degree relatives, including males and non-reproductive-age females, have reproductive and metabolic phenotypes consistent with a genetic susceptibility to these traits. PCOS is now recognized as a complex trait influenced by both genetic and environmental factors. Genome-wide association studies have identified ∼30 loci linked to PCOS, implicating pathways involved in gonadotropin secretion and action, folliculogenesis, steroidogenesis, age at menopause, and carbohydrate metabolism. Next-generation sequencing has found rare variants in AMH, AMHR2, and DENND1A, supporting these genes' central role in developing PCOS. Epigenetic mechanisms such as DNA methylation and non-coding RNAs influence gene regulation and may contribute to phenotypic heterogeneity. Unsupervised clustering has identified distinct reproductive and metabolic subtypes with unique genetic architectures, providing a biologically meaningful framework for classification. This shift from expert opinion-based diagnosis to data-driven classification has the potential to transform PCOS management and enable precision medicine approaches tailored to distinct subtypes of the disorder.
    Keywords:  Cluster Analysis; GWAS; Mendelian Randomization; Next-Generation Sequencing; PCOS
    DOI:  https://doi.org/10.1530/REP-25-0126
  9. Front Immunol. 2025 ;16 1662942
       Introduction: Numerous pre-vaccination factors are known to be associated with differential responses to influenza vaccination, including age, prior infection, vaccination history, immune cell frequencies, and transcriptomic profiles. However, plasma chemokines and cytokines are relatively unexplored. Given that older individuals have generally higher levels of inflammatory molecules in circulation, termed inflammaging, and also respond poorly to vaccination, plasma immune profiles likely play a role in effective response to influenza vaccination.
    Methods: A cohort of 100 people were sampled pre- (Day 0) and post-vaccination (Day 7) with the inactivated, quadrivalent Fluzone construct in the autumn of 2019 (UGA4). Plasma chemokines and cytokines were quantified by 38-plex Luminex assay, with ultrasensitive quantification of additional analytes by Single Molecule Array Technology (Simoa) assay. Antibodies against individual strains of influenza and serological response to vaccination were determined by Day 0 hemagglutination inhibition (HAI) titer and change in HAI titers from Day 0 to Day 28, respectively.
    Results: Age was strongly associated with pre-vaccination HAI titers and differences in plasma analytes, but not changes in HAI titers post-vaccination. High plasma levels of eotaxin (CCL11) and MDC (CCL22) pre-vaccination were associated respectively with ineffective and effective serological response to vaccination. Increasing plasma levels of IFN-γ, IL-17A, and IL-15 from Day 0 to Day 7 post-vaccination were associated with effective serological response to vaccination.
    Discussion: In conclusion, plasma chemokines and cytokine levels prior to or in the first few days post-influenza vaccination may be predictive of serological responses to vaccination, with changes in IFN-γ, IL-17A, and IL-15 post-vaccination possibly indicative of the activation of cell-mediated immunity. These findings support the need for larger, high-resolution studies exploring the role of plasma proteomics in serological responses to influenza vaccination.
    Keywords:  FluZone vaccination; chemokines; cytokines; influenza; serological immunity
    DOI:  https://doi.org/10.3389/fimmu.2025.1662942
  10. Reprod Biol Endocrinol. 2025 Sep 01. 23(1): 120
       BACKGROUND: This retrospective cohort study aimed to evaluate the impact of insulin resistance (IR) on clinical outcomes in polycystic ovary syndrome (PCOS) patients undergoing in vitro fertilization (IVF) or intracytoplasmic sperm injection (ICSI) treatment.
    METHODS: A total of 1,768 PCOS patients undergoing IVF/ICSI cycles at Shenzhen Zhongshan Obstetrics & Gynecology Hospital between October 2010 and November 2024 were stratified into two cohorts: non-IR group (HOMA index < 2.69, n = 867) and IR group (HOMA index ≥ 2.69, n = 901). Baseline characteristics and clinical outcomes were compared between the groups. Linear logistic regression and multivariate logistic regression analysis were conducted to assess the independent impact of IR on fertilization efficiency and pregnancy outcomes.
    RESULTS: Patients with IR exhibited significantly higher BMI (25.44 ± 3.55 vs. 21.59 ± 3.20, p < 0.001), longer infertility duration (3.74 ± 2.75 vs. 3.25 ± 2.43, p < 0.001), increased antral follicle counts (26.74 ± 10.74 vs. 25.05 ± 9.79, p < 0.001) and lower basal follicle-stimulating hormone (FSH) level (9.78 ± 3.25 vs. 10.64 ± 3.83, p < 0.001) compared to those without IR. Additionally, the fertilization rate (82.02% vs. 83.86%, p = 0.005) and 2PN rate (81.07% vs. 83.96%, p < 0.001) were significantly lower in PCOS patients with IR. Linear regression indicated that IR had a more pronounced inverse effect on 2PN rate (B: -2.540, p = 0.009) than on fertilization rate (B: -0.664, p = 0.490). Subgroup analysis and interaction analysis demonstrated that IR functioned as an independent risk factor for impaired oocyte fertilization in normal-weight PCOS patients (B: -22.694, p = 0.011). No statistically significant associations between IR status and clinical or live birth pregnancy outcomes were observed in the regression models.
    CONCLUSIONS: IR adversely affects oocyte fertilization competence and early embryonic development in normal-weight PCOS patients undergoing assisted reproductive technology (ART). These effects may be attributable to IR-induced metabolic dysregulation, which compromises folliculogenic and cytoplasmic maturation processes critical to gamete competence. These findings underscore the importance of addressing metabolic dysfunction in IR-affected PCOS populations to optimize ART outcomes.
    TRIAL REGISTRATION: This is a retrospective study.
    Keywords:  Fertilization rate; HOMA; IVF/ICSI; Insulin resistance; Polycystic ovary syndrome
    DOI:  https://doi.org/10.1186/s12958-025-01453-5
  11. Front Immunol. 2025 ;16 1605972
      Tissue-resident memory T cells (TRM) represent a heterogeneous population of T cells that exhibit both effector and memory functionalities. They express specific gene signatures that enable them to occupy tissues without recirculating, thus providing a first response against reencountered pathogens or antigens. TRM have been implicated in the pathogenesis of various diseases, including autoimmune disorders, infections, and cancers. This has prompted interest in targeting TRM as a potential therapeutic strategy. Epigenetic modifications, which frequently occur in immune cells across various disease states, play a significant role not only in tissue homeostasis but also in disease progression. Emerging evidence suggests that the epigenetic landscape of TRM is altered in pathogenic conditions, impacting their differentiation, maintenance, and function. Nevertheless, the precise mechanisms remain poorly understood. This review seeks to provide a comprehensive overview of the epigenetic regulation of TRM, focusing on key areas such as chromatin accessibility, DNA methylation, histone modifications, and non-coding RNAs. Importantly, a deeper understanding of these epigenetic mechanisms will pave the way for novel therapeutic strategies, such as modulating TRM activity in autoimmune diseases, enhancing tissue-specific immunity through vaccines, or improving immunotherapeutic efficacy in cancer.
    Keywords:  DNA methylation; chromatin accessibility; epigenetics; histone modification; non-coding RNAs; tissue resident memory T cells
    DOI:  https://doi.org/10.3389/fimmu.2025.1605972
  12. J Clin Invest. 2025 Sep 02. pii: e192469. [Epub ahead of print]135(17):
      Trained immunity (TRIM) is a form of long-lasting functional reprogramming of innate immune cells and their progenitors that enhances responsiveness to subsequent stimuli. Although first characterized in myeloid cells, TRIM was recently extended to nonmyeloid cell types, including endothelial and glial cells, which also exhibit stimulus-driven, memory-like behavior. While initially recognized as a protective mechanism, particularly in the context of vaccines and acute infections, TRIM can also become maladaptive, promoting chronic inflammation, immune dysfunction, and disease. This Review focuses on virus-induced TRIM while also addressing microbial, metabolic, and endogenous inducers. We examine key ligands and receptors that initiate TRIM and dissect the associated signaling and epigenetic pathways. Importantly, we argue that maladaptive TRIM arises not from a specific ligand, receptor, or molecular event, but from contextual factors such as stimulus persistence, dose, tissue microenvironment, and preexisting inflammation. The nature of the secondary challenge also shapes whether a trained response is adaptive or maladaptive. We further discuss TRIM induction in the bone marrow, involvement of both myeloid and nonmyeloid cells, and the role of lipid rafts in sustaining TRIM. We review maladaptive TRIM's potential contribution to systemic diseases, such as atherosclerosis, diabetes, sepsis, cancer, and autoimmunity, along with its influence on viral vaccine responses. Finally, we outline potential strategies to redirect maladaptive TRIM and propose key outstanding questions for future research.
    DOI:  https://doi.org/10.1172/JCI192469
  13. Am J Physiol Endocrinol Metab. 2025 Aug 28.
      The circadian system coordinates 24-hour cycles of internal biological processes with the environmental light-dark cycle. Abrupt shifts in the timing of the light-dark cycle misalign internal circadian clocks with the environment and cause jet lag until resynchronization occurs. The objective of this study was to investigate the sex difference in simulated jet lag in mice. Female mice resynchronized faster than male mice to 6-hour advances of the light-dark cycle that mimicked eastward travel. Circulating estradiol was necessary and sufficient for rapid resynchronization in female mice since ovariectomized females resynchronized slower than mice treated with estradiol. Disabling estrogen receptor alpha (ERα), but not ERβ or G-protein coupled estrogen receptor 1 (GPER1), abolished the sex difference in resynchronization. To investigate ERα-dependent mechanisms that regulate the rate of resynchronization, we measured the endogenous circadian period and the magnitudes of phase shifts to light pulses in male and female wild-type and ERα knockout mice. Wild-type females had shorter periods and greater phase delays in response to light pulses given in the early subjective night than male mice. Disabling ERα abolished the these sex differences by lengthening circadian period and reducing the magnitudes of phase delays. Together these data suggest that ERα alters the rate of resynchronization to shifted light-dark cycles by regulating period length and phase shift magnitude in female mice. Understanding the mechanisms underlying the sex difference in resynchronization to shifted light-dark cycles can be used to develop strategies to alleviate jet lag and circadian misalignment.
    Keywords:  circadian; estrogens; females; males; photoentrainment
    DOI:  https://doi.org/10.1152/ajpendo.00268.2025
  14. Front Neurol. 2025 ;16 1616992
       Introduction: Leber's hereditary optic neuropathy (LHON) is a maternally inherited condition due to mitochondrial DNA (mtDNA) mutations usually affecting young men within their thirties, while women seem protected by estrogens with a female-to-male ratio of 1:3. Late-onset cases (over 40 years of age) are usually associated to toxic exposure to tobacco smoke or drugs causing mitochondrial dysfunction.
    Results: We describe two cases of LHON remarkable for their late onset (> 60 years) in the absence of classic toxic factors. They were both affected by advanced prostate cancer and developed LHON after introduction of enzalutamide, an antagonist of androgens' receptor, in association with leuprolide, a gonadotropin-releasing hormone (GnRH) analogue, used in the context of Androgen deprivation therapy (ADT). Both patients presented very low serum levels of gonadotropin, estrogens and androgens compatible with hormonotherapy. MtDNA copy number in our probands resulted significantly reduced (like other LHON affected cases), compared to age-matched LHON unaffected mutation carriers and controls.
    Discussion: The role of hormones in LHON pathogenesis remains still debated. Recent evidence suggests a protective effect of estrogens in increasing mitochondrial biogenesis (and mtDNA copy number), partially explaining the gender bias of the disease, while the role of androgens is yet to be fully understood. Considering the effect of the ADT on circulating hormonal levels and their reciprocal interactions, we hypothesize that in a context of already low estrogens levels due to GnRH analogue, the block of androgens receptors by Leuprolide further imbalance the estrogens to androgens ratio and eventually trigger the disease.
    Keywords:  Leber’s hereditary optic neuropathy; androgen deprivation therapy; estrogens; hormones; mitochondrial disease
    DOI:  https://doi.org/10.3389/fneur.2025.1616992
  15. Biol Reprod. 2025 Aug 30. pii: ioaf197. [Epub ahead of print]
       PURPOSE: Polycystic ovary syndrome (PCOS) affects 5-10% of women of reproductive age, with insulin resistance (IR) playing a central role in its pathophysiology in up to 80% of cases. This review aims to elucidate the molecular mechanisms by which insulin resistance disrupts ovarian function, contributing to menstrual irregularities and hyperandrogenism. It also evaluates current and emerging therapeutic strategies, with an emphasis on individualized management.
    METHODS: A comprehensive review of recent literature was conducted, focusing on molecular studies, clinical trials, and meta-analyses related to insulin signaling pathways in PCOS, as well as therapeutic interventions. Special attention was paid to ethnic variations, particularly in East Asian populations, and advances in genomic and metabolomic profiling.
    RESULTS: PCOS is characterized by selective insulin resistance, wherein metabolic insulin signaling is impaired, but steroidogenic and mitogenic pathways remain responsive, promoting hyperandrogenism and anovulation. East Asian women exhibit significant insulin resistance despite lower BMI compared to Western populations. Insulin resistance in PCOS also increases cardiometabolic risks and psychological burden. While lifestyle modification, insulin sensitizers, and hormonal therapy remain first-line treatments, novel approaches such as microbiome-targeted therapies and anti-inflammatory agents show promise.
    CONCLUSION: Understanding the complex interplay between insulin resistance and ovarian dysfunction is crucial for effective PCOS management. Integrating emerging molecular insights with digital health tools can facilitate personalized, multidisciplinary approaches that address both reproductive and metabolic aspects of PCOS, ultimately improving patient outcomes.
    Keywords:  Hyperandrogenism; Insulin resistance; Menstrual irregularities; Polycystic ovary syndrome; Treatment strategies
    DOI:  https://doi.org/10.1093/biolre/ioaf197
  16. Epigenomics. 2025 Sep 05. 1-13
      DNA methylation (DNAm) is a key epigenetic modification that dynamically regulates eukaryotic development over time. DNAm has been found to influence a variety of biological processes in both normative and pathological states, such as depression. Since DNAm can serve as an interface between environmental influence and gene expression, it is a mechanism studied in the context of many pathologies, including psychiatric. Depression is a complex and heterogeneous disorder strongly influenced by puberty, as evidenced by increased rates in both sexes after sexual maturation. However, this effect is more pronounced in females, contributing to its twofold increased lifetime prevalence compared to males. Additionally, depression is consistently associated with altered DNAm at specific genomic sites. In this review, we discuss how DNAm programming can affect functional pathways during puberty and in turn, influence disease outcomes. Here, we highlight the bidirectional relationship of steroid hormone surges during this sensitive period and DNAm, adding a layer of complexity and insight into the pathophysiology of depression. Specifically, we explore the extent of DNAm change throughout puberty, how it contributes to individual and sex-specific differences in puberty, and how it may influence the risk for depression.
    Keywords:  DNA methylation; depression; epigenomic regulation; puberty; steroid hormones
    DOI:  https://doi.org/10.1080/17501911.2025.2554569
  17. Endocrinology. 2025 Aug 29. pii: bqaf137. [Epub ahead of print]
      The hypothalamic-pituitary-ovarian (HPO) axis is a complex endocrine feedback mechanism controlling ovulation in female vertebrates. Balance of the HPO axis requires correct secretion of sex steroids from the ovarian follicle to inhibit release of gonadotropins from the pituitary. Several conditions of ovarian dysfunction such as menopause, Primary Ovarian Insufficiency (POI) and Polycystic Ovary Syndrome (PCOS) involve imbalances in the HPO axis, contributing to infertility. Intriguingly, these disorders also share a higher incidence of cognitive and emotional dysregulations, as well as a heightened risk of certain neurodegenerative conditions with age. It is understood that estradiol exerts neuroprotective functions, but gonadotropin signaling is less understood. High concentrations of circulating Follicle Stimulating Hormone (FSH) and Luteinizing Hormone (LH) have shown to contribute to neurodegenerative disease states, but are not addressed as part of traditional Hormone Replacement Therapy (HRT). To identify the mechanistic connections between ovarian disorders and heightened susceptibility of the brain to pathological aging, a multi-system experimental approach is required, considering each HPO axis player as an individual effector. In this review, we will summarize current knowledge on the effects of estradiol, progesterone, FSH and LH on neuronal susceptibility to pathology. We will describe ways in which the HPO axis becomes imbalanced during ovarian dysfunction, and how systemic inflammation can become an additional HPO axis effector. Finally, we will recommend solutions to the presented gaps in knowledge, and suggest avenues of future research to pursue development of therapeutics targeting both ovarian and brain health in patients.
    Keywords:  endocrine dysfunction; gonadotropins; neurodegeneration; neuroprotection; ovarian disorders; sex hormones
    DOI:  https://doi.org/10.1210/endocr/bqaf137
  18. Semin Oncol. 2025 Sep 03. pii: S0093-7754(25)00106-X. [Epub ahead of print]52(6): 152414
      While the efficacy of immune checkpoint inhibitors (ICIs) in advanced non-small-cell lung cancer (NSCLC) is well-established, sex-based differences in treatment responses remain insufficiently explored. This study examines how sex disparities impact ICI treatment outcomes in advanced-stage NSCLC, focusing on the role of tumor mutational burden (TMB) in these differences. This study analyzed data from 174 advanced-stage, chemotherapy-naïve, NSCLC patients treated with ICIs, including PD-1/PD-L1 and CTLA-4 inhibitors, to assess sex differences in treatment response and survival outcomes. Male patients with low TMB (<10 mut/Mb) had worse treatment responses compared to female patients. In contrast, no sex differences were observed in patients with high TMB, where both sexes exhibited similar therapeutic responses. These results suggest that high TMB may reduce the impact of sex on ICI efficacy, with male and female patients showing comparable outcomes. Furthermore, sex disparities in disease progression and overall survival were more evident in low-TMB patients, emphasizing the role of TMB in modulating sex-related differences in immunotherapy outcomes. This study highlights the importance of incorporating both sex and TMB into precision oncology. High TMB appears to equalize treatment responses between sexes, while low TMB may necessitate more personalized treatment strategies, particularly for male patients. Further research into the biological mechanisms underlying these differences is essential to optimize ICI therapies and enhance patient outcomes. Integrating both sex and TMB into clinical decision-making will help to develop more tailored and effective cancer immunotherapy.
    Keywords:  Immune checkpoint inhibitor; Immunotherapy; Non–small-cell lung cancer; Sex
    DOI:  https://doi.org/10.1016/j.seminoncol.2025.152414
  19. Mol Hum Reprod. 2025 Sep 05. pii: gaaf045. [Epub ahead of print]
      Human fertilization is a coordinated process involving nteraction of sperm with the oocyte. As the sperm pass through the female reproductive tract (FRT), they are presented with numerous challenges. These include navigating through highly viscous cervical mucus while evading immune responses to successfully fertilize the oocyte. The female immune system facilitates sperm selection while providing protection against pathogens. Neutrophils, a major component of the innate immune system, use mechanisms such as phagocytosis, neutrophil extracellular trap (NET) formation, and trogocytosis to play a crucial role in this process. With the help of sialic acid residues and N glycans present on its glycocalyx as well as certain proteins in the seminal plasma, the sperm modulate the immune responses in the FRT to their advantage. This review examines the various interactions which take place between the sperm and the FRT, the neutrophil mediated immune reaction occurring in the FRT, as well as the adaptations the sperm employ to overcome the immune challenges. Understanding these mechanisms provides critical insights into fertility and potential therapeutic targets for infertility.
    Keywords:  METs; NETs; female reproductive tract (FRT); fertilization; innate immunity; neutrophil; sperm; trogocytosis
    DOI:  https://doi.org/10.1093/molehr/gaaf045
  20. Immunol Rev. 2025 Sep;334(1): e70056
      Humoral immune responses are critical for protection against immune challenge by pathogens and transformed cells, while dysregulated antibody production is a hallmark of autoimmune diseases. T follicular helper (Tfh) cells are central to the development of humoral immunity, regulating B-cell maturation, including immunoglobulin class switch recombination and somatic hypermutation, and development of memory B and antibody-producing plasma cells. These events occur as B cells migrate to and differentiate within B cell follicles of secondary lymphoid organs, with this classical program of follicular B cell maturation providing systemic immune protection. Local humoral responses are also necessary for organismal defense against immune challenge. Accordingly, T-dependent B-cell help occurs outside of B-cell follicles, including in non-lymphoid tissues such as the lung, central nervous system, joints, and kidneys. The phenotype and function of T cells that provide humoral protection against pathogens and tumors and conversely promote autoimmunity at the tissue level both overlap with and are distinct from those of canonical Tfh cells. Here, we summarize current knowledge of these tissue T-B helper cells, focusing on their differentiation and function in infection, cancer, and autoimmunity.
    Keywords:  B cells; T cells; T follicular helper cells; T peripheral helper cells; autoimmunity
    DOI:  https://doi.org/10.1111/imr.70056
  21. Nat Rev Immunol. 2025 Sep 03.
      MHC-E is a highly conserved, non-polymorphic MHC protein that engages inhibitory and activating receptors on natural killer (NK) cells and T cells and can also present antigens to T cell receptors. NK cell responses driven by activating receptor interactions with MHC-E are implicated in controlling chronic viral infections and cancer. Immunotherapeutic targeting of interactions between MHC-E and inhibitory receptors to increase the activation of NK cells and T cells shows promise in improving antitumour immune responses. Furthermore, MHC-E-restricted CD8+ T cells elicited by cytomegalovirus-based vaccines might, for certain infections and cancers, be more effective than CD8+ T cells restricted by classical MHC class I or class II molecules. The ability of MHC-E to regulate or mediate both innate and adaptive immune responses independently of the MHC haplotype of an individual raises the possibility of new, universally effective vaccines and immunotherapies for infectious disease and cancer. Although the therapeutic exploitation of MHC-E is still in its infancy, recent advances in the understanding of MHC-E biology show enormous potential, as described in this Review.
    DOI:  https://doi.org/10.1038/s41577-025-01218-6
  22. World J Gastroenterol. 2025 Aug 28. 31(32): 104277
       BACKGROUND: Hepatocellular carcinoma (HCC) is a global health concern, representing the second most common cause of malignancy-related mortality in the world. The primary cause of HCC in the United States is chronic infection with the hepatitis C virus (HCV). Clinical observations have established sex-based differences in HCV infection with the disease progressing more severely and more rapidly in males and postmenopausal females compared to premenopausal females, suggesting that estrogens and their receptors may play an important role in hepatic defenses and development of HCV-mediated HCC. However, the precise mechanism of estrogen protection and their effects on inflammation is poorly understood.
    AIM: To determine whether estrogen receptor (ER) expression is correlated with the expression of tumor necrosis factor-alpha (TNF-α) in males and females with HCV-associated diseases.
    METHODS: The role of ERs in modulating innate immune responses was investigated using human liver tissues with HCV/cirrhosis and HCV/HCC. Messenger RNA (mRNA) and protein (nuclear and cytoplasmic) expression were measured for all markers of interest and compared to normal human liver tissue samples.
    RESULTS: ERβ was reported for the first time to have a greater mRNA expression than ERα in normal liver (P ≤ 0.001). In addition, ERβ mRNA expression was found to be decreased in diseased livers (P ≤ 0.05), while TNF-α expression was increased (P ≤ 0.0001). Upon stratifying by sex within each disease group, ESR1 was found to be negatively correlated with ESR2 in females with HCV/cirrhosis (r = -0.84, P ≤ 0.001), whereas males with HCV/cirrhosis were found to have a significant positive correlation (r = 0.57, P ≤ 0.05). ESR2 mRNA expression had a significant positive correlation with TNF-α in both HCV/cirrhosis (r = 0.61, P ≤ 0.001) and HCV/HCC patients (r = 0.45, P ≤ 0.05).
    CONCLUSION: All together, these findings indicate that changes in ERβ and TNF-α expression are associated with worsening disease, and may be part of the sex-dependent factors in HCC pathogenesis.
    Keywords:  Estrogen receptor; Hepatitis C virus; Hepatocellular carcinoma; Inflammation; Liver cirrhosis; Tumor necrosis factor alpha
    DOI:  https://doi.org/10.3748/wjg.v31.i32.104277
  23. Front Immunol. 2025 ;16 1646719
      Natural killer (NK) cells are lymphoid-derived cells that play a critical role in bridging innate and adaptive immunity. Given their ability to recognize and directly kill targets possessing missing or altered self-proteins and to induce indirect killing via recruitment of adaptive immunity, they are in a unique position to modulate host immunologic responses. These complex immune sentinels typically circulate in the peripheral blood and/or reside in lymphoid tissues. As the largest organ, human skin functions in front line immunological defense, though it has not historically been categorized as lymphoid tissue. Whether tissue-resident ILC populations originally derive from conventional circulating NK cells, or whether they interface as developmentally distinct entities with phenotypic overlap within particular inflammatory contexts remains a subject of ongoing investigation. This review seeks to consolidate the currently available literature regarding NK cell and ILC skin homing and innate immune function in healthy vs. lesional human skin (including infection, inflammatory/autoimmune conditions, and cutaneous malignancy). Importantly, we elucidate significant gaps in the understanding of the complex role for NK cells in skin homeostasis and pathology, and posit unique opportunities the accessibility of this secondary lymphoid organ provides for translational studies to improve our understanding of cutaneous immunity.
    Keywords:  cutaneous disease; innate immunity; innate lymphoid cells; natural killer cells; skin
    DOI:  https://doi.org/10.3389/fimmu.2025.1646719
  24. Oncogene. 2025 Sep 04.
      Androgen receptor (AR)-mediated signaling is essential for PC tumorigenesis. In the TCGA database we observed a positive correlation between ECD and AR expression. Consistently, Dihydrotestosterone (DHT) treatment of PC cell lines increased ECD mRNA and protein levels, and AR knockdown (KD) reduced ECD expression. Bioinformatic analysis predicted three consensus androgen response elements in the ECD promoter, and DHT treatment increased AR occupancy at the ECD promoter, and enhanced ECD promoter activity. Enzalutamide treatment decreased ECD levels, and ECD knockout (KO) in PC cells reduced oncogenic traits, suggesting a functional role of ECD to maintain PC oncogenesis. ECD mRNA and protein are overexpressed in PC patient tissues, and its overexpression predicts shorter survival. Overexpression of ECD in PC cell lines enhanced the oncogenic traits in vitro and developed faster and larger highly proliferative xenograft tumors. RNA-seq analysis of mouse tumors revealed an increase in mRNA levels of several glycolytic genes. ECD associates with mRNA of key glycolytic genes and is required for their stability, consistent with our recent demonstration of ECD is an RNA binding protein. Higher glucose uptake and glycolysis was seen upon ECD overexpression in PC cells. Together, we demonstrate the role of a novel AR target gene ECD in PC tumorigenesis.
    DOI:  https://doi.org/10.1038/s41388-025-03559-x