bims-simsho Biomed News
on Systems immunology and sex hormones
Issue of 2025–11–16
34 papers selected by
Chun-Chi Chang, Lunds universitet



  1. Biol Futur. 2025 Nov 11.
      The immune system is a complex network of cells and small molecules that play a crucial role in defending the body against pathogens and maintaining overall health. It has long been recognized that gonadal steroid hormones, such as estrogen, progesterone and testosterone, exert significant influences on immune responses, leading to gender-specific variations in immune function and disease susceptibility. This comprehensive review aims to provide a thorough analysis of the current knowledge regarding the interplay between sex hormones and immune responses, with a specific focus on gender differences. The article explores the fundamental mechanisms underlying the impact of sex hormones on the immune system, highlighting the intricate interactions between hormones and immune cells, including B- and T-lymphocytes and natural killer cells. Moreover, the review sheds light on the dynamic changes in sex hormone levels throughout one's lifespan and their potential implications on immune functions at different developmental stages, including puberty, pregnancy, and menopause. Notably, the role of sex steroid hormones in modulating innate and adaptive immune responses, cytokine production, immune cell trafficking, and immune-mediated tissue damages are also reviewed here. Importantly, the potential of these findings in the development of personalized medicine is huge, as understanding the intricate relationship between sex hormones and immune responses could lead to the design of targeted therapeutic interventions that consider gender-specific factors. In conclusion, this review details the crucial role of sex hormones in shaping immune responses and highlights the profound impact of gender on immune-related diseases.
    Keywords:  Anticancer; Estrogen; Gender differences; Immune responses; Sex hormones; Sex-hormone deprivation; Testosterone
    DOI:  https://doi.org/10.1007/s42977-025-00291-1
  2. J Leukoc Biol. 2025 Nov 10. pii: qiaf161. [Epub ahead of print]
      Neutrophils, our frontline of defense against pathogens, exhibit pronounced sexual dimorphism in ontogeny, phenotype, and effector functions. Throughout the human lifespan, estrogen and androgen signaling, together with sex-linked genetic regulators, orchestrate neutrophil production, maturation, and immune activity, contributing to immunological differences between sexes observed across lifespan. Differences in neutrophil antimicrobial and immune responses contribute to disease susceptibility, with females having stronger antimicrobial defenses but also a higher risk of autoimmunity, while males experience greater severity of infections and different cancer risks. This review summarizes and discusses the existing evidence on regulation of neutrophil biology by sex: We (I) describe the dynamics of neutrophils throughout human life in both females and males, (II) delineate sex-specific regulation of neutrophil phenotype and function, and (III) examine the significance of these differences in the susceptibility and outcomes of neutrophil-driven diseases.
    Keywords:  Granulopoiesis; Neutrophil function; Neutrophils; Sex hormones; Sexual dimorphism
    DOI:  https://doi.org/10.1093/jleuko/qiaf161
  3. Int J Mol Sci. 2025 Oct 29. pii: 10523. [Epub ahead of print]26(21):
      Although female (XX) and male (XY) placentas generally function the same, it is evident that there are sex-specific postnatal health outcomes following placental dysfunction and pregnancy complications. Although the underlying causes for these sex differences are unclear, it is postulated that differences in XX and XY placental function are involved due to sex chromosomes and/or sex steroids. Studies in breast and prostate cancer cells demonstrated a role for the citrullination enzyme peptidylarginine deiminase 2 (PAD2) in post-translational regulation of estrogen (ESR) and androgen receptor (AR) signaling. The goal of this study is to determine if PAD2 is present in mouse placentas and if XX versus XY differences exist in the relative level of PAD2. Fetuses and placentas were collected from three pregnant mice (C57BL6) at 14 days of gestation. Total RNA and protein were isolated from XX and XY placentas, and relative mRNA and protein were analyzed by real-time PCR and Western blot. AR and PAD2 levels were significantly higher in XY than in XX placentas. This study is the first to demonstrate XX and XY differences in PAD2 and AR in the placenta. It suggests a role for PAD2 regulation of androgen receptor signaling in the XY placenta.
    Keywords:  PAD2; androgen receptor; estrogen receptor 1; fetus; peptidylarginine deiminase; placenta
    DOI:  https://doi.org/10.3390/ijms262110523
  4. Elife. 2025 Nov 14. pii: e108276. [Epub ahead of print]14
      Trained immunity is reshaping our understanding of host defense by demonstrating that innate immune cells once thought to lack memory can be reprogrammed to mount heightened responses to subsequent challenges. Unlike tolerance, differentiation, or priming, trained immunity relies on epigenetic and metabolic rewiring of resident myeloid cells, particularly in mucosal barriers such as the skin, gut, and lungs, where these cells provide continuous protection against toxins and pathogens. Here, we review recent advances showing how an initial stimulus endows monocytes and macrophages with long-lasting functional changes that can be either protective or maladaptive upon re-exposure. We highlight therapeutic opportunities that harness trained immunity to boost vaccine efficacy and discuss strategies to modulate this program in cancer and hyper-inflammatory disorders. Finally, we propose new directions for enhancing or dampening trained immunity to promote human health.
    Keywords:  bone marrow; immunology; infection; infectious disease; inflammation; innate immune memory; innate immunity; lung; macrophage; microbiology; monocyte; trained immunity
    DOI:  https://doi.org/10.7554/eLife.108276
  5. Endocrinology. 2025 Nov 05. pii: bqaf161. [Epub ahead of print]
      Prenatal androgen excess (PNA), an etiologic factor for polycystic ovary syndrome (PCOS), is implicated in programming long-term reproductive deficits in females such as anovulation, subfertility, and hyperandrogenism. Impaired steroid hormone feedback is a key neuroendocrine feature suspected to underpin the development of reproductive dysfunction in both clinical PCOS and in PNA mice exposed to dihydrotestosterone during late gestation. PNA is suspected to act in the brain to programme the impaired sensitivity of the gonadotropin-releasing hormone (GnRH) neuronal network to progesterone negative feedback, centrally dysregulating the hypothalamic-pituitary-ovarian axis (HPO) controlling reproduction. To test the hypothesis that androgen-sensitive neurons mediate PNA programming, we generated PNA female mice with a neuron-specific deletion of androgen receptors (NeurARKO) using Cre-lox transgenics. Following confirmation of embryonic AR deletion, PNA NeurARKO females were reproductively phenotyped and assessed for changes in progesterone receptor (PR) expression in the brain. PNA-induced reproductive traits including delayed pubertal onset, acyclicity, altered ovarian morphology, and subfertility were not different between NeurARKO and wild-type (WT) mice. In contrast, downregulation of PR expression in PNA WT mice was protected against in PNA NeurARKO mice. Together, these findings suggest that while neuronal AR may contribute to PCOS-like impaired sensitivity to progesterone feedback, their deletion alone is insufficient to rescue reproductive dysfunction associated with PCOS.
    Keywords:  PCOS; androgen receptor; prenatal androgen; progesterone receptor; reproduction
    DOI:  https://doi.org/10.1210/endocr/bqaf161
  6. Cancers (Basel). 2025 Oct 23. pii: 3414. [Epub ahead of print]17(21):
      Several major cancer types exhibit significant sex dimorphism in incidence and survival. Whether and how sex as a biological factor impacts tumorigenesis, progression, and survival warrants full investigation, as such knowledge may lead to novel, precise prevention and treatment strategies. We reviewed epidemiological and molecular data on sex differences in cancers of the esophagus, bladder, head and neck, lung, liver, kidney, stomach, and skin melanoma, as well as the potential role of androgens and androgen receptor (AR) activity in these cancers. The potential molecular mechanisms are briefly discussed. Elevated testosterone (T) levels seemed to be associated with increased liver cancer and cutaneous melanoma incidences, and with reduced esophageal cancer risk. AR activity does not always correlate with T levels in tumorigenesis and progression. Higher AR expressions are associated with poorer survival in ESCC, whereas the role of AR in the survival of HNSCC and melanoma patients is inconsistent. The molecular impact of AR in liver cancer, kidney cancer, melanoma, and lung cancer is controversial. However, AR is likely to promote tumor growth and/or progression in esophagus, bladder, head and neck, and stomach cancers, and thus is associated with poor survival. Patients diagnosed with a tumor in this latter group could potentially benefit from therapeutic approaches targeting AR. Overall, the research on sex hormone androgens and AR in these cancers is limited. Further research is needed to determine a possible U-shaped relationship of T with cancer risk, and to decipher the role of testosterone and AR in some of these tumors to facilitate our understanding of sex dimorphism and to explore novel T/AR-based treatment options.
    Keywords:  androgen receptor; cancer; sex dimorphism; testosterone
    DOI:  https://doi.org/10.3390/cancers17213414
  7. Int J Mol Sci. 2025 Oct 25. pii: 10381. [Epub ahead of print]26(21):
      Ferroptosis is a novel kind of regulated cell death that occurs when redox equilibrium is disrupted, leading to iron-dependent lipid peroxidation. Ferroptosis is defined by the buildup of deleterious lipid hydroperoxides, the inactivation of glutathione peroxidase 4 (GPX4), and mitochondrial shrinkage, setting it apart from apoptosis and necrosis. The relevance of this route to human reproduction remains unknown, despite its thorough investigation in neurodegeneration and cancer. Recent studies demonstrate that the ovarian follicular milieu is especially susceptible to ferroptosis owing to its high content of polyunsaturated fatty acids, iron-dependent metabolism, and the generation of reactive oxygen species. Dysregulation of ferroptosis may result in infertility by affecting granulosa cell survival, oocyte maturation, and embryonic competence. Ferroptotic activity correlates with oxidative stress indicators identified in clinical diseases including polycystic ovary syndrome, reduced ovarian reserve, and insufficient responsiveness to ovarian stimulation. Potential indicators include GPX4 expression, decreased glutathione levels, and the accumulation of lipid reactive oxygen species in granulosa cells and follicular fluid. Melatonin, which boosts antioxidant defences, and ferrostatin-1, a prototype inhibitor of ferroptosis that lowers lipid peroxidation, are two early candidates for treatment. For future evaluations, these agents should be used with standardised FF biomarker panels. Significantly, vitamin E, coenzyme Q10, and small-molecule ferroptosis inhibitors have shown efficacy in halting ferroptosis in experimental settings. These approaches have shown protective benefits in alternative systems and may signify viable treatment options for assisted reproduction. This narrative review encapsulates ferroptosis inside the ovarian follicle, its influence on oocyte quality, and the implications for in vitro fertilization results.
    Keywords:  IVF outcomes; diminished ovarian reserve; endometriosis; follicular fluid; granulosa cells; lipid peroxidation; ovarian aging; oxidative stress; polycystic ovary syndrome; redox homeostasis
    DOI:  https://doi.org/10.3390/ijms262110381
  8. Vascul Pharmacol. 2025 Nov 08. pii: S1537-1891(25)00098-9. [Epub ahead of print] 107559
      Estrogen's role in cardiovascular health remains inconsistent. This study investigates the non-genomic vasorelaxant effects of 17β-estradiol, focusing on sex-specific differences in estrogen receptor (ER) subtypes (ERα, ERβ, GPER), endothelial function, and phosphoinositide 3-kinase (PI3K) signaling pathway. Ex vivo experiments using thoracic aorta from male and female wild-type mice assessed ER expression by immunofluorescence and vascular reactivity by wire myography. Vasorelaxant responses to 17β-estradiol and ER-selective agonists (PPT, DPN, G1) were examined. Some experiments were conducted in the presence of pharmacological inhibitors targeting endothelium-dependent relaxation pathways-L-NMMA, indomethacin, and apamin/charybdotoxin-as well as the PI3K pathway inhibitor LY294002 hydrochloride. In vitro, primary mouse aortic smooth muscle cells (MASMCs) were used to assess short-term dose-dependent effects of 17β-estradiol on myosin light chain (MLC) and Akt phosphorylation via western blot. ER subtypes were detected in both endothelial and vascular smooth muscle layers. 17β-estradiol induced vasorelaxation, with no significant sex differences. Inhibition of nitric oxide synthase (NOS) reduced vasorelaxation, with modest sex specific differences among ER subtypes. PI3K inhibition augmented this vasorelaxation regardless of endothelial integrity, exhibiting slight sex-specific variability across distinct ER subtypes. MASMCs showed a trend toward increased MLC phosphorylation with 17β-estradiol without significant Akt activation. In conclusion, estrogen induces NOS dependent vasorelaxation in mouse aorta, with no significant sex differences. However, subtle sex differences among ER subtypes were observed in the contribution of NOS and PI3K pathways. The PI3K pathway may attenuate vasorelaxant effect by promoting MLC phosphorylation in vascular smooth muscle, and its inhibition unmasks the vasorelaxant potential of estrogen.
    Keywords:  Aortic smooth muscle cells; Endothelium; Estrogen; LY294002; Nitric oxide; Non-genomic signaling; PI3K
    DOI:  https://doi.org/10.1016/j.vph.2025.107559
  9. Microbiol Spectr. 2025 Nov 12. e0179025
      Biological sex significantly influences susceptibility to tuberculosis (TB), with males typically experiencing a greater disease burden than females. However, the mechanisms underlying sex-specific immune regulation during TB pathogenesis remain incompletely understood. In this study, we examined sex-based differences in pulmonary immune cell composition, inflammatory signaling pathways, and macrophage transcriptomic responses in C57BL/6 mice infected with the Mycobacterium tuberculosis (Mtb) HN878 strain during the sub-acute stage of infection. Our results show that female mice had significantly lower pulmonary Mtb burdens and enhanced protein kinase B (AKT)-interferon gamma (IFN-γ) signaling, which is associated with autophagy, lysosomal activation, and effective intracellular bacterial clearance. In contrast, male mice exhibited higher bacterial loads and elevated IL-6 signaling, a pathway linked to exacerbated inflammation and impaired pathogen control. Histological analysis revealed greater lymphocytic aggregation in female lungs, despite comparable levels of pulmonary macrophages between sexes. Importantly, transcriptomic profiling of lung macrophages uncovered distinct sex-specific gene expression patterns, wherein female macrophages upregulated genes involved in tissue remodeling, phagocytosis, autophagy/lipophagy, and cell survival, whereas male macrophages showed enrichment of genes related to pro-inflammatory and adaptive immune responses. Notably, male macrophages expressed higher levels of Ighg1 (Immunoglobulin Heavy Constant Gamma 1), suggesting a potential sex-dependent modulation of humoral immunity during TB. Together, these findings demonstrate that biological sex shapes immune cell programming and host-pathogen interactions during TB, underscoring the importance of incorporating sex as a biological variable in TB research and therapeutic development.IMPORTANCEMen and women often respond differently to diseases like tuberculosis (TB), with men typically facing more severe illness, though the underlying reasons are unclear. To this end, our study investigated how male and female mice combat TB infection at a cellular and molecular level. We discovered that female mice controlled TB more effectively, as their immune systems activated specific pathways to break down and clear bacteria efficiently. Conversely, male mice showed higher bacterial loads and triggered more inflammatory, yet less effective, immune responses. Crucially, despite similar numbers of key immune cells (macrophages) in the lungs, their functional responses differed significantly by sex. These findings underscore that biological sex profoundly impacts the immune system's fight against TB, paving the way for more personalized treatments and improved outcomes for all.
    Keywords:  AKT-IFN-γ signaling; IL-6 signaling; Immunoglobulin Heavy Constant Gamma 1 (Ighg1); Mycobacterium tuberculosis; autophagy; host-pathogen interactions; macrophages; pulmonary infection; sex-specific immune regulation; transcriptomic profiling
    DOI:  https://doi.org/10.1128/spectrum.01790-25
  10. Front Immunol. 2025 ;16 1670112
      The omentum is a critical intraperitoneal organ essential for peritoneal homeostasis, yet detailed characterization of its cellular composition remains limited by the lack of validated markers. Here, we employed single-cell RNA sequencing to systematically define cellular heterogeneity in naive and activated mouse omentum from both sexes. Our analysis identified previously uncharacterized immune and stromal cell subsets, including three macrophage subtypes with activation-dependent gene expression patterns, implying specialized roles in inflammation and immune regulation. Comparative analysis revealed marked transcriptional differences between omental and peritoneal macrophages, underscoring tissue-specific microenvironments. Additionally, sexually dimorphic gene expression in omental stromal cells correlated with peritoneal macrophage polarization, indicating sex-specific regulatory mechanisms. Critically, macrophages from omentum of female mice with ovarian cancer metastases showed unique gene signatures associated with tumor migration and invasion. Collectively, we provide the first comprehensive atlas of omental cell populations stratified by sex and activation state, offering novel insights into peritoneal immunity and identifying potential therapeutic targets for inflammatory and metastatic diseases.
    Keywords:  inflammation; macrophages; omentum; peritoneum; sexually dimorphisms; single-cell RNA sequencing
    DOI:  https://doi.org/10.3389/fimmu.2025.1670112
  11. Eur J Immunol. 2025 Nov;55(11): e70083
      Recent studies add to our understanding of T cell residency in the human bone marrow (BM). In the May 2025 issue of EJI, Schneider Revueltas et al. demonstrate that CD69- CD4+ and CD8+ memory T cells, presumed to be recirculating, have a distinct transcriptional cluster profile and a distinct TCR repertoire from their blood counterparts. These findings are in line with CD69- memory T cells taking up residence in the BM, similar to their CD69+ counterparts. In parallel, Pulvirenti et al. identify a subset of CD69+EOMES+GzmK+ Tr1-like cells in the BM maintained by IL-15. Together, these studies refine our understanding of the BM as a heterogeneous immune niche and suggest a broader definition of resident cells within this tissue.
    Keywords:  T cells; bone marrow; tissue‐resident memory
    DOI:  https://doi.org/10.1002/eji.70083
  12. J Ovarian Res. 2025 Nov 12. 18(1): 255
       BACKGROUND: Polycystic ovary syndrome (PCOS) is a complex endocrine and metabolic disorder, and its diagnosis remains controversial due to heterogeneous phenotypes and varying diagnostic criteria. Insulin resistance and its metabolic consequences are central features of PCOS management. Metabolomics has increasingly been applied to elucidate the pathophysiology of complex disorders. In this study, we sought to determine which serum metabolites and metabolic pathways are differentially altered in women with PCOS compared with healthy controls, thereby addressing whether metabolomic profiling can reveal candidate biomarkers for early diagnosis and potential therapeutic targets.
    METHODS: Fifty patients diagnosed with PCOS and 50 healthy controls matched for age and body mass index (BMI) were included in the study. Blood samples were collected for metabolomic analysis and routine biochemical parameters. Metabolomic analysis was performed by UPLC-HRMS and data were processed using MZmine, TidyMass and MetaboAnalyst. Metabolite annotation was performed using databases such as HMDB, MassBank and MoNA.
    RESULTS: Metabolomic analysis revealed 49 compounds in the serum of PCOS patients, 39 of which were upregulated and 10 of which were downregulated. Compounds such as di(2-ethylhexyl) phthalate (DEHP), promethazine N-oxide, tetrahydromagnolol, 5-methyl-5-phenylhydantoin, valerenic acid, butylparaben, erucamide, DDAO, d-erythro-sphinganin-1-phosphate and 1-arachidoyl-2-hydroxy-sn-glycero-3-phosphocholine were significantly higher in the PCOS group. Compounds such as L-methyladenosine, cystine, glu-gln and 2,2'-methylene-bis(6-tert-butyl-4-methylphenol) were significantly lower. In the pathway analysis performed using KEGG database, sphygolipid metabolism, sphygolipid signaling pathway, neuroactive ligand-receptor interaction and phenylalanine metabolism were found to be the most affected pathways.
    CONCLUSION: This study demonstrates distinct alterations in lipid and amino acid metabolism in PCOS and highlights the accumulation of exogenous molecules, including endocrine disruptors, in patient serum. By integrating metabolomic profiling with clinical phenotyping, our findings provide novel insights into PCOS pathophysiology and suggest potential serum biomarkers that may support early diagnosis and personalized therapeutic approaches.
    Keywords:  Metabolomics analysis; Polycystic ovary syndrome; Serum metabolites; UPLC-HRMS
    DOI:  https://doi.org/10.1186/s13048-025-01842-9
  13. Arterioscler Thromb Vasc Biol. 2025 Nov 13.
      Innate immune cells can develop a long-lasting hyperresponsive phenotype by metabolic and epigenetic reprogramming after brief exposure to inflammatory stimuli. Several experimental studies convincingly demonstrated that this immunologic phenomenon, which is called trained immunity, can accelerate the development of atherosclerosis. In this brief review, we summarize current evidence that diets and specific dietary components can modulate trained immunity. In mice, intermittent high-fat diets can reprogram bone marrow myeloid progenitor cells, resulting in hyperinflammatory monocytes and neutrophils that aggravate atherosclerosis. Diet-induced obesity also leads to persistent proinflammatory epigenetic reprogramming of myeloid cells and adipocytes. Hyperglycemia and high-salt diets can also induce trained immunity in murine models. Recent intervention studies in Tanzania revealed that urban Western-style diets trigger systemic inflammation and immune activation, whereas a traditional plant-based heritage diet limits inflammation. Ex vivo studies suggest that this is caused, at least in part, by modulation of trained immunity. Various individual dietary components, such as the flavone apigenin and the polyphenol resveratrol, are able to prevent trained immunity in vitro. It is exciting to speculate how further molecular elucidation on the modulation of trained immunity by diets or isolated dietary components could help to prevent cardiovascular diseases.
    Keywords:  atherosclerosis; cardiovascular diseases; diet, high-fat; myeloid progenitor cells; trained immunity
    DOI:  https://doi.org/10.1161/ATVBAHA.125.322608
  14. Trends Mol Med. 2025 Nov 12. pii: S1471-4914(25)00256-4. [Epub ahead of print]
      Men have a higher incidence of specific types of cancer and neurodegenerative disease. Mounting evidence suggests that androgen receptor (AR)-mediated androgen signaling is a key determinant at the core of this sex discrepancy. Herein we review the role of androgens in disorders characterized by altered AR activity, focusing on transcriptional coregulators that shape receptor specificity. In particular, we highlight the roles of protein arginine methyltransferase 6 (PRMT6) and lysine-specific demethylase 1 (LSD1), enzymes associated with epigenetic repression, yet functioning as AR coactivators. By enhancing AR transcriptional output, PRMT6 and LSD1 contribute to malignant transformation and progression across multiple cell types. We further explore how these insights inform combinatorial therapeutic strategies targeting AR, PRMT6, and LSD1, with implications for both cancer and neurodegeneration.
    Keywords:  LSD1; PRMT6; androgens; cancer; epigenetic factors; neurodegenerative diseases
    DOI:  https://doi.org/10.1016/j.molmed.2025.10.005
  15. Radiol Case Rep. 2026 Jan;21(1): 329-332
      Morris syndrome, also known as Complete Androgen Insensitivity Syndrome (CAIS), is a rare genetic disorder of sex development characterized by a 46, XY karyotype with female external genitalia due to androgen receptor mutations. We present a case of a 15-year-old phenotypically female patient who presented with primary amenorrhea despite normal secondary sexual characteristics. Physical examination revealed a BMI of 14.81 kg/m2, developing breasts and sparse pubic hair consistent with Tanner stage 3, normal external female genitalia, and a blind-ended vaginal canal. Imaging studies demonstrated absence of uterus and ovaries, with 2 solid oval-shaped hypoechoic structures in the right inguinal region consistent with undescended testes, while the left inguinal region remained empty. Hormonal analysis revealed markedly elevated testosterone levels (643.6 ng/dL) within the male reference range and decreased estradiol levels (25.45 pg/mL). Cytogenetic analysis confirmed a 46, XY karyotype in all cells examined. This case highlights the importance of considering androgen insensitivity syndrome in phenotypically female patients presenting with primary amenorrhea, elevated testosterone levels, and absent uterine structures. Early diagnosis is crucial for appropriate management, including psychological support, hormone replacement therapy planning, and timely gonadectomy due to increased malignancy risk in undescended testes.
    Keywords:  46, XY karyotype; Complete androgen insensitivity syndrome (CAIS); Morris syndrome; Primary amenorrhea; Undescended testes
    DOI:  https://doi.org/10.1016/j.radcr.2025.09.075
  16. Biogerontology. 2025 Nov 14. 26(6): 204
      Homeostatic systems (nervous, immune, and endocrine) are crucial for maintaining health throughout life and, consequently, relevant for the rate of aging and the longevity achieved. In many species, male and female mammals show different lifespans, attributed to distinct redox states, but it is scarcely known whether sex differences in the functioning of these systems are involved. This study investigated, in an integrative view, sex differences in the nervous and immune systems of Swiss strain mice by analyzing behavior, immune function, and redox biomarkers across aging, to determine whether possible sex differences in homeostatic systems affect longevity. A longitudinal study was conducted on 20 female and male Swiss mice. At their young (2 mon), adult (7 mon), and old (18 mon) ages, subjects were subjected to a battery of behavioral tests, and peritoneal leukocytes were extracted to assess immune function and redox biomarkers. The natural deaths of animals were recorded for a longevity study. Our results indicate that sexual differences begin at a young age, and several are maintained until old age. Females, in general, show better behavior, immune function, and redox biomarkers, contributing to their higher longevity compared to males. The enhanced longevity in females may be attributable, in part, to the preservation of robust immune competence, with emphasis on innate immune functions and lower oxidative stress. The integration of behavioral and immunological profiles, together with redox biomarkers, underscores the critical importance of incorporating both sex as a biological variable in the design of aging-related research.
    Keywords:  Aging; Behavior; Immunology; Longevity; Redox state; Sex differences
    DOI:  https://doi.org/10.1007/s10522-025-10350-8
  17. Cell Rep Med. 2025 Nov 07. pii: S2666-3791(25)00522-1. [Epub ahead of print] 102449
      Long COVID (LC) manifests with sex-specific differences, particularly in those with myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). Our study reveals that female LC patients (LCF) with ME/CFS show a shift toward myelopoiesis, reduced lymphocytes, increased neutrophils/monocytes, and depleted regulatory T cells-suggesting persistent immune activation. Elevated CD71+ erythroid cells and disrupted erythropoiesis contribute to fatigue and tissue damage in LCF. Cytokine profiling indicates a stronger pro-inflammatory response in LCF compared to males (LCM), along with markers of gut barrier dysfunction. Hormonal analysis shows reduced testosterone in LCF and estradiol in LCM. Transcriptomic data reveal neuroinflammatory signatures in LCF, potentially explaining cognitive symptoms. We also identify biomarkers that distinguish LCF from LCM and correlate with sex-specific clinical symptoms. Overall, LC with ME/CFS is characterized by sex-specific immune, hormonal, and transcriptional alterations, with females exhibiting more severe inflammation. These insights underscore the need for sex-tailored interventions, including consideration of hormone replacement therapy.
    Keywords:  Reelin; artemin; chronic fatigue syndrome; cortisol; erythropoiesis; galectin-9; growth hormone; long COVID; sex-related differences; testosterone
    DOI:  https://doi.org/10.1016/j.xcrm.2025.102449
  18. Medicine (Baltimore). 2025 Nov 14. 104(46): e46099
      Polycystic ovary syndrome (PCOS) is one of the causes of endocrine disorders and infertility in women of childbearing age, which seriously affects women's health. The etiology of PCOS is still unknown, and metabolic abnormalities are the focus of current research. A large number of studies have shown that gut microbiota is related to metabolic abnormalities. This project intends to study the relationship between gut microbiota and PCOS and analyze the molecular mechanism of its impact on PCOS. The project is designed to conduct 16S rRNA sequencing and analysis using feces from patients with polycystic ovary syndrome. 48 samples were included in the study and divided into 3 groups (NOR, POS1, POS2), Venn diagram shows that there are 372 operational taxonomic units in total, and 96, 43 and 20 unique to the 3 groups respectively. PLS-DA analysis shows that there are significant differences among the 3 groups. Species abundance analysis shows that there are differences among the 3 groups at the level of class, family, genus, order, phylum and species. There are more lachnoanaerobcaulum and Klebsiella in POS1 group, and more enterococcaceae and enterococcu in POS2 group. The correlation analysis showed that Klebsiella and Lachnoanaerobaculum were positive associated with BMI, and LH level, respectively, and Enterococcus was negative correlated with the levels of LH and testosterone. This study can provide reference basis for clinical accurate and personalized dietary intervention and drug treatment of PCOS.
    Keywords:  16S rRNA gene sequencing; gut microbiota; polycystic ovary syndrome
    DOI:  https://doi.org/10.1097/MD.0000000000046099
  19. Immunol Rev. 2025 Nov;336(1): e70074
      Decades of experimental work have helped define the heterogeneity of the various cell types that compose the immune system. The different cell types arise from distinct hematopoietic stem and progenitor cells in a coordinated fashion during ontogeny, providing a set of diverse cells that contribute to host defense. Cells can also differentiate into different subsets in response to the cytokine and tissue environment, creating a level of cellular heterogeneity that helps direct the nature and magnitude of the immune response. Here we are discussing a variation whereby cellular heterogeneity arises due to the expression of X-linked immune genes that escape X chromosome inactivation, giving an advantage to a subset of cells more prone to respond to stimulation by external (pathogens) but also internal signals (i.e., mechanosensing). Interestingly, these inflammatory subsets are much more likely to be differentially enriched in patients with autoimmunity or inflammatory diseases which are well known to be predominant in females. We are using plasmacytoid dendritic cells (pDCs) as a model cell type, as these cells are a rare but critical subset of innate immune cells, with a rapid and massive capacity to produce type I IFNs (IFN-I) upon sensing of nucleic acids from pathogens, but also from the self, and these cells have been linked to the pathogenesis of many autoimmune diseases.
    Keywords:  TLRs; X chromosome inactivation; autoimmunity; plasmacytoid DCs
    DOI:  https://doi.org/10.1111/imr.70074
  20. Int J Mol Sci. 2025 Nov 06. pii: 10796. [Epub ahead of print]26(21):
      The human endometrium, previously considered a sterile environment, is now recognized as a low-biomass but biologically active microbial niche critical to reproductive health. Advances in sequencing technologies, particularly shotgun metagenomics, have provided unprecedented insights into the taxonomic and functional complexity of the endometrial microbiome. While 16S rRNA sequencing has delineated the distinction between Lactobacillus-dominant and non-dominant microbial communities, shotgun metagenomics has revealed additional diversity at the species and strain level, uncovering microbial signatures that remain undetected by amplicon-based approaches. Current evidence supports the association of Lactobacillus dominance with endometrial homeostasis and favorable reproductive outcomes. Dysbiosis, characterized by increased microbial diversity and enrichment of anaerobic taxa such as Gardnerella, Atopobium, Prevotella, and Streptococcus, is linked to chronic endometritis, implantation failure, and adverse IVF results. Beyond compositional differences, the endometrial microbiome interacts with the host through immunological, metabolic, and epigenetic mechanisms. These interactions modulate cytokine signaling, epithelial barrier integrity, and receptivity-associated gene expression, ultimately influencing embryo implantation. However, discrepancies between published studies reflect the lack of standardized protocols for sampling, DNA extraction, and bioinformatic analysis, as well as the inherent challenges of studying low-biomass environments. Factors such as geography, ethnicity, hormonal status, and antibiotic exposure further contribute to interindividual variability. Culturomics approaches complement sequencing by enabling the isolation of viable bacterial strains, offering perspectives for microbiome-based biotherapeutics. Emerging 3D endometrial models provide additional tools to dissect microbiome-host interactions under controlled conditions. Taken together, the growing body of data highlights the potential of endometrial microbiome profiling as a biomarker for reproductive success and as a target for personalized interventions. Future research should focus on integrating multi-omics approaches and functional analyses to establish causal relationships and translate findings into clinical practice. This review gives a new insight into current knowledge on the uterine microbiome and its impact on implantation success, analyzed through the lenses of microbiology, immunology, and oxidative stress.
    Keywords:  3D human endometrial culture models; Lactobacillus; antioxidant activity of lactobacilli; endometrial microbiome; immunological mechanisms; oxidative stress
    DOI:  https://doi.org/10.3390/ijms262110796
  21. J Assist Reprod Genet. 2025 Nov 15.
       PURPOSE: Transgender men (TGM) are individuals who identify as males but were assigned female sex at birth. Gender-affirming testosterone therapy (GATT) is used to induce virilization, and its effects on fertility are discussed. To provide TGM with complete fertility counseling, we synthesized data concerning the impact of GATT on ovarian reserve and oocyte quality.
    METHODS: We conducted a narrative review of published data on the ovarian and oocyte features induced by GATT in TGM.
    RESULTS: Data on ovarian morphology and histology showed a normal proportion of primordial, primary, pre-antral, and antral follicles, and a significantly higher number of atretic and cystic follicles. Additionally, ovaries exhibited diffuse ovarian stromal hyperplasia and a thicker ovarian cortex, tunica albuginea, and basal membrane. No impact on ovarian reserve was observed. Oocyte quality following ovarian stimulation did not appear to be impaired and led to the birth of healthy offspring.
    CONCLUSION: These findings may reassure transgender patients and their care providers that satisfactory reproductive outcomes are possible, even if GATT has already been initiated. However, further investigation is needed to assess the reversibility and functional impact of these changes on ovarian folliculogenesis, ART success rates, pregnancy, and child health.
    Keywords:  Anti-Müllerian hormone; Fertility; Gender-affirming testosterone therapy; Oocytes; Ovarian stimulation; Ovaries; Transgender men
    DOI:  https://doi.org/10.1007/s10815-025-03701-4
  22. BJU Int. 2025 Nov 14.
      
    Keywords:  androgen deprivation therapy; prostate cancer; quality of life; transdermal oestradiol; treatment decision‐making
    DOI:  https://doi.org/10.1111/bju.70088
  23. Cell Mol Biol (Noisy-le-grand). 2025 Nov 02. 71(10): 82-88
      Polycystic ovary syndrome (PCOS) is one of the most common endocrine diseases characterized by hyperandrogenemia and anovulation. The present study aimed to estimate heat shock protein 70 (HSP-70) in PCOS patients. This case-control study involved 90 females aged 15 to 45 years, divided into two groups: 45 controls and 45 PCOS patients. Levels of HSP-70, anti-Müllerian hormone (AMH), luteinizing hormone (LH), follicle-stimulating hormone (FSH), prolactin, and testosterone were measured using enzyme-linked immunosorbent assay (ELISA). The study revealed a significant increase (p ≤ 0.01) in HSP-70 and AMH levels in PCOS patients compared to controls, with mean ± SD values of 16.49 ± 2.79 and 2386.14 ± 530.09, respectively. Hormonal parameters such as LH, testosterone, and prolactin were elevated, whereas FSH was decreased in PCOS patients. The findings suggest that elevated HSP-70 plays a key role in the pathogenesis of PCOS.
    DOI:  https://doi.org/10.14715/cmb/2025.71.10.11
  24. Front Endocrinol (Lausanne). 2025 ;16 1652731
       Background: Polycystic ovary syndrome (PCOS) is a prevalent endocrine-metabolic disorder characterized by Insulin Resistance (IR), hyperandrogenism, and ovulatory dysfunction, with gut dysbiosis emerging as a key pathophysiological driver. Exercise, a non-pharmacological intervention, ameliorates PCOS symptoms, yet the molecular mechanisms linking exercise-induced gut microbiota remodeling to metabolic improvements remain elusive.
    Objective: This review synthesizes evidence on how exercise reshapes gut microbiota to reverse core PCOS pathologies through integrated molecular pathways.
    Results: Exercise enriches beneficial taxa (e.g., Faecalibacterium, Roseburia, Akkermansia muciniphila) and reduces pro-inflammatory pathogens (e.g., Proteobacteria), elevating short-chain fatty acids (SCFAs) and secondary bile acids (BAs) while suppressing lipopolysaccharide (LPS) translocation. We propose three core mechanisms:(1) SCFAs network reconstruction: Butyrate/propionate enhance gut barrier integrity (via ZO-1/Occludin), inhibit histone deacetylases (suppressing CYP17A1), activate GLP-1 secretion (FFAR3-dependent), and mitigate inflammation. (2) BA-FXR axis activation: Exercise increases secondary BAs (e.g., deoxycholic acid), activating hepatic FXR to inhibit gluconeogenesis (*PEPCK/G6Pase*) and upregulate androgen-clearance enzymes (*SULT2A1/CYP3A4*). (3) LPS-inflammation inhibition: Reduced LPS blunts TLR4/NF-κB signaling and NLRP3 inflammasome activation, resolving chronic inflammation. These axes converge to improve tissue-specific PCOS features: ovarian androgen synthesis (HDAC/NF-κB inhibition), hepatic IR (FXR/PI3K-Akt), and ovulatory function (AhR-mediated Treg/Th17 balance). Exercise modality differentially impacts PCOS subtypes-endurance training benefits IR-dominant phenotypes via SCFAs producers, while resistance training reduces inflammation in obese PCOS.
    Conclusion: Exercise remodels the gut microbiota-metabolism-immune network to reverse PCOS pathophysiology. Targeting microbial metabolites (e.g., butyrate, BAs) or their receptors (FXR, GPR43) offers novel therapeutic strategies. Future research must address PCOS heterogeneity and optimize exercise protocols for microbiota-directed precision medicine.
    Keywords:  IR; exercise; gut microbiota; molecular mechanisms; pcos
    DOI:  https://doi.org/10.3389/fendo.2025.1652731
  25. Nat Commun. 2025 Nov 11. 16(1): 9915
      CD16a triggers antibody-dependent cellular cytotoxicity (ADCC) and phagocytosis by natural killer (NK) cells and macrophages in anti-tumor immunity. However, CD16a undergoes cleavage by ADAM17 that dampens its anti-tumor immunity. We here develop a monoclonal antibody (F9H4) that binds to CD16a and inhibits its cleavage. F9H4 retains CD16a on the surface of NK cells and macrophages, without triggering or blocking CD16a. F9H4 also binds to and inhibits shedding of CD16b by neutrophils, and inhibits CD16a/b shedding by leukocytes in tumor samples from lung cancer patients. F9H4 promotes ADCC against lung cancer cells that are opsonized by cetuximab, an epidermal growth factor receptor antibody that engages CD16a. F9H4 synergizes with cetuximab to inhibit human lung adenocarcinoma development in immunodeficient mice reconstituted with human NK cells. F9H4 combining with cetuximab also inhibits murine lung carcinoma growth in Fc gamma receptor-humanized mice, and such effect is mediated by NK cells and macrophages. The efficacy of F9H4+cetuximab in lung cancer models is the proof-of-concept for this new approach that promotes anti-tumor functions of Fc-enabled antibodies.
    DOI:  https://doi.org/10.1038/s41467-025-64862-5
  26. Medicine (Baltimore). 2025 Nov 14. 104(46): e45831
      Endometrial cancer (EC) is a major reproductive system tumor and a common cancer in women. Polycystic ovary syndrome (PCOS) is one of the most prevalent female reproductive endocrine disorders. The incidence of EC is significantly higher in individuals with PCOS. This study seeks to elucidate the organic correlations and interaction mechanisms between the 2 diseases through series of exploration of key genes with a bioinformatics analysis. The PCOS sample data and the EC single-cell dataset were downloaded from the gene expression omnibus database. The EC sample data were retrieved from the cancer genome atlas public database. The random survival forest method was employed to identify key genes associated with the prognosis of PCOS and EC comorbidity. Corresponding analyses on functional pathway enrichment, regulatory networks, and immune micro-environment are conducted. From a bioinformatics perspective, the association and interaction mechanisms between PCOS and EC comorbidity were explored to provide research and development references for the prevention and control of PCOS and EC comorbidity. Five key genes associated with the prognosis of PCOS and EC comorbidity were identified using the random survival forest method. The identified genes are SYTL1, PARVG, ID4, IL1RN, and S100A9. The abnormal expression of these key genes has impacted various enrichment pathways, including the TGF-β signaling-pathway, motif regulatory networks (such as motif cisbp__M4556), and miRNA regulatory networks, which encompass genes such as ATM, BARD1 and BRCA1. Furthermore, these also influence the immune cell microenvironment, such as T cells regulatory. Collectively, these key genes play a significant role in the occurrence and progression of comorbidities through the pathways mentioned above. The dysregulation of key genes (SYTL1, PARVG, ID4, IL1RN, S100A9) in the context of PCOS-EC comorbidities, along with their associated enrichment pathways, including the TGF-β signaling-pathway and immune microenvironment, plays a significant role in the occurrence and progression of EC.
    Keywords:  bioinformatics; comorbidity; endometrial cancer; endometrial proliferative lesions; polycystic ovary syndrome
    DOI:  https://doi.org/10.1097/MD.0000000000045831
  27. Neoplasia. 2025 Nov 13. pii: S1476-5586(25)00133-2. [Epub ahead of print]71 101253
      Androgen receptor (AR) signaling remains a key driver of castration-resistant prostate cancer (CRPC), with AR splice variants like AR-V7 contributing to resistance against second-generation antiandrogens. Targeting the AR N-terminal domain (NTD) provides a strategy to bypass ligand-binding domain (LBD)-mediated resistance. We developed ITRI-148, a CRBN-based AR-NTD degrader incorporating a rigid piperidine-alkyne linker optimized for oral pharmacokinetics. ITRI-148 efficiently degrades full-length AR, AR-V7, and clinically relevant mutants (L702H, H875Y). It facilitates the recruitment of active AR species to CRBN in the nucleus, promoting their polyubiquitination and proteasomal degradation. In CRPC and enzalutamide-resistant models, ITRI-148 robustly suppresses AR signaling and inhibits cell viability, outperforming enzalutamide. With long-term treatment, it achieves sustained AR suppression without inducing compensatory AR-V7 upregulation or PSA re-expression. In vivo, ITRI-148 demonstrates potent antitumor efficacy in both castrated and hormone-intact CRPC models, supported by favorable pharmacokinetic properties, stability and safety profiles. These findings position ITRI-148 as a promising next-generation AR-targeting agent capable of degrading resistant AR variants and providing durable inhibition of AR signaling in advanced prostate cancer.
    Keywords:  AR, AR-V7; Drug resistance; PROTAC; Prostate cancer
    DOI:  https://doi.org/10.1016/j.neo.2025.101253
  28. Animal Model Exp Med. 2025 Nov 12.
      Robust preclinical models of transgender male (TGM) gender-affirming hormone therapy (GAHT) can inform clinicians of the isolated effects of GAHT; however existing models vary significantly in approach. We aimed to assess existing methodology and how it influences circulating sex-hormone levels in rodent models of TGM GAHT to provide recommendations of best practise. PubMed, Embase, and Scopus databases were systematically searched for studies that investigated GAHT in rodent models and were published from inception to the 1st of August 2024. Study characteristics and methodology were extracted and compared. Post-intervention circulating sex hormone concentrations were the primary outcome used to determine whether successful gender affirming hormone therapy had been achieved. Sixteen experimental rodent studies were included. Studies were performed on mice (n = 11) and rats (n = 5). Subcutaneous (SC) pellets and SC silastic implants were featured in some studies but weekly SC injections of testosterone enanthate was the preferred method. Sesame oil was the preferred solvent for injected testosterone formulations. Weekly doses of ~450 μg (mice) and ~420-900 μg (rats) consistently induced the testosterone levels of the male counterpart. Similarly, 10 mg of unesterified testosterone in a SC silastic implant in mice or 10 mg/100 g in rats were also successful methods. Most studies administered hormones for 6-8 weeks before performing post-treatment assessments. This review demonstrates that methods largely varied across studies and successfully identifies the effective methodological approaches that improve the reproducibility and accuracy of preclinical models. Representing an integral step forward to bridging gaps in preclinical transgender healthcare research.
    Keywords:  animal model; endocrinology; hormone therapy; methodology; transgender
    DOI:  https://doi.org/10.1002/ame2.70088
  29. Br J Dermatol. 2025 Nov 13. pii: ljaf451. [Epub ahead of print]
      Atopic dermatitis (AD) is a common chronic inflammatory skin disease with diverse clinical and histological features. While primarily immune-mediated, genetic studies have also highlighted the role of epithelium-expressed gene abnormalities (e.g., filaggrin mutations) as a key factor. The approaches to treat AD are multifaceted, involving barrier restoration, local anti-inflammatory treatment, and, if needed, systemic immunosuppressive therapy. Genetic variations in the stratum corneum and the immune system are linked to an unbalance between the host and its microbiota, known as dysbiosis. An impaired skin barrier and immune responses can alter the microbial composition, while the skin microbiota itself can influence skin immunity and barrier formation. A hallmark of AD is increased bacterial colonization with Staphylococcus aureus (S. aureus), which is found on lesional skin in over 90% of patients. It contributes to disease severity driving further breakdown of the skin barrier and immune stimulation. The most common treatment for S. aureus infections in AD is topical or systemic antibiotic administration. While these treatments are typically reserved for active infections, they are sometimes prescribed to AD patients without clear skin infection. However, these treatments can disrupt commensal skin and gut microbiota, which play a critical role in maintaining skin and gut health. In this review we describe various therapies targeting the skin microbiome to reduce infection and inflammation in AD, including transplantation of microbiota, and the use of prebiotics, probiotics, and postbiotics. In addition, we provide a perspective to engineer and to harness bacteria of the skin microbiome as next-generation probiotics, also known as engineered live biotherapeutic products (eLBPs), using synthetic biology to create strains that can sense skin signals, such as immune signals, and environmental factors, and produce therapeutic treatments for AD on demand.
    DOI:  https://doi.org/10.1093/bjd/ljaf451
  30. Front Dent Med. 2025 ;6 1653315
       Background: Epidemiologic assessment of periodontitis prevalence and extent demonstrates age, sex, and race/ethnicity effects. However, the biological sources of these observations regarding sex differences with an elevated incidence in males remain unclear.
    Methods: This study used a model of experimental ligature-induced periodontitis in young nonhuman primates (Macaca mulatta) to evaluate gingival transcriptomic differences stratified based on the sex of the animal. The animals represent humans aged 10-25 years of age, with gingival tissue samples obtained at baseline, 0.5 months (initiation), and 1 and 3 months (progression). Microarray analysis was used to quantify gene expression profiles in the gingival tissues.
    Results: The results demonstrated clear gene expression differences in healthy (baseline) tissues between the sexes, with elevations in females associated with immune responses and elevated gene expression in males related to tissue structural genes. With disease initiation, fewer genes differed between the sexes, although a pattern of a greater number of unique gene expression changes was observed in females at late progression. Overexpressed biological processes showed tissue structural/functional genes at initiation, with host response pathways altered during disease progression.
    Conclusion: These findings support that this model can be used to explore processes that contribute to sex as a biological variable in periodontitis.
    Keywords:  aging; nonhuman primate; periodontitis; sex; transcriptome
    DOI:  https://doi.org/10.3389/fdmed.2025.1653315
  31. J Vis Exp. 2025 Oct 24.
      Human ovary follicular contents, including follicular fluid (FF), oocytes, cumulus cells, and somatic cells, offer a powerful yet underutilized opportunity to study ovarian physiology. Assisted reproductive technologies (ART) routinely generate these biological byproducts, which are often discarded after mature oocytes are used for clinical care. However, they provide valuable insights into fertility, reproductive aging, and ovarian dysfunction. Here, we present a standardized protocol for the systematic collection and downstream processing of follicular components from women undergoing oocyte retrieval. Fully expanded cumulus-oocyte complexes (COCs) are microdissected to isolate cumulus cells without enzymatic denudation, preserving transcriptomic integrity. FF is centrifuged, with the supernatant analyzed for cytokines, hormones, metabolites, and environmental toxicants, while the pellet is cryopreserved and processed via density gradient centrifugation to isolate viable granulosa cells. A stepwise protocol enables the preparation of single-cell suspensions from fresh FF for single-cell RNA sequencing (scRNA-Seq), including enzymatic digestion, serial filtration, and red blood cell lysis, and yields >65% viable cells representing a diverse composition, including granulosa cells, macrophages, T cells, smooth muscle-like cells, and antigen-presenting cells. In addition, immature germinal vesicle (GV) oocytes can be matured in vitro, with 56% reaching the MII stage within 24 h. Together, these protocols yield high-quality RNA, enriched granulosa cell populations, and complex single-cell datasets that capture the heterogeneity of the ovarian microenvironment. They provide a framework to leverage discarded reproductive tissues for translational research in human fertility and reproductive biology.
    DOI:  https://doi.org/10.3791/69122
  32. J Endocrinol. 2025 Nov 11. pii: JOE-25-0315. [Epub ahead of print]
      Prostate cancer is a leading cause of death. As a hormone-driven cancer, prostate cancer is often treated with drugs (luteinizing hormone receptor agonists; LHRHa) or surgical approaches (orchidectomy; ORX) with the goal of reducing androgens. These approaches cause side effects like bone loss. It is unclear if the side effects of these approaches are due to loss of androgens or loss of estrogens, as these approaches reduce both. We seek to evaluate if LHRHa and ORX have equivalent effects on bone, if the bone loss can be ameliorated by estrogen supplementation, and if estrogen supplementation alone is sufficient to improve bone mass while reducing androgen production. Herein, we evaluated bone microarchitecture, mechanical properties and the cellular mechanism of LHRHa with subsequent hormone addback on bone. We find that LHRHa negatively affects bone microarchitecture but has more mild effects on bone than ORX. Estrogen supplementation - but not androgen supplementation - improves bone mass and strength in mice treated with LHRHa. Estrogen supplementation alone is also sufficient to improve bone mass and strength while also reducing androgen production. However, estrogen supplementation also increases osteoblast and osteoclast activity, which may promote prostate cancer metastasis in bone. Future studies should evaluate estrogen as a modulator of the metastatic niche.
    Keywords:  Androgen; Cancer; Estrogen; Prostate; Skeletal Biology
    DOI:  https://doi.org/10.1530/JOE-25-0315
  33. Cell Death Dis. 2025 Nov 10. 16(1): 817
      Thioredoxin-Interacting Protein (TXNIP) is an arrestin at the crossroad of redox and glycolytic metabolisms. Prostate cancer (PCa) exhibits a unique metabolic profile due to the glycolytic nature of healthy prostate tissue. We hypothesize that TXNIP plays a pivotal role in the progression of PCa to castration-resistant prostate cancer (CRPC), an incurable stage of the disease characterized by profound metabolic reprogramming and independence from androgens. Only a subset of patients progresses to CRPC, and current stratification tools lack robust biomarkers. TXNIP expression is directly suppressed by androgens and diminishes during tumor initiation and progression, as demonstrated in both human samples and a prostate adenocarcinoma mouse model (TRAMP). TXNIP regulates glucose metabolism by sequestering the glucose transporter GLUT1 away from the membrane, shifting metabolism from glycolysis to glutaminolysis. Nuclear-localized TXNIP induces cell cycle arrest through the upregulation of p27kip1 which is downregulated together with TXNIP in CRPC. The response to androgen deprivation therapy (ADT) strongly depends on TXNIP expression. In the murine model, TXNIP levels were significantly higher in ADT responders compared to non-responders. Furthermore, TRAMP-Txnip-/- prostate tumors exhibited a poorer response to ADT, with increased Ki67 and enhanced viability. In clinical samples, all patients on relapse showed low levels of TXNIP and progressed to CRPC. Our findings identify TXNIP as a critical regulator of cell cycle and glucose metabolism in PCa and emphasize for the first time its essential role in mediating therapeutic responses to ADT.
    DOI:  https://doi.org/10.1038/s41419-025-08128-4
  34. Nat Commun. 2025 Nov 11. 16(1): 9918
      In mammals, X-linked dosage compensation involves X-chromosome inactivation to balance X chromosome dosage between males and females, and hyperactivation of the remaining X-chromosome (Xa-hyperactivation) to achieve X-autosome balance in both sexes. Studies of both processes have largely focused on coding genes and have not accounted for transposable elements which comprise 50% of the X-chromosome with numerous epigenetic functions. Here we develop a new bioinformatic pipeline tailored to repetitive elements with capability for allelic discrimination. We then apply the pipeline to our recent So-Smart-Seq analysis of single embryos to comprehensively interrogate whether X-linked transposable elements are subject to either X-chromosome inactivation or Xa-hyperactivation. We observe significant differences in repeat silencing in parentally driven "imprinted" versus zygotically driven "random" X-chromosome inactivation. Chromosomal positioning, genetic background and evolutionary age impact their silencing. In contrast, transposable elements do not undergo Xa-hyperactivation. Evolutionary and functional implications are discussed.
    DOI:  https://doi.org/10.1038/s41467-025-64865-2