1
|
Halloran KM, Saadat N, Pallas B, Vyas AK, Padmanabhan V. Exploratory analysis of differences at the transcriptional interface between the maternal and fetal compartments of the sheep placenta and potential influence of fetal sex. Mol Cell Endocrinol 2025; 603:112546. [PMID: 40222550 DOI: 10.1016/j.mce.2025.112546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 04/10/2025] [Accepted: 04/11/2025] [Indexed: 04/15/2025]
Abstract
An understanding of the inner workings of the placenta is imperative to elucidate how the maternal and fetal compartments coordinate to mediate fetal development. The two compartments can be separated and studied before term in sheep, a feat not possible in humans, thus providing a valuable translational model. This study investigated differential expression of gene signaling networks in the maternal and fetal compartments of the placenta and explored the potential influence of fetal sex. On approximately gestational day 120 (term: 147 days), ewes were euthanized and fetuses removed and sexed. Placentomes [n = 5 male, n = 3 female] were collected, and caruncles (maternal) and cotyledons (fetal) were separated and sequenced to assess RNA expression. Analysis revealed 2627 differentially expressed genes (FDR<0.01, abslog2FC ≥ 2) contributing to key transcriptional differences between maternal and fetal compartments, which suggested that the maternal compartment drives extracellular signaling at the interface whereas the fetal compartment controls internal mechanisms crucial for fetal-placental development. X-chromosome inactivation equalized expression of a vast majority of X-linked genes in the fetal compartment. Additionally, the female placenta had more fine-tuned regulation of key pathways for fetal-placental development, such as DNA replication, mRNA surveillance, and RNA transport, compared to males, which had enrichment of metabolic pathways including TCA cycle and galactose metabolism. These findings, in addition to supporting differences in expression in the maternal and fetal placental compartments and the possible influence of fetal sex, offer a transcriptional platform to compare placental perturbations that occur at the maternal-fetal interface that contribute to adverse pregnancy outcomes.
Collapse
Affiliation(s)
| | - Nadia Saadat
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - Brooke Pallas
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Arpita K Vyas
- Department of Pediatrics, Washington University St. Louis, MO, USA
| | | |
Collapse
|
2
|
Schmitz EG, Griffith M, Griffith OL, Cooper MA. Identifying genetic errors of immunity due to mosaicism. J Exp Med 2025; 222:e20241045. [PMID: 40232243 PMCID: PMC11998702 DOI: 10.1084/jem.20241045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 02/24/2025] [Accepted: 03/24/2025] [Indexed: 04/16/2025] Open
Abstract
Inborn errors of immunity are monogenic disorders of the immune system that lead to immune deficiency and/or dysregulation in patients. Identification of precise genetic causes of disease aids diagnosis and advances our understanding of the human immune system; however, a significant portion of patients lack a molecular diagnosis. Somatic mosaicism, genetic changes in a subset of cells, is emerging as an important mechanism of immune disease in both young and older patients. Here, we review the current landscape of somatic genetic errors of immunity and methods for the detection and validation of somatic variants.
Collapse
Affiliation(s)
- Elizabeth G. Schmitz
- Division of Rheumatology/Immunology, Department of Pediatrics, Washington University in St. Louis, St. Louis, MO, USA
| | - Malachi Griffith
- Division of Oncology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Obi L. Griffith
- Division of Oncology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Megan A. Cooper
- Division of Rheumatology/Immunology, Department of Pediatrics, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
3
|
Adams DM, Rayner JG, Hex SBSW, Wilkinson GS. DNA Methylation Dynamics Reflect Sex and Status Differences in Mortality Rates in a Polygynous Bat. Mol Ecol 2025; 34:e17745. [PMID: 40119542 PMCID: PMC12010472 DOI: 10.1111/mec.17745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 02/21/2025] [Accepted: 03/10/2025] [Indexed: 03/24/2025]
Abstract
Males of polygynous mammals often do not live as long as females and, in some cases, exhibit evidence of earlier senescence. Patterns of DNA methylation (DNAm) have recently been used to predict chronological age in mammals. Whether DNAm also changes as a consequence of survival and senescence is largely untested in wild animals. In this study, we estimate mortality rates using recaptures of 2700 greater spear-nosed bats, Phyllostomus hastatus, over 34 years and DNAm profiled for over 300 adult bats. In this species, one male typically controls mating access to a group of unrelated females. Bayesian analysis reveals that mortality risk in males is 1.8 times that of females, and comparison of age-associated differences in DNAm indicates that DNAm changes 1.4 times faster in males than females. Therefore, even though the age of either sex is predicted by a common set of sites, the methylome of males is more dynamic than that of females. Sites associated with sex differences in the rate of DNAm change are sensitive to androgens and enriched on the X chromosome. Sites that exhibit hypermethylation are enriched in promoters of genes involved in the regulation of metabolic processes. Unexpectedly, subordinate males have higher mortality rates than reproductively dominant males and exhibit faster DNAm change than dominants at dozens of sites. Our results reveal that differences in mortality associated with sex and social status are reflected by changes in DNA methylation, providing novel insights into mechanisms of aging and mortality in this and likely other wild animal populations.
Collapse
Affiliation(s)
| | - Jack G. Rayner
- Department of BiologyUniversity of MarylandCollege ParkMarylandUSA
| | | | | |
Collapse
|
4
|
Peckham H, Radziszewska A, Sikora J, de Gruijter NM, Restuadi R, Kartawinata M, Martin-Gutierrez L, Robinson GA, Deakin CT, Wedderburn LR, Jury EC, Butler G, Chambers ES, Rosser EC, Ciurtin C. Estrogen influences class-switched memory B cell frequency only in humans with two X chromosomes. J Exp Med 2025; 222:e20241253. [PMID: 40049222 PMCID: PMC11893172 DOI: 10.1084/jem.20241253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 12/04/2024] [Accepted: 01/17/2025] [Indexed: 03/12/2025] Open
Abstract
Sex differences in immunity are well-documented, though mechanisms underpinning these differences remain ill-defined. Here, in a human-only ex vivo study, we demonstrate that postpubertal cisgender females have higher levels of CD19+CD27+IgD- class-switched memory B cells compared with age-matched cisgender males. This increase is only observed after puberty and before menopause, suggesting a strong influence for sex hormones. Accordingly, B cells express high levels of estrogen receptor 2 (ESR2), and class-switch-regulating genes are enriched for ESR2-binding sites. In a gender-diverse cohort, blockade of natal estrogen in transgender males (XX karyotype) reduced class-switched memory B cell frequency, while gender-affirming estradiol treatment in transgender females (XY karyotype) did not increase these levels. In postmenopausal cis-females, class-switched memory B cells were increased in those taking hormone replacement therapy (HRT) compared with those who were not. These data demonstrate that sex hormones and chromosomes work in tandem to impact immune responses, with estrogen only influencing the frequency of class-switched memory B cells in individuals with an XX chromosomal background.
Collapse
Affiliation(s)
- Hannah Peckham
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH and GOSH, London, UK
- Centre for Rheumatology, University College London, London, UK
- Infection, Immunity and Inflammation Research and Teaching Department – UCL Great Ormond Street Institute of Child Health, London, UK
| | - Anna Radziszewska
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH and GOSH, London, UK
- Centre for Rheumatology, University College London, London, UK
| | - Justyna Sikora
- Centre for Immunobiology, Blizard Institute, Queen Mary University of London, London, UK
| | - Nina M. de Gruijter
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH and GOSH, London, UK
- Centre for Rheumatology, University College London, London, UK
| | - Restuadi Restuadi
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH and GOSH, London, UK
- Infection, Immunity and Inflammation Research and Teaching Department – UCL Great Ormond Street Institute of Child Health, London, UK
| | - Melissa Kartawinata
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH and GOSH, London, UK
- Infection, Immunity and Inflammation Research and Teaching Department – UCL Great Ormond Street Institute of Child Health, London, UK
| | - Lucia Martin-Gutierrez
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH and GOSH, London, UK
- Centre for Rheumatology, University College London, London, UK
| | - George A. Robinson
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH and GOSH, London, UK
- Centre for Rheumatology, University College London, London, UK
| | - Claire T. Deakin
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH and GOSH, London, UK
- Infection, Immunity and Inflammation Research and Teaching Department – UCL Great Ormond Street Institute of Child Health, London, UK
- NIHR Biomedical Research Centre at Great Ormond Street Hospital, London, UK
- School of Population Health, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
| | - Lucy R. Wedderburn
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH and GOSH, London, UK
- Infection, Immunity and Inflammation Research and Teaching Department – UCL Great Ormond Street Institute of Child Health, London, UK
- NIHR Biomedical Research Centre at Great Ormond Street Hospital, London, UK
| | - Elizabeth C. Jury
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH and GOSH, London, UK
- Centre for Rheumatology, University College London, London, UK
| | - Gary Butler
- Infection, Immunity and Inflammation Research and Teaching Department – UCL Great Ormond Street Institute of Child Health, London, UK
- University College London Hospital, London, UK
| | - Emma S. Chambers
- Centre for Immunobiology, Blizard Institute, Queen Mary University of London, London, UK
| | - Elizabeth C. Rosser
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH and GOSH, London, UK
- Centre for Rheumatology, University College London, London, UK
| | - Coziana Ciurtin
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH and GOSH, London, UK
- Centre for Rheumatology, University College London, London, UK
- University College London Hospital, London, UK
| |
Collapse
|
5
|
Marinelli A, Dragonieri S, Portacci A, Quaranta VN, Carpagnano GE. Reconsidering Gender in Asthma: Is It All About Sex? A Perspective Review. J Clin Med 2025; 14:2506. [PMID: 40217954 PMCID: PMC11989258 DOI: 10.3390/jcm14072506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 03/28/2025] [Accepted: 04/02/2025] [Indexed: 04/14/2025] Open
Abstract
Asthma is a prevalent chronic condition, affecting an estimated 260 million people worldwide, according to the 2021 Global Burden of Disease Study. This condition significantly impacts individuals of all ages. One notable finding is that asthma prevalence among adults was higher in females than males. Recent evidence suggests that these disparities in asthma prevalence and outcomes are likely due to complex interactions among hormonal, anatomical, and environmental factors, coupled with societal and behavioral influences. The interchangeable use of the terms "sex" and "gender" in the scientific literature is frequently inconsistent. Biological sex is defined by anatomical and physiological characteristics determined by genetics; "gender", on the other hand, is a more complex construct and a universally accepted definition is still lacking. This lack of clarity, coupled with potential knowledge gaps, misunderstandings, or the inherent difficulty in differentiating sex- and gender-related effects, often leads to the terms being poorly defined or used interchangeably. Such imprecise usage hinders accurate data interpretation and research progress. This paper provides a perspective review synthesizing current knowledge regarding the influence of sex and gender on asthma, specifically focusing on their impact on disease pathogenesis, clinical presentation, severity, and management strategies.
Collapse
Affiliation(s)
| | - Silvano Dragonieri
- Department of Respiratory Diseases, University of Bari, 70121 Bari, Italy; (A.M.); (A.P.); (V.N.Q.); (G.E.C.)
| | | | | | | |
Collapse
|
6
|
Perry RN, Lenert G, Benavente ED, Ma A, Barbera N, Mokry M, de Kleijn DPV, de Winther MPJ, Mayr M, Björkegren JLM, den Ruijter HM, Civelek M. Female-biased vascular smooth muscle cell gene regulatory networks predict MYH9 as a key regulator of fibrous plaque phenotype. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.28.645955. [PMID: 40236025 PMCID: PMC11996327 DOI: 10.1101/2025.03.28.645955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Atherosclerosis, a chronic inflammatory condition driving coronary artery disease (CAD), manifests in two primary plaque types: unstable atheromatous plaques and stable fibrous plaques. While significant research has focused on atheromatous plaques, recent studies emphasize the growing importance of fibrous plaques, particularly in females under 50 years of age, where erosion on fibrous plaques significantly contributes to coronary thrombosis. The molecular mechanisms underlying sex differences in atherosclerotic plaque characteristics, including vascular smooth muscle cell (VSMC) contributions, remain understudied. Therefore, we utilized sex-specific gene regulatory networks (GRNs) derived from VSMC gene expression data from 119 male and 32 female heart transplant donors to identify molecular drivers of fibrous plaques. GRN analysis revealed two female-biased networks in VSMC, GRN floralwhite and GRN yellowgreen , enriched for inflammatory signaling and actin remodeling pathways, respectively. Single-cell RNA sequencing of carotid plaques from female and male patients confirmed the sex specificity of these networks in VSMCs. Further sub cellular phenotyping of the single-cell RNA sequencing revealed a sex-specific gene expression signature within GRN yellowgreen for VSMCs enriched for contractile and vasculature development pathways. Bayesian network modeling of the GRN yellowgreen identified MYH9 as a key driver gene. Indeed, elevated MYH9 protein expression in atherosclerotic plaques was associated with higher smooth muscle cell content and lower lipid content in female plaques, suggesting its involvement in fibrous plaque formation. Further proteomic analysis confirmed MYH9's upregulation in female fibrous plaques only and its correlation with stable plaque features. These findings provide novel insights into sex-specific molecular mechanisms regulating fibrous plaque formation.
Collapse
|
7
|
Lovell CD, Anguera MC. More X's, more problems: how contributions from the X chromosomes enhance female predisposition for autoimmunity. Curr Opin Immunol 2025; 93:102543. [PMID: 40020257 PMCID: PMC11909602 DOI: 10.1016/j.coi.2025.102543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/17/2025] [Accepted: 02/22/2025] [Indexed: 03/08/2025]
Abstract
Many autoimmune diseases exhibit a strong female bias. While sex hormones may influence sex bias in disease, recent studies suggest that the X chromosome itself directly contributes to female-biased susceptibility to autoimmunity. Females with two X chromosomes utilize X Chromosome Inactivation (XCI) to silence gene expression from one X chromosome, equalizing expression between the sexes. The X chromosome is highly enriched with immune-related genes, and recent work indicates that the fidelity of XCI maintenance in lymphocytes from female systemic lupus erythematosus patients is compromised, suggesting that aberrant X-linked gene expression contributes to autoimmune phenotypes. XCI is initiated and maintained by the long noncoding RNA XIST/Xist through its interactions with the inactive X chromosome and numerous interacting proteins, and recent studies also implicate XIST/Xist RNA in driving endosomal Toll-like receptor signaling and XIST/Xist RNA-protein complexes in serving as a source of autoantigens to respectively drive autoimmunity. Here, we will review these three distinct pathways that underscore the significance of X-linked genetics for understanding the origins of the female bias in autoimmune disease.
Collapse
Affiliation(s)
- Claudia D Lovell
- Department of Biomedical Science, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Montserrat C Anguera
- Department of Biomedical Science, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
8
|
He W, Luo Q, Zhao J, Wang M, Zhao A, Feng L, Reda A, Lindgren E, Stukenborg J, Chen J, Deng Q. X-Linked Gene Dosage and SOX2 Act as Key Roadblocks for Human Germ Cell Specification in Klinefelter Syndrome. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410533. [PMID: 39996497 PMCID: PMC12005746 DOI: 10.1002/advs.202410533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 02/03/2025] [Indexed: 02/26/2025]
Abstract
Klinefelter syndrome (KS), characterized by the presence of at least one extra X-chromosome, is a common cause of male infertility. However, the mechanism underlying the failure of germline specification is not well studied. Intriguingly, the differentiation efficiency of female human pluripotent stem cells (hPSCs) is often lower than that of male. This study investigates how X-linked gene dosage affects human primordial germ cell-like cells (hPGCLCs) specification in both healthy and diseased conditions. This work reveals that X-linked genes play a multifaceted role against the fate competency to hPGCLCs, with escape genes IGSF1 and CHRDL1 inhibiting the TGF-beta/Activin A and BMP pathways, respectively. Notably, this work identifies a previously unrecognized role of SOX2, upregulated by the escape gene USP9X, elucidating a species-specific function in the mammalian germline. The USP9X-SOX2 regulatory axis profoundly influenced cellular metabolism, mitochondrial morphology, and progenitor competence in hPGCLCs specification. Furthermore, the inability to downregulate SOX2 and upregulate SOX17 in response to BMP signaling impedes downstream gene activation due to motif binding competition. These findings shed novel insights into the human germline specification by elucidating the divergent roles of SOX2 versus SOX17 in mammals, influenced by X-linked gene dosage effects. These results offer potential applications for improving the induction efficiency of hPGCLCs, facilitating disease mechanistic studies.
Collapse
Affiliation(s)
- Wenteng He
- Department of Physiology and PharmacologyKarolinska InstitutetStockholm171 77Sweden
| | - Qing Luo
- Department of Physiology and PharmacologyKarolinska InstitutetStockholm171 77Sweden
| | - Jian Zhao
- Department of Physiology and PharmacologyKarolinska InstitutetStockholm171 77Sweden
- Department of Oncology‐PathologyKarolinska InstitutetStockholm171 77Sweden
| | - Mengting Wang
- Clinical and Translational Research Center of Shanghai First Maternity and Infant HospitalShanghai Key Laboratory of Signaling and Disease ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Allan Zhao
- Department of Physiology and PharmacologyKarolinska InstitutetStockholm171 77Sweden
| | - Luohua Feng
- Department of Physiology and PharmacologyKarolinska InstitutetStockholm171 77Sweden
| | - Ahmed Reda
- Department of Physiology and PharmacologyKarolinska InstitutetStockholm171 77Sweden
| | - Eva Lindgren
- Department of Physiology and PharmacologyKarolinska InstitutetStockholm171 77Sweden
| | - Jan‐Bernd Stukenborg
- NORDFERTIL Research Lab StockholmChildhood Cancer Research UnitDepartment of Women's and Children's HealthKarolinska InstitutetKarolinska University HospitalStockholm17 165Sweden
| | - Jiayu Chen
- Clinical and Translational Research Center of Shanghai First Maternity and Infant HospitalShanghai Key Laboratory of Signaling and Disease ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
- Frontier Science Center for Stem Cell ResearchTongji UniversityShanghai200092China
| | - Qiaolin Deng
- Department of Physiology and PharmacologyKarolinska InstitutetStockholm171 77Sweden
- Department of Molecular Biosciences, The Wenner‐Gren InstituteStockholm UnviersityStockholm11418Sweden
| |
Collapse
|
9
|
Khawaja RR, Martín-Segura A, Santiago-Fernández O, Sereda R, Lindenau K, McCabe M, Macho-González A, Jafari M, Scrivo A, Gomez-Sintes R, Chavda B, Saez-Ibanez AR, Tasset I, Arias E, Xie X, Kim M, Kaushik S, Cuervo AM. Sex-specific and cell-type-specific changes in chaperone-mediated autophagy across tissues during aging. NATURE AGING 2025; 5:691-708. [PMID: 39910244 PMCID: PMC12003181 DOI: 10.1038/s43587-024-00799-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 12/18/2024] [Indexed: 02/07/2025]
Abstract
Aging leads to progressive decline in organ and tissue integrity and function, partly due to loss of proteostasis and autophagy malfunctioning. A decrease with age in chaperone-mediated autophagy (CMA), a selective type of lysosomal degradation, has been reported in various organs and cells from rodents and humans. Disruption of CMA recapitulates features of aging, whereas activating CMA in mice protects against age-related diseases such as Alzheimer's, retinal degeneration and/or atherosclerosis. However, sex-specific and cell-type-specific differences in CMA with aging remain unexplored. Here, using CMA reporter mice and single-cell transcriptomic data, we report that most organs and cell types show CMA decline with age, with males exhibiting a greater decline with aging. Reduced CMA is often associated with fewer lysosomes competent for CMA. Transcriptional downregulation of CMA genes may further contribute to CMA decline, especially in males. These findings suggest that CMA differences may influence organ vulnerability to age-related degeneration.
Collapse
Affiliation(s)
- Rabia R Khawaja
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA.
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, USA.
| | - Adrián Martín-Segura
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, USA
- IMDEA Food, Madrid, Spain
| | - Olaya Santiago-Fernández
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Rebecca Sereda
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Kristen Lindenau
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Mericka McCabe
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Adrián Macho-González
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Maryam Jafari
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Aurora Scrivo
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, USA
- Bellvitge Biomedical Research Institute, IDIBELL, University of Barcelona, Barcelona, Spain
| | - Raquel Gomez-Sintes
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Madrid, Spain
| | - Bhakti Chavda
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ana Rosa Saez-Ibanez
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Inmaculada Tasset
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Biochemistry and Molecular Biology, University of Córdoba, Córdoba, Spain
| | - Esperanza Arias
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Medicine, Marion Bessin Liver Research Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Xianhong Xie
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Mimi Kim
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Susmita Kaushik
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ana Maria Cuervo
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA.
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, USA.
- Department of Medicine, Marion Bessin Liver Research Center, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
10
|
Park DE, Aziz M, Salazar JE, Pham T, Nelson SG, Villani J, Weber NO, Price LB, Hungate BA, Liu CM. The nasal microbiome modulates risk for SARS-CoV-2 infection. EBioMedicine 2025:105660. [PMID: 40210576 DOI: 10.1016/j.ebiom.2025.105660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 02/20/2025] [Accepted: 03/06/2025] [Indexed: 04/12/2025] Open
Abstract
BACKGROUND The nasal microbiome may influence host risk for COVID-19 by modulating the expression of key proteins that facilitate SARS-CoV-2 entry, including angiotensin-converting enzyme 2 (ACE2), which binds the virus, and transmembrane serine protease 2 (TMPRSS2), which activates viral entry into nasal epithelial cells. This study examined whether the expression levels of ACE2 and TMPRSS2 in the nasal cavity predict the risk of SARS-CoV-2 infection and whether the host nasal microbiome modulates their expression. METHODS Using 1548 self-collected nasal swabs from a population-based surveillance testing of community-dwelling adults in Washington D.C., we conducted two retrospective case-control studies (cross-sectional: n = 111 cases and 343 controls; longitudinal: n = 97 cases, 286 controls) and a nasal microbiome study (n = 428). Cases, defined as individuals with a positive SARS-CoV-2 test, were matched with controls based on age and test date. Pre-infection samples were analysed. We measured nasal ACE2/TMPRSS2 expression using RT-qPCR and characterized the nasal microbiome using 16S rRNA gene-based qPCR and sequencing. We used machine learning and regression analysis to determine if nasal ACE2/TMPRSS2 expression predicts SARS-CoV-2 infection and whether the nasal microbiome influences their expression. FINDINGS Elevated nasal ACE2/TMPRSS2 expression was associated with 3.6-fold increased risk of contracting COVID-19 (95% CI = 1.71-7.47) compared to those with no detectable levels of ACE2 or TMPRSS2. Before testing positive for SARS-CoV-2, cases also had significantly higher and more unstable ACE2/TMPRSS2 expression in their nasal cavity than controls. Having high densities of Staphylococcus aureus, Haemophilus influenzae, or Moraxella catarrhalis/nonliquefaciens was linked to increased nasal ACE2/TMPRSS2 expression. In contrast, having high densities of Dolosigranulum pigrum was associated with decreased nasal ACE2/TMPRSS2 expression. INTERPRETATION These results suggest that natural variation in the nasal microbiome significantly impacts ACE2/TMPRSS2 expression in the nasal cavity and the near-term risk of SARS-CoV-2 infection in adults. Modifying the nasal microbiome could potentially reduce COVID-19 risk. FUNDING Research reported in this article was supported by the Milken Institute School of Public Health, the George Washington University and the National Institute of Allergy and Infectious Diseases, National Institutes of Health under award number R01AI168182. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.
Collapse
Affiliation(s)
- Daniel E Park
- Antibiotic Resistance Action Center, Department of Environmental and Occupational Health, Milken Institute School of Public Health, George Washington University, Washington, DC, 20052, USA
| | - Maliha Aziz
- Antibiotic Resistance Action Center, Department of Environmental and Occupational Health, Milken Institute School of Public Health, George Washington University, Washington, DC, 20052, USA
| | - Juan E Salazar
- Antibiotic Resistance Action Center, Department of Environmental and Occupational Health, Milken Institute School of Public Health, George Washington University, Washington, DC, 20052, USA
| | - Tony Pham
- Antibiotic Resistance Action Center, Department of Environmental and Occupational Health, Milken Institute School of Public Health, George Washington University, Washington, DC, 20052, USA
| | - Sydney G Nelson
- Antibiotic Resistance Action Center, Department of Environmental and Occupational Health, Milken Institute School of Public Health, George Washington University, Washington, DC, 20052, USA
| | - Jack Villani
- Antibiotic Resistance Action Center, Department of Environmental and Occupational Health, Milken Institute School of Public Health, George Washington University, Washington, DC, 20052, USA
| | - Nathan O Weber
- Antibiotic Resistance Action Center, Department of Environmental and Occupational Health, Milken Institute School of Public Health, George Washington University, Washington, DC, 20052, USA
| | - Lance B Price
- Antibiotic Resistance Action Center, Department of Environmental and Occupational Health, Milken Institute School of Public Health, George Washington University, Washington, DC, 20052, USA
| | - Bruce A Hungate
- Center for Ecosystem Science and Society, Northern Arizona University, Flagstaff, AZ, 86011, USA; Department of Biological Sciences, Northern Arizona University, Flagstaff, AZ, 86011, USA
| | - Cindy M Liu
- Antibiotic Resistance Action Center, Department of Environmental and Occupational Health, Milken Institute School of Public Health, George Washington University, Washington, DC, 20052, USA.
| |
Collapse
|
11
|
Raposo AC, Caldas P, Jeremias J, Arez M, Cazaux Mateus F, Barbosa P, Sousa-Luís R, Água F, Oxley D, Mupo A, Eckersley-Maslin M, Casanova M, Grosso AR, da Rocha ST. Gene reactivation upon erosion of X chromosome inactivation in female hiPSCs is predictable yet variable and persists through differentiation. Stem Cell Reports 2025:102472. [PMID: 40185090 DOI: 10.1016/j.stemcr.2025.102472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 03/06/2025] [Accepted: 03/07/2025] [Indexed: 04/07/2025] Open
Abstract
Female human induced pluripotent stem cells frequently undergo X-chromosome inactivation (XCI) erosion, marked by X-inactive specific transcript (XIST) RNA loss and partial reactivation of the inactive X (Xi). This overlooked phenomenon limits our understanding of its impact on stem cell applications. Here, we show that XCI erosion is frequent and heterogeneous, leading to the reactivation of several X-linked genes. These are primarily located on the short arm of the X chromosome, particularly near escape genes and within H3K27me3-enriched domains, with reactivation linked to reduced promoter DNA methylation. Interestingly, escape genes further increase their expression from Xi upon XCI erosion, highlighting the critical role of XIST in their dosage regulation. Importantly, global (hydroxy)methylation levels and imprinted regions remain unaffected, and analysis of trilineage commitment and cardiomyocyte formation reveals that XCI erosion persists across differentiation. These findings underscore the need for greater awareness of the implications of XCI erosion for stem cell research and clinical applications.
Collapse
Affiliation(s)
- Ana Cláudia Raposo
- iBB - Institute for Bioengineering and Biosciences and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal; Associate Laboratory i4HB Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal; Instituto de Medicina Molecular, João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Paulo Caldas
- Associate Laboratory i4HB Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal; Department of Life Sciences, UCIBIO - Applied Molecular Biosciences Unit, NOVA School of Science and Technology, NOVA University Lisbon, Caparica, Portugal
| | - Joana Jeremias
- iBB - Institute for Bioengineering and Biosciences and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal; Associate Laboratory i4HB Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Maria Arez
- iBB - Institute for Bioengineering and Biosciences and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal; Associate Laboratory i4HB Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Francisca Cazaux Mateus
- iBB - Institute for Bioengineering and Biosciences and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal; Associate Laboratory i4HB Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Pedro Barbosa
- Instituto de Medicina Molecular, João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal; LASIGE, Departamento de Informática, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisbon, Portugal
| | - Rui Sousa-Luís
- Instituto de Medicina Molecular, João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal; Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Frederico Água
- Associate Laboratory i4HB Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal; Department of Life Sciences, UCIBIO - Applied Molecular Biosciences Unit, NOVA School of Science and Technology, NOVA University Lisbon, Caparica, Portugal
| | - David Oxley
- Mass Spectrometry Facility, The Babraham Institute, Cambridge, UK
| | - Annalisa Mupo
- Epigenetics Programme, Babraham Institute, Cambridge CB22 3AT, UK; Altos Labs, Cambridge, UK
| | - Melanie Eckersley-Maslin
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3010, Australia; Department of Anatomy and Physiology, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Miguel Casanova
- iBB - Institute for Bioengineering and Biosciences and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal; Associate Laboratory i4HB Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal; Instituto de Medicina Molecular, João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Ana Rita Grosso
- Associate Laboratory i4HB Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal; Department of Life Sciences, UCIBIO - Applied Molecular Biosciences Unit, NOVA School of Science and Technology, NOVA University Lisbon, Caparica, Portugal
| | - Simão Teixeira da Rocha
- iBB - Institute for Bioengineering and Biosciences and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal; Associate Laboratory i4HB Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal; Instituto de Medicina Molecular, João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
12
|
Sierra I, Toothacre NE, van der Weide RH, Lovell CD, Nguyen SC, Barnett RJ, Cook AL, Ryu HS, Pyfrom S, Wang H, Beiting D, Philips-Cremins JE, Joyce EF, Anguera MC. B cell stimulation changes the structure and higher-order organization of the inactive X chromosome. Cell Rep 2025; 44:115351. [PMID: 40014450 PMCID: PMC11998099 DOI: 10.1016/j.celrep.2025.115351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 12/17/2024] [Accepted: 02/05/2025] [Indexed: 03/01/2025] Open
Abstract
X chromosome inactivation (XCI) equalizes X-linked gene expression between sexes. B cells exhibit dynamic XCI, with Xist RNA/heterochromatic marks absent on the inactive X (Xi) in naive B cells but returning following mitogenic stimulation. The impact of dynamic XCI on Xi structure and maintenance was previously unknown. Here, we find dosage compensation of the Xi with state-specific XCI escape genes in naive and in vitro-activated B cells. Allele-specific OligoPaints indicate similar Xi and active X (Xa) territories in B cells that are less compact than in fibroblasts. Allele-specific Hi-C reveals a lack of TAD-like structures on the Xi of naive B cells and stimulation-induced alterations in TAD-like boundary strength independent of gene expression. Notably, Xist deletion in B cells changes TAD boundaries and large-scale Xi compaction. Altogether, our results uncover B cell-specific Xi plasticity, which could underlie sex-biased biological mechanisms.
Collapse
Affiliation(s)
- Isabel Sierra
- Department of Biomedical Science, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Natalie E Toothacre
- Department of Biomedical Science, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Robin H van der Weide
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, the Netherlands; Oncode Institute, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Claudia D Lovell
- Department of Biomedical Science, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Son C Nguyen
- Department of Genetics, Penn Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - R Jordan Barnett
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ashley L Cook
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Han-Seul Ryu
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sarah Pyfrom
- Department of Biomedical Science, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Harrison Wang
- Department of Biomedical Science, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daniel Beiting
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jennifer E Philips-Cremins
- Department of Genetics, Penn Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA; Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Eric F Joyce
- Department of Genetics, Penn Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Montserrat C Anguera
- Department of Biomedical Science, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA.
| |
Collapse
|
13
|
Sarel-Gallily R, Gunapala KM, Benvenisty N. Large-scale analysis of loss of chromosome Y in human pluripotent stem cells: Implications for Turner syndrome and ribosomopathies. Stem Cell Reports 2025:102471. [PMID: 40185088 DOI: 10.1016/j.stemcr.2025.102471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 03/04/2025] [Accepted: 03/04/2025] [Indexed: 04/07/2025] Open
Abstract
Loss of chromosome Y (LOY) occurs in aging and cancers, but its extent and implications in human embryonic stem cells (hESCs) have not been studied. Here, we analyzed over 2,650 RNA sequencing (RNA-seq) samples from hESCs and their differentiated derivatives to detect LOY. We found that 12% of hESC samples have lost their chromosome Y and identified LOY in all three germ layers. Differential expression analysis revealed that LOY samples showed a decrease in expression of pluripotency markers and in ribosomal protein (RP) genes. Strikingly, significant RP transcription downregulation was observed in most RP genes, although there is only one expressed Y-linked RP gene. We further analyzed RP expression in Turner syndrome and Diamond-Blackfan anemia samples and observed overall downregulation of RP transcription. This broad analysis sheds light on the scope and effects of LOY in hESCs, suggesting a novel dosage-sensitive mechanism regulating RP gene transcription in LOY and autosomal ribosomopathies.
Collapse
Affiliation(s)
- Roni Sarel-Gallily
- The Azrieli Center for Stem Cells and Genetic Research, Department of Genetics, Institute of Life Sciences, The Hebrew University, Jerusalem 91904, Israel
| | - Keith M Gunapala
- The Azrieli Center for Stem Cells and Genetic Research, Department of Genetics, Institute of Life Sciences, The Hebrew University, Jerusalem 91904, Israel; Department of Biomedicine, University of Basel, Mattenstrasse 28, CH-4058 Basel, Switzerland.
| | - Nissim Benvenisty
- The Azrieli Center for Stem Cells and Genetic Research, Department of Genetics, Institute of Life Sciences, The Hebrew University, Jerusalem 91904, Israel.
| |
Collapse
|
14
|
Oliva J, Ruffin M, Calmel C, Gibeaud A, Pizzorno A, Gaudin C, Chardonnet S, de Almeida Bastos V, Rosa-Calatrava M, Soulé A, Emad A, Rousseau S, Corvol H, Terrier O, Guillot L. Divergent responses to SARS-CoV-2 infection in bronchial epithelium with pre-existing respiratory diseases. iScience 2025; 28:111999. [PMID: 40104058 PMCID: PMC11914195 DOI: 10.1016/j.isci.2025.111999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 12/04/2024] [Accepted: 02/07/2025] [Indexed: 03/20/2025] Open
Abstract
Pre-existing respiratory diseases may influence coronavirus disease (COVID-19) susceptibility and severity. However, the molecular mechanisms underlying the airway epithelial response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection severity in patients with chronic respiratory diseases remain unelucidated. Using an in vitro model of differentiated primary bronchial epithelial cells, we aimed to investigate the molecular mechanisms of SARS-CoV-2 infection in pre-existing cystic fibrosis (CF) and chronic obstructive pulmonary disease (COPD). Our study revealed reduced susceptibility of CF and COPD airway epithelia to SARS-CoV-2, relative to that in healthy controls. Mechanistically, reduced transmembrane serine protease 2 (TMPRSS2) activity potentially contributed to this resistance of CF epithelium. Upregulated complement and inflammatory pathways in CF and COPD epithelia potentially primed the antiviral state prior to infection. Analysis of a COVID-19 patient cohort validated our findings, correlating specific inflammatory markers (IP-10, SERPINA1, and CFB) with COVID-19 severity. This study elucidates SARS-CoV-2 pathogenesis and identifies potential biomarkers for clinical monitoring.
Collapse
Affiliation(s)
- Justine Oliva
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 69007 Lyon, France
| | - Manon Ruffin
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), 75012 Paris, France
| | - Claire Calmel
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), 75012 Paris, France
| | - Aurélien Gibeaud
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 69007 Lyon, France
| | - Andrés Pizzorno
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 69007 Lyon, France
- Virnext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, 69008 Lyon, France
- International Research Laboratory RespiVir France - Canada, Centre de Recherche en Infectiologie, Faculté de Médecine RTH Laennec 69008 Lyon, Université Claude Bernard Lyon 1, Université de Lyon, INSERM, CNRS, ENS de Lyon, France, Centre Hospitalier Universitaire de Québec - Université Laval, Québec, QC G1V 4G2, Canada
| | - Clémence Gaudin
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), 75012 Paris, France
| | - Solenne Chardonnet
- Sorbonne Université, INSERM, UMS PASS, Plateforme Post-génomique de la Pitié-Salpêtrière (P3S), 75013 Paris, France
| | - Viviane de Almeida Bastos
- Sorbonne Université, INSERM, UMS PASS, Plateforme Post-génomique de la Pitié-Salpêtrière (P3S), 75013 Paris, France
| | - Manuel Rosa-Calatrava
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 69007 Lyon, France
- Virnext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, 69008 Lyon, France
- International Research Laboratory RespiVir France - Canada, Centre de Recherche en Infectiologie, Faculté de Médecine RTH Laennec 69008 Lyon, Université Claude Bernard Lyon 1, Université de Lyon, INSERM, CNRS, ENS de Lyon, France, Centre Hospitalier Universitaire de Québec - Université Laval, Québec, QC G1V 4G2, Canada
- Centre de Recherche en Infectiologie, Centre Hospitalier Universitaire de Québec, Université Laval, Québec, QC G1V 4G2, Canada
| | - Antoine Soulé
- Department of Electrical and Computer Engineering, McGill University, Montréal, QC, Canada
| | - Amin Emad
- Department of Electrical and Computer Engineering, McGill University, Montréal, QC, Canada
- Mila, Québec AI Institute, Montréal, QC, Canada
| | - Simon Rousseau
- The Meakins-Christie Laboratories at the Research Institute of the McGill University Health Centre Research Institute, Department of Medicine, Faculty of Medicine, McGill University, Montréal, QC, Canada
| | - Harriet Corvol
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), 75012 Paris, France
- Pneumologie Pédiatrique, APHP, Hôpital Trousseau, 75012 Paris, France
| | - Olivier Terrier
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 69007 Lyon, France
| | - Loïc Guillot
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), 75012 Paris, France
| |
Collapse
|
15
|
Dumitrescu L, Seto M, Clifton M, Gomez ML, Coughlan G, Gifford K, Jefferson A, Jager PD, Bennett D, Wang Y, Barnes L, Schneider J, Hohman T, Buckley R. Sex-specific Associations of Gene Expression with Alzheimer's Disease Neuropathology and Ante-mortem Cognitive Performance. RESEARCH SQUARE 2025:rs.3.rs-5938205. [PMID: 40166028 PMCID: PMC11957198 DOI: 10.21203/rs.3.rs-5938205/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The biological mechanisms underlying the increased prevalence of Alzheimer's disease (AD) in women remain undefined. While previous case/control studies have identified sex-biased molecular pathways, the sex-specific relationships between gene expression and AD endophenotypes, particularly involving sex chromosomes, are underexplored. With bulk transcriptomic data across 3 brain regions from 767 decedents, we investigated sex-specific associations between gene expression and post-mortem β-amyloid and tau, as well as antemortem longitudinal cognition. Among 23,118 significant gene associations, 10% were sex-specific, with 73% of these identified in females and primarily associated with tau tangles and longitudinal cognition (90%). Notably, four X-linked genes, MCF2, HDAC8, FTX, and SLC10A3, demonstrated significant sex differences in their associations with AD endophenotypes (i.e., significant sex × gene interaction). Our results also uncovered sex-specific biological pathways, including a female-specific role of neuroinflammation and neuronal development, underscoring the importance of sex-aware analyses to advance precision medicine approaches in AD.
Collapse
Affiliation(s)
| | - Mabel Seto
- Massachusetts General Hospital/Harvard Medical School
| | - Michelle Clifton
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center
| | - Melisa Lara Gomez
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center
| | | | - Katherine Gifford
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Ahern DT, Bansal P, Faustino IV, Chambers OM, Banda EC, Glatt-Deeley HR, Massey RE, Kondaveeti Y, Pinter SF. Isogenic hiPSC models of Turner syndrome development reveal shared roles of inactive X and Y in the human cranial neural crest network. Am J Hum Genet 2025; 112:615-629. [PMID: 39922196 PMCID: PMC11947172 DOI: 10.1016/j.ajhg.2025.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/13/2025] [Accepted: 01/16/2025] [Indexed: 02/10/2025] Open
Abstract
Viable human aneuploidy can be challenging to model in rodents due to syntenic boundaries or primate-specific biology. Human monosomy-X (45,X) causes Turner syndrome (TS), altering craniofacial, skeletal, endocrine, and cardiovascular development, which in contrast remain unaffected in X-monosomic mice. To learn how monosomy-X may impact embryonic development, we turned to 45,X and isogenic euploid human induced pluripotent stem cells (hiPSCs) from male and female mosaic donors. Because the neural crest (NC) is hypothesized to give rise to craniofacial and cardiovascular changes in TS, we assessed differential expression of hiPSC-derived anterior NC cells (NCCs). Across three independent isogenic panels, 45,X NCCs show impaired acquisition of PAX7+SOX10+ markers and disrupted expression of other NCC-specific genes relative to isogenic euploid controls. Additionally, 45,X NCCs increase cholesterol biosynthesis genes while reducing transcripts with 5' terminal oligopyrimidine (TOP) motifs, including those of ribosomal and nuclear-encoded mitochondrial proteins. Such metabolic pathways are also over-represented in weighted co-expression modules that are preserved in monogenic neurocristopathy and reflect 28% of all TS-associated terms of the human phenotype ontology. We demonstrate that 45,X NCCs reduce protein synthesis despite activation of mammalian target of rapamycin (mTOR) but are partially rescued by mild mTOR suppression. Our analysis identifies specific sex-linked genes that are expressed from two copies in euploid males and females alike and qualify as candidate haploinsufficient drivers of TS phenotypes in NC-derived lineages. This study demonstrates that isogenic hiPSC-derived NCC panels representing monosomy-X can serve as powerful models of early NC development in TS and inform new hypotheses toward its etiology.
Collapse
Affiliation(s)
- Darcy T Ahern
- Graduate Program in Genetics and Developmental Biology, UCONN Health, University of Connecticut, Farmington, CT, USA; Department of Genetics and Genome Sciences, UCONN Health, University of Connecticut, Farmington, CT, USA
| | - Prakhar Bansal
- Graduate Program in Genetics and Developmental Biology, UCONN Health, University of Connecticut, Farmington, CT, USA; Department of Genetics and Genome Sciences, UCONN Health, University of Connecticut, Farmington, CT, USA
| | - Isaac V Faustino
- Department of Genetics and Genome Sciences, UCONN Health, University of Connecticut, Farmington, CT, USA
| | - Owen M Chambers
- Department of Genetics and Genome Sciences, UCONN Health, University of Connecticut, Farmington, CT, USA
| | - Erin C Banda
- Department of Genetics and Genome Sciences, UCONN Health, University of Connecticut, Farmington, CT, USA
| | - Heather R Glatt-Deeley
- Department of Genetics and Genome Sciences, UCONN Health, University of Connecticut, Farmington, CT, USA
| | - Rachael E Massey
- Graduate Program in Genetics and Developmental Biology, UCONN Health, University of Connecticut, Farmington, CT, USA; Department of Genetics and Genome Sciences, UCONN Health, University of Connecticut, Farmington, CT, USA; Institute for Systems Genomics, University of Connecticut, Farmington, CT, USA
| | - Yuvabharath Kondaveeti
- Department of Genetics and Genome Sciences, UCONN Health, University of Connecticut, Farmington, CT, USA
| | - Stefan F Pinter
- Graduate Program in Genetics and Developmental Biology, UCONN Health, University of Connecticut, Farmington, CT, USA; Department of Genetics and Genome Sciences, UCONN Health, University of Connecticut, Farmington, CT, USA; Institute for Systems Genomics, University of Connecticut, Farmington, CT, USA.
| |
Collapse
|
17
|
Ansere VA, Kim SS, Marino F, Morillo K, Dubal DB, Murphy CT, Suh Y, Benayoun BA. Strategies for studying sex differences in brain aging. Trends Genet 2025:S0168-9525(25)00027-7. [PMID: 40037936 DOI: 10.1016/j.tig.2025.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 02/06/2025] [Accepted: 02/07/2025] [Indexed: 03/06/2025]
Abstract
Studying sex effects and their underlying mechanisms is of major relevance to understanding brain health. Despite growing interests, experimentally studying sex differences, particularly in the context of aging, remains challenging. Since sex chromosomal content influences gonadal development, separating the effects of gonadal hormones and chromosomal factors requires specific model systems. Here, we highlight rodent and tractable models for examining sex dimorphism in brain and cognitive aging. In addition, we discuss multi-omic and bioinformatic approaches that yield biological insights from animal and human studies. This review provides a comprehensive overview of the diverse toolkit now available to advance our understanding of sex differences in brain aging.
Collapse
Affiliation(s)
- Victor A Ansere
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Seung-Soo Kim
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Francesca Marino
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Katherine Morillo
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Dena B Dubal
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, CA, USA.
| | - Coleen T Murphy
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA; LSI Genomics, Princeton University, Princeton, NJ, USA.
| | - Yousin Suh
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA.
| | - Bérénice A Benayoun
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA; Molecular and Computational Biology Department, USC Dornsife College of Letters, Arts and Sciences, Los Angeles, CA 90089, USA; Biochemistry and Molecular Medicine Department, USC Keck School of Medicine, Los Angeles, CA 90089, USA.
| |
Collapse
|
18
|
Inkster AM, Matthews AM, Phung TN, Plaisier SB, Wilson MA, Brown CJ, Robinson WP. Breaking rules: the complex relationship between DNA methylation and X-chromosome inactivation in the human placenta. Biol Sex Differ 2025; 16:18. [PMID: 40038810 DOI: 10.1186/s13293-025-00696-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 01/28/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND The human placenta is distinct from most organs due to its uniquely low-methylated genome. DNA methylation (DNAme) is particularly depleted in the placenta at partially methylated domains and on the inactive X chromosome (Xi) in XX samples. While Xi DNAme is known to be critical for X-chromosome inactivation (XCI) in other tissues, its role in the placenta remains unclear. Understanding X-linked DNAme variation in the placenta may provide insights into XCI and have implications for prenatal development and phenotypic sex differences. METHODS DNAme data were analyzed from over 350 human placental (chorionic villus) samples, along with samples from cord blood, amnion and chorion placental membranes, and fetal somatic tissues. We characterized X chromosome DNAme variation in the placenta relative to sample variables including cell composition, ancestry, maternal age, placental weight, and fetal birth weight, and compared these patterns to other tissues. We also evaluated the relationship between X-linked DNAme and previously reported XCI gene expression status in placenta. RESULTS Our findings confirm that the placenta exhibits significant depletion of DNAme on the Xi compared to other tissues. Additionally, we observe that X chromosome DNAme profiles in the placenta are influenced by cell composition, particularly trophoblast proportion, with minimal DNAme variation across gestation. Notably, low promoter DNAme is observed at most genes on the Xi regardless of XCI status, challenging known associations in somatic tissues between low promoter DNAme and escape from XCI. CONCLUSIONS This study provides evidence that the human placenta has a distinct Xi DNAme landscape, which may inform our understanding of sex differences during prenatal development. Future research should explore the mechanisms underlying the placenta's unique X-linked DNAme profile, and the factors involved in placental XCI maintenance.
Collapse
Affiliation(s)
- Amy M Inkster
- BC Children's Hospital Research Institute, 950 W 28th Ave, Vancouver, BC, V6H 3N1, Canada.
- Department of Medical Genetics, University of British Columbia, 4500 Oak St, Vancouver, BC, V6H 3N1, Canada.
| | - Allison M Matthews
- BC Children's Hospital Research Institute, 950 W 28th Ave, Vancouver, BC, V6H 3N1, Canada
- Department of Pathology & Laboratory Medicine, University of British Columbia, 221 Wesbrook Mall, Vancouver, BC, V6T 1Z7, Canada
| | - Tanya N Phung
- Center for Evolution and Medicine, School of Life Sciences, Arizona State University, 427 E Tyler Mall, Tempe, AZ, 85281, USA
| | - Seema B Plaisier
- Center for Evolution and Medicine, School of Life Sciences, Arizona State University, 427 E Tyler Mall, Tempe, AZ, 85281, USA
| | - Melissa A Wilson
- Center for Evolution and Medicine, School of Life Sciences, Arizona State University, 427 E Tyler Mall, Tempe, AZ, 85281, USA
| | - Carolyn J Brown
- Department of Medical Genetics, University of British Columbia, 4500 Oak St, Vancouver, BC, V6H 3N1, Canada
| | - Wendy P Robinson
- BC Children's Hospital Research Institute, 950 W 28th Ave, Vancouver, BC, V6H 3N1, Canada
- Department of Medical Genetics, University of British Columbia, 4500 Oak St, Vancouver, BC, V6H 3N1, Canada
| |
Collapse
|
19
|
Du Y, Faz-Lopez B, Ah Kioon MD, Cenac C, Pierides M, Lakin KS, Spiera RF, Chaumeil J, Truchetet ME, Gordon JK, Guéry JC, Barrat FJ. Altered X-chromosome inactivation of the TLR7/8 locus and heterogeneity of pDCs in systemic sclerosis. J Exp Med 2025; 222:e20231809. [PMID: 39670995 PMCID: PMC11639950 DOI: 10.1084/jem.20231809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 09/26/2024] [Accepted: 11/21/2024] [Indexed: 12/14/2024] Open
Abstract
Systemic sclerosis (SSc) is an autoimmune disease that has a strong female predominance. Both the X-linked TLR7 and TLR8 can induce type I IFN (IFN-I) by plasmacytoid DCs (pDCs), which can promote fibrosis. We identified five subclusters of pDCs, including ISGhigh clusters that were over-represented in SSc patients. We observed that both TLR7 and TLR8 genes escape from X chromosome inactivation (XCI) at higher frequency in pDCs of SSc patients, which was associated with changes in TLR7 protein profile. Combined DNA/RNA FISH analysis revealed that the TLR7/8 locus is preferentially located outside of the inactive X (Xi) territory when TLR7 is expressed, suggesting that higher-order loop formation is linked to TLR7/8 expression from the Xi. Furthermore, the expression levels of XIST and the transcriptional repressor SPEN were reduced in SSc pDCs. Hence, our data revealed the heterogeneity of pDCs in SSc and suggested that altered XCI at the TLR7/8 locus may contribute to the chronic IFN-I activity of pDCs in female SSc patients.
Collapse
Affiliation(s)
- Yong Du
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Inflammation and Autoimmunity Program, Hospital for Special Surgery, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medical College of Cornell University, New York, NY, USA
| | - Bérénice Faz-Lopez
- Institut Toulousain des Maladies Infectieuses et Inflammatoires, Université de Toulouse, INSERM, CNRS, UPS, Toulouse, France
| | - Marie Dominique Ah Kioon
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Inflammation and Autoimmunity Program, Hospital for Special Surgery, New York, NY, USA
| | - Claire Cenac
- Institut Toulousain des Maladies Infectieuses et Inflammatoires, Université de Toulouse, INSERM, CNRS, UPS, Toulouse, France
| | - Michael Pierides
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Inflammation and Autoimmunity Program, Hospital for Special Surgery, New York, NY, USA
| | - Kimberly S. Lakin
- Division of Rheumatology and Scleroderma and Vasculitis Center, Department of Medicine, Hospital for Special Surgery, New York, NY, USA
| | - Robert F. Spiera
- Division of Rheumatology and Scleroderma and Vasculitis Center, Department of Medicine, Hospital for Special Surgery, New York, NY, USA
| | - Julie Chaumeil
- Institut Cochin, Université Paris Cité, CNRS, INSERM, Paris, France
| | - Marie-Elise Truchetet
- ImmunoConcEpt, CNRS, UMR 5164, University of Bordeaux, Talence, France
- Rheumatology Department, CHU de Bordeaux, Bordeaux, France
| | - Jessica K. Gordon
- Division of Rheumatology and Scleroderma and Vasculitis Center, Department of Medicine, Hospital for Special Surgery, New York, NY, USA
| | - Jean-Charles Guéry
- Institut Toulousain des Maladies Infectieuses et Inflammatoires, Université de Toulouse, INSERM, CNRS, UPS, Toulouse, France
| | - Franck J. Barrat
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Inflammation and Autoimmunity Program, Hospital for Special Surgery, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medical College of Cornell University, New York, NY, USA
| |
Collapse
|
20
|
Fang H, Tronco AR, Bonora G, Nguyen T, Thakur J, Berletch JB, Filippova GN, Henikoff S, Shendure J, Noble WS, Duan Z, Disteche CM, Deng X. CTCF-mediated insulation and chromatin environment modulate Car5b escape from X inactivation. BMC Biol 2025; 23:68. [PMID: 40025499 PMCID: PMC11874400 DOI: 10.1186/s12915-025-02137-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 01/21/2025] [Indexed: 03/04/2025] Open
Abstract
BACKGROUND Genes that escape X-chromosome inactivation (XCI) in female somatic cells vary in number and levels of escape among mammalian species and tissues, potentially contributing to species- and tissue-specific sex differences. CTCF, a master chromatin conformation regulator, is enriched at escape regions and may play an important role in regulating escape, but the molecular mechanisms remain elusive. RESULTS CTCF binding profiles and epigenetic features were systematically examined at escape genes (escapees) using mouse allelic systems with skewed XCI to distinguish the inactive X (Xi) and active X (Xa) chromosomes. We found that six constitutive and two facultative escapees are located inside 30-800 kb domains marked by convergent arrays of CTCF binding sites, consistent with the formation of chromatin loops. Facultative escapees show clear differences in CTCF binding depending on their XCI status in specific cell types/tissues. In addition, sets of strong and in some cases divergent CTCF binding sites located at the boundary between an escapee and its adjacent neighbors subject to XCI would also help insulate domains. Indeed, deletion but not inversion of a CTCF binding site at the boundary between the facultative escapee Car5b and its silent neighbor Siah1b results in a dramatic reduction of Car5b escape. This is associated with reduced CTCF and cohesin binding, which indicates loss of looping and insulation and is supported by 3C combined with Hi-C analysis. In addition, enrichment in the repressive mark H3K27me3 invades the Car5b domain in deleted cells, consistent with loss of expression from the Xi. In contrast, cells with an inversion of the CTCF binding site retain CTCF and cohesin binding, as well as looping, in line with persistence of escape. Interestingly, the levels of escape increase in cells with deletion of either Dxz4, which disrupts the Xi-specific compact 3D structure, or Firre, which results in lower H3K27me3 enrichment on the Xi, indicating that the structural and epigenetic features of the Xi constrain escape from XCI in wild type conditions. CONCLUSIONS Taken together, our findings support the idea that escape from XCI in female somatic cells is modulated by both the topological insulation of domains via CTCF binding and the surrounding heterochromatin environment.
Collapse
Affiliation(s)
- He Fang
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA
| | - Ana R Tronco
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA
| | - Giancarlo Bonora
- Department of Genome Sciences, University of Washington, Seattle, WA, 98195, USA
| | - Truong Nguyen
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA
| | - Jitendra Thakur
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Joel B Berletch
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA
| | - Galina N Filippova
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA
| | - Steven Henikoff
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Jay Shendure
- Department of Genome Sciences, University of Washington, Seattle, WA, 98195, USA
| | - William S Noble
- Department of Genome Sciences, University of Washington, Seattle, WA, 98195, USA
- Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, WA, 98195, USA
| | - Zhijun Duan
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, 98195, USA
- Division of Hematology, University of Washington, Seattle, WA, 98195, USA
| | - Christine M Disteche
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA.
- Department of Medicine, University of Washington, Seattle, WA, 98195, USA.
| | - Xinxian Deng
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
21
|
DeCasien AR, Tsai K, Liu S, Thomas A, Raznahan A. Evolutionary divergence between homologous X-Y chromosome genes shapes sex-biased biology. Nat Ecol Evol 2025; 9:448-463. [PMID: 39856216 DOI: 10.1038/s41559-024-02627-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 12/10/2024] [Indexed: 01/27/2025]
Abstract
Sex chromosomes are a fundamental aspect of sex-biased biology, but the extent to which homologous X-Y gene pairs ('the gametologs') contribute to sex-biased phenotypes remains hotly debated. Although these genes tend to exhibit large sex differences in expression throughout the body (XX females can express both X members, and XY males can express one X and one Y member), there is conflicting evidence regarding the degree of functional divergence between the X and Y members. Here we develop and apply co-expression fingerprint analysis to characterize functional divergence between the X and Y members of 17 gametolog gene pairs across >40 human tissues. Gametolog pairs exhibit functional divergence between the sexes that is driven by divergence between the X versus Y members (assayed in males), and this within-pair divergence is greatest among pairs with evolutionarily distant X and Y members. These patterns reflect that X versus Y gametologs show coordinated patterns of asymmetric coupling with large sets of autosomal genes, which are enriched for functional pathways and gene sets implicated in sex-biased biology and disease. Our findings suggest that the X versus Y gametologs have diverged in function and prioritize specific gametolog pairs for future targeted experimental studies.
Collapse
Affiliation(s)
- Alex R DeCasien
- Section on Developmental Neurogenomics, Human Genetics Branch, NIMH IRP, NIH, Bethesda, MD, USA.
- Computational and Evolutionary Neurogenomics Unit, Laboratory of Neurogenetics, NIA IRP, NIH, Bethesda, MD, USA.
| | - Kathryn Tsai
- Section on Developmental Neurogenomics, Human Genetics Branch, NIMH IRP, NIH, Bethesda, MD, USA
| | - Siyuan Liu
- Section on Developmental Neurogenomics, Human Genetics Branch, NIMH IRP, NIH, Bethesda, MD, USA
| | - Adam Thomas
- Data Science and Sharing Team, NIMH IRP, NIH, Bethesda, MD, USA
| | - Armin Raznahan
- Section on Developmental Neurogenomics, Human Genetics Branch, NIMH IRP, NIH, Bethesda, MD, USA.
| |
Collapse
|
22
|
Chen HN, Hu YN, Ran LL, Wang M, Zhang Z. Sexual dimorphism in aortic aneurysm: A review of the contributions of sex hormones and sex chromosomes. Vascul Pharmacol 2025; 158:107460. [PMID: 39716526 DOI: 10.1016/j.vph.2024.107460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/23/2024] [Accepted: 12/18/2024] [Indexed: 12/25/2024]
Abstract
Aortic aneurysm is a common cardiovascular disease. Over time, the disease damages the structural and functional integrity of the aorta, causing it to abnormally expand and potentially rupture, which can be fatal. Sex differences are evident in the disease, with men experiencing an earlier onset and higher incidence. However, women may face a worse prognosis and a higher risk of rupture. While there are some studies on the cellular and molecular mechanisms of aneurysm formation, it remains unclear how sex factors contribute to sexual dimorphism. Therefore, this review aims to summarize the role of sex in the occurrence of aortic aneurysms, offering valuable insights for disease prevention and the development of appropriate treatment options.
Collapse
Affiliation(s)
- Hao-Nan Chen
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, China
| | - Yan-Ni Hu
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, China
| | - Li-Ling Ran
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, China
| | - Mi Wang
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, China
| | - Zheng Zhang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Central South University, Changsha 410013, Hunan, China.
| |
Collapse
|
23
|
Franco-Enzástiga Ú, Inturi NN, Natarajan K, Mwirigi JM, Mazhar K, Schlachetzki JC, Schumacher M, Price TJ. Epigenomic landscape of the human dorsal root ganglion: sex differences and transcriptional regulation of nociceptive genes. Pain 2025; 166:614-630. [PMID: 39928726 PMCID: PMC11819886 DOI: 10.1097/j.pain.0000000000003508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 10/14/2024] [Indexed: 02/12/2025]
Abstract
ABSTRACT Cell states are influenced by the regulation of gene expression orchestrated by transcription factors capable of binding to accessible DNA regions. To uncover if sex differences exist in chromatin accessibility in the human dorsal root ganglion (hDRG), where nociceptive neurons innervating the body are found, we performed bulk and spatial assays for transposase-accessible chromatin technology followed by sequencing (ATAC-seq) from organ donors without a history of chronic pain. Using bulk ATAC-seq, we detected abundant sex differences in the hDRG. In women, differentially accessible regions (DARs) mapped mostly to the X chromosome, whereas in men, they mapped to autosomal genes. Hormone-responsive transcription factor binding motifs such as EGR1/3 were abundant within DARs in women, while JUN, FOS, and other activating protein 1 factor motifs were enriched in men, suggesting a higher activation state of cells compared with women. These observations were consistent with spatial ATAC-seq data. Furthermore, we validated that EGR1 expression is biased to female hDRG using RNAscope. In neurons, spatial ATAC-seq revealed higher chromatin accessibility in GABAergic, glutamatergic, and interferon-related genes in women and in Ca2+-signaling-related genes in men. Strikingly, XIST, responsible for inactivating 1 X chromosome by compacting it and maintaining at the periphery of the nucleus, was found to be highly dispersed in female neuronal nuclei. This is likely related to the higher chromatin accessibility in X in female hDRG neurons observed using both ATAC-seq approaches. We have documented baseline epigenomic sex differences in the hDRG which provide important descriptive information to test future hypotheses.
Collapse
Affiliation(s)
- Úrzula Franco-Enzástiga
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080
| | - Nikhil N. Inturi
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080
| | - Keerthana Natarajan
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080
| | - Juliet M. Mwirigi
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080
| | - Khadijah Mazhar
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080
| | - Johannes C.M. Schlachetzki
- Department of Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0651, USA
| | - Mark Schumacher
- Department of Anesthesia and Perioperative Care and the UCSF Pain and Addiction Research Center, University of California, San Francisco, California, 94143 USA
| | - Theodore J. Price
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080
| |
Collapse
|
24
|
Watanabe-Okochi N, Tsuneyama H, Kumamoto M, Tanaka S, Nakazono T, Ichinomiya K, Suzuki Y, Ogasawara K, Uchikawa M, Naganuma S, Hayashi S, Igarashi H, Tsuno NH, Muroi K. Genetic background of anti-CD99 producers in Japan and analysis of hemolytic transfusion reactions due to anti-CD99. Transfusion 2025; 65:604-614. [PMID: 39925213 DOI: 10.1111/trf.18126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 12/10/2024] [Accepted: 12/23/2024] [Indexed: 02/11/2025]
Abstract
BACKGROUND The XG blood group system comprises two antigens, Xga and CD99. CD99 is known to be carried on both the X and Y chromosomes in pseudoautosomal region 1. We identified five unrelated Japanese individuals with anti-CD99 and investigated their genomic background as well as the clinical significance of anti-CD99. STUDY DESIGN AND METHODS Analysis of CD99 expression on RBCs and a modified monocyte monolayer assay was performed using flow cytometry. Genomic DNA was obtained from the five anti-CD99 producers to identify the deleted region responsible for the lack of CD99, and we conducted a long polymerase chain reaction using primer pairs specific for CD99 and GYG2. RESULTS CD99 expression from the Y chromosome was higher than that from the X chromosome. The five anti-CD99 plasma samples gave varied agglutination strengths with the red blood cells (RBCs) expressing high and low CD99 levels, in the antiglobulin test. The phagocytosis rate of anti-CD99-sensitized RBCs was 76.6% in one case indicating a risk of hemolytic transfusion reactions (HTR), and it correlated with the level of CD99 expression. The deleted region spanned 115 kb, from CD99 exon 3 to GYG2 exon 1. All five anti-CD99 producers were homozygous for the large deletion allele. DISCUSSION All five anti-CD99 producers were females with a history of pregnancy in Kyushu, Japan, and this deletion allele may thus be endemic. Our results indicated the possibility of HTR due to anti-CD99, and the risk is low when transfusing RBC products from Xg(a-) females with a low expression of CD99.
Collapse
Affiliation(s)
- Naoko Watanabe-Okochi
- Department of Laboratory, Japanese Red Cross Kanto-Koshinetsu Block Blood Center, Tokyo, Japan
| | - Hatsue Tsuneyama
- Department of Laboratory, Japanese Red Cross Kanto-Koshinetsu Block Blood Center, Tokyo, Japan
| | - Makoto Kumamoto
- Department of Laboratory, Japanese Red Cross Kyushu Block Blood Center, Tokyo, Japan
| | - Sho Tanaka
- Department of Laboratory, Japanese Red Cross Kyushu Block Blood Center, Tokyo, Japan
| | - Tomoko Nakazono
- Department of Laboratory, Japanese Red Cross Kyushu Block Blood Center, Tokyo, Japan
| | | | - Yumi Suzuki
- Department of Laboratory, Japanese Red Cross Kanto-Koshinetsu Block Blood Center, Tokyo, Japan
| | - Kenichi Ogasawara
- Team of Red Blood Cells, Japanese Red Cross Central Blood Institute, Tokyo, Japan
| | - Makoto Uchikawa
- Department of Laboratory, Japanese Red Cross Kanto-Koshinetsu Block Blood Center, Tokyo, Japan
| | - Shinichi Naganuma
- Department of Laboratory, Japanese Red Cross Kanto-Koshinetsu Block Blood Center, Tokyo, Japan
| | - Sumie Hayashi
- Department of Laboratory, Japanese Red Cross Kanto-Koshinetsu Block Blood Center, Tokyo, Japan
| | - Hiroyuki Igarashi
- Department of Laboratory, Japanese Red Cross Kanto-Koshinetsu Block Blood Center, Tokyo, Japan
| | - Nelson-Hirokazu Tsuno
- Department of Laboratory, Japanese Red Cross Kanto-Koshinetsu Block Blood Center, Tokyo, Japan
| | - Kazuo Muroi
- Department of Laboratory, Japanese Red Cross Kanto-Koshinetsu Block Blood Center, Tokyo, Japan
| |
Collapse
|
25
|
Qin S, Luo Z, Wang J, Wang X, Chen X, Ye M, Leng X. Nephrogenic diabetes insipidus results from a novel in-frame deletion of AVPR2 gene in monozygotic-twin boys and their mother and grandmother. J Pediatr Endocrinol Metab 2025; 38:162-171. [PMID: 39648407 DOI: 10.1515/jpem-2024-0301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 11/21/2024] [Indexed: 12/10/2024]
Abstract
OBJECTIVES Mutations in the AVPR2 gene are the most common cause of nephrogenic diabetes insipidus (NDI). In-frame deletions of the AVPR2 gene are a rare variant that results in NDI. We report a novel variant of the p.H138del in an NDI family with twin male patients and three female carriers of different clinical phenotypes. METHODS The proband's blood genome was sequenced with a panel, and the variants were classified according to ACMG/AMP (2015) guidelines. X chromosome inactivation (XCI) was analyzed in the peripheral blood of his mother, grandmother, and maternal aunt, respectively. The haplotypes of the X chromosome were determined using their STR loci. RESULTS A novel in-frame deletion in the AVPR2 gene was detected in monozygotic-twin boys, and his mother, grandmother, and maternal aunt were heterozygous carriers. The two boys showed typical NDI, and their mother and grandmother presented polydipsia, polydipsia, and polyuria, but the maternal aunt did not have similar symptoms. The blood XCI results of the mother, grandmother, and maternal aunt showed random inactivation (36.18 , 48.37, and 49.30 %, respectively). The X haplotype indicated that the variant of the mother and grandmother was on their activated X chromosomes(Xa), while the maternal aunt's variant was on her inactivated X chromosome(Xi). CONCLUSIONS In-frame deletion of the AVPR2 gene within its functional domain can significantly affect protein function, which is one of the vital causes of NDI. The clinical variability of female carriers of AVPR2 is associated with underlying environmental and epigenetic factors or complex recombination of the X chromosomes.
Collapse
Affiliation(s)
- Shengfang Qin
- Department of Medical Genetics and Prenatal Diagnosis, Sichuan Provincial Women's and Children's Hospital/The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Zemin Luo
- Department of Internal Medicine and Paediatrics, Sichuan Provincial Women's and Children's Hospital/The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Jin Wang
- Department of Medical Genetics and Prenatal Diagnosis, Sichuan Provincial Women's and Children's Hospital/The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Xueyan Wang
- Department of Medical Genetics and Prenatal Diagnosis, Sichuan Provincial Women's and Children's Hospital/The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Ximin Chen
- Department of Medical Genetics and Prenatal Diagnosis, Sichuan Provincial Women's and Children's Hospital/The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Mengling Ye
- Department of Medical Genetics and Prenatal Diagnosis, Sichuan Provincial Women's and Children's Hospital/The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Xiangyou Leng
- Department of Medical Genetics and Prenatal Diagnosis, Sichuan Provincial Women's and Children's Hospital/The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| |
Collapse
|
26
|
Cáceres A, Pérez-Jurado LA, Alegret-García A, Dwaraka VB, Smith R, González JR. Defective X-chromosome inactivation and cancer risk in women. Commun Biol 2025; 8:289. [PMID: 39987288 PMCID: PMC11846847 DOI: 10.1038/s42003-025-07691-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 02/06/2025] [Indexed: 02/24/2025] Open
Abstract
X-chromosome inactivation (XCI) is a fundamental mechanism in placental mammals that compensates for gene dosage differences between sexes. Using methylation levels of genes under XCI, we establish defective levels of XCI as a new source of interindividual variation among cancer types in females, characterized by a significant and consistent lowering of XIST expression and enrichment of differentially expressed genes under XCI. We show that defective XCI is an additive factor to the cancer risk of XCI escape deregulation in women. Defective XCI of more than 10% has an attributable risk of 40% among 12 different cancers from The Cancer Genome Atlas. Validations between independent studies of breast cancer samples show that defective XCI increases triple-negative subtype frequency, decreases survival rates, and is reduced by chemotherapy treatment. Mechanistically, it is associated with somatic mutations at TP53 and top MYC gains. In independent studies, defective XCI is detectable in blood and increases with aging, menopause, and cancer diagnosis.
Collapse
Affiliation(s)
- Alejandro Cáceres
- Instituto de Salud Global de Barcelona (ISGlobal), Barcelona, Spain.
- Centro de Investigación Biomédica en Red en Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain.
| | - Luis A Pérez-Jurado
- Genetics Unit, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
- Genetics Service, Hospital del Mar and Institut Hospital del Mar d'Investigacions Mediques (IMIM), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain
| | - Albert Alegret-García
- Instituto de Salud Global de Barcelona (ISGlobal), Barcelona, Spain
- Centro de Investigación Biomédica en Red en Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | | | | | - Juan R González
- Instituto de Salud Global de Barcelona (ISGlobal), Barcelona, Spain
- Centro de Investigación Biomédica en Red en Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| |
Collapse
|
27
|
Liu X, Fan Q, Deng M, Xu Y, Guo J, Cao P, Zhou C, Xu Y. GGN repeat length of the androgen receptor gene is associated with antral follicle count in Chinese women undergoing controlled ovarian stimulation. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2025; 45:213-222. [PMID: 40031964 DOI: 10.12122/j.issn.1673-4254.2025.02.01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
OBJECTIVES To evaluate the association of GGN repeat polymorphism of androgen receptor (AR) with ovarian reserve and ovarian response in controlled ovarian stimulation (COS). METHODS This genetic association study was conducted among a total of 361 women aged ≤40 years with basal FSH≤12 U/L undergoing the GnRH-agonist long protocol for COS in a university-affiliated IVF center. GGN repeat in the AR gene was analyzed with Sanger sequencing. The primary endpoint was the number of antral follicle counts (AFCs), and the secondary endpoints were stimulation days, total dose of gonadotropin (Gn) used, total number of retrieved oocytes, ovarian sensitivity index, and follicular output rate. RESULTS The GGN repeat in exon 1 of the AR gene ranged from 13 to 24, and the median repeat length was 22. Based on the genotypes (S for GGN repeats <22, L for GGN repeats ≥22), the patients were divided into 3 groups: SS, SL, and LL. Generalized regression analysis indicated that the number of AFCs in group SS was significantly lower than those in group SL (adjusted β=1.8, 95% CI: 0.2-3.4, P=0.024) and group LL (adjusted β=1.5, 95% CI: 0.2-2.7, P=0.021). No significant difference was observed in the number of AFCs between group SL and group LL (P>0.05). Generalized regression analysis indicated no significant differences in ovarian stimulation parameters among the 3 groups, either before or after adjusting for confounding factors (P>0.05). CONCLUSIONS GGN repeat length on the AR gene is associated with AFC but not with ovarian response in Chinese women, indicating that AR gene polymorphisms may affect ovarian reserve.
Collapse
Affiliation(s)
- Xinyan Liu
- Guangdong Provincial Key Laboratory of Reproductive Medicine, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510000, China
- Reproductive Medicine Center, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510000, China
| | - Qi Fan
- Reproductive Medicine Center, Huizhou Municipal Central Hospital, Huizhou 516001, China
| | - Mingfen Deng
- Guangdong Provincial Key Laboratory of Reproductive Medicine, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510000, China
- Reproductive Medicine Center, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510000, China
| | - Yan Xu
- Guangdong Provincial Key Laboratory of Reproductive Medicine, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510000, China
- Reproductive Medicine Center, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510000, China
| | - Jing Guo
- Guangdong Provincial Key Laboratory of Reproductive Medicine, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510000, China
- Reproductive Medicine Center, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510000, China
| | - Ping Cao
- Research School for Developmental Biology (GROW), Maastricht University, Maastricht, The Netherlands
- Division of Obstetrics and Gynecology,Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institute, Stockholm, Sweden
| | - Canquan Zhou
- Guangdong Provincial Key Laboratory of Reproductive Medicine, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510000, China
- Reproductive Medicine Center, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510000, China
| | - Yanwen Xu
- Guangdong Provincial Key Laboratory of Reproductive Medicine, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510000, China
- Reproductive Medicine Center, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510000, China
| |
Collapse
|
28
|
Yao C, Dong Y, Zhou H, Zou X, Alhaskawi A, Ezzi SHA, Wang Z, Lai J, Kota VG, Abdulla MHAH, Liu Z, Abdalbary SA, Alenikova O, Lu H. COVID-19 and acute limb ischemia: latest hypotheses of pathophysiology and molecular mechanisms. J Zhejiang Univ Sci B 2025; 26:333-352. [PMID: 40274383 DOI: 10.1631/jzus.b2300512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 01/01/2024] [Indexed: 04/26/2025]
Abstract
Coronavirus disease 2019 (COVID-19) is a multi-system disease that can lead to various severe complications. Acute limb ischemia (ALI) has been increasingly recognized as a COVID-19-associated complication that often predicts a poor prognosis. However, the pathophysiology and molecular mechanisms underlying COVID-19-associated ALI remain poorly understood. Hypercoagulability and thrombosis are considered important mechanisms, but we also emphasize the roles of vasospasm, hypoxia, and acidosis in the pathogenesis of the disease. The angiotensin-converting enzyme 2 (ACE2) pathway, inflammation, and platelet activation may be important molecular mechanisms underlying these pathological changes induced by COVID-19. Furthermore, we discuss the hypotheses of risk factors for COVID-19-associated ALI from genetic, age, and gender perspectives based on our analysis of molecular mechanisms. Additionally, we summarize therapeutic approaches such as use of the interleukin-6 (IL-6) blocker tocilizumab, calcium channel blockers, and angiotensin-converting enzyme inhibitors, providing insights for the future treatment of coronavirus-associated limb ischemic diseases.
Collapse
Affiliation(s)
- Chengjun Yao
- Department of Orthopaedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yanzhao Dong
- Department of Orthopaedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Haiying Zhou
- Department of Orthopaedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Xiaodi Zou
- Department of Orthopaedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310005, China
| | - Ahmad Alhaskawi
- Department of Orthopaedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Sohaib Hasan Abdullah Ezzi
- Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Department of Orthopaedics, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Zewei Wang
- Department of Orthopaedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Jingtian Lai
- Department of Orthopaedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Vishnu Goutham Kota
- Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | | | - Zhenfeng Liu
- Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Sahar Ahmed Abdalbary
- Department of Orthopaedic Physical Therapy, Faculty of Physical Therapy, Nahda University, Beni Suef 2711860, Egypt
| | - Olga Alenikova
- Republic Scientific Practical Center of Neurology and Neurosurgery, Ministry of Health of the Republic of Belarus, Minsk 220004, Belarus
| | - Hui Lu
- Department of Orthopaedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.
| |
Collapse
|
29
|
Chaturvedi SM, Sarafinovska S, Selmanovic D, McCullough KB, Swift RG, Maloney SE, Dougherty JD. Chromosomal and gonadal sex have differing effects on social motivation in mice. Biol Sex Differ 2025; 16:13. [PMID: 39966983 PMCID: PMC11837725 DOI: 10.1186/s13293-025-00690-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 01/25/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Sex differences in brain development are thought to lead to sex variation in social behavior. Sex differences are fundamentally driven by both gonadal hormones and sex chromosomes, yet little is known about the independent effects of each on social behavior. Further, mouse models of the genetic liability for the neurodevelopmental disorder MYT1L Syndrome have shown sex-specific deficits in social motivation. In this study, we aimed to determine if gonadal hormones or sex chromosomes primarily mediate the sex differences seen in mouse social behavior, both at baseline and in the context of Myt1l haploinsufficiency. METHODS Four-core genotypes (FCG) mice, which uncouple gonadal and chromosomal sex, were crossed with MYT1L heterozygous mice to create eight different groups with unique combinations of sex factors and MYT1L genotype. A total of 131 mice from all eight groups were assayed for activity and social behavior via the open field and social operant paradigms. Measures of social seeking and orienting were analyzed for main effects of chromosome, gonads, and their interactions with Myt1l mutation. RESULTS The FCGxMYT1L cross revealed independent effects of both gonadal and chromosomal sex on activity and social behavior. Specifically, the presence of ovarian hormones led to greater overall activity, social seeking, and social orienting regardless of MYT1L genotype. In contrast, sex chromosomes affected social behavior mainly in the MYT1L heterozygous group, with XX MYT1L mutant mice demonstrating elevated levels of social orienting and seeking compared to XY MYT1L mutant mice. CONCLUSIONS Gonadal and chromosomal sex have independent mechanisms of driving greater social motivation in females. Additionally, genes on the sex chromosomes may interact with neurodevelopmental risk genes to influence sex variation in atypical social behavior.
Collapse
Affiliation(s)
- Sneha M Chaturvedi
- Department of Genetics, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO, 63110, USA
- Department of Psychiatry, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO, 63110, USA
| | - Simona Sarafinovska
- Department of Genetics, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO, 63110, USA
- Department of Psychiatry, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO, 63110, USA
| | - Din Selmanovic
- Department of Genetics, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO, 63110, USA
- Department of Psychiatry, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO, 63110, USA
| | - Katherine B McCullough
- Department of Genetics, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO, 63110, USA
- Department of Psychiatry, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO, 63110, USA
| | - Raylynn G Swift
- Department of Genetics, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO, 63110, USA
- Department of Psychiatry, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO, 63110, USA
| | - Susan E Maloney
- Department of Psychiatry, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO, 63110, USA
- Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, St. Louis, MO, 63130, USA
| | - Joseph D Dougherty
- Department of Genetics, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO, 63110, USA.
- Department of Psychiatry, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO, 63110, USA.
- Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, St. Louis, MO, 63130, USA.
| |
Collapse
|
30
|
Arthurs AL, Dietrich B, Knöfler M, Lushington CJ, Thomas PQ, Adikusuma F, Williamson JM, Babikha S, Damhuis T, Jankovic-Karasoulos T, Smith MD, Pringle KG, Roberts CT. Genetically edited human placental organoids cast new light on the role of ACE2. Cell Death Dis 2025; 16:78. [PMID: 39920116 PMCID: PMC11806113 DOI: 10.1038/s41419-025-07400-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 01/05/2025] [Accepted: 01/27/2025] [Indexed: 02/09/2025]
Abstract
ACE2 expression is altered in pregnancy disorders and ACE2 gene variants are associated with several major pregnancy complications including small-for-gestational-age, fetal growth restriction and preeclampsia. This study utilised gene-editing to generate both ACE2 knockout and ACE2 rs2074192 placental organoids, facilitating mechanistic studies into the role of ACE2 in placental development, and the effect of fetal carriage of ACE2 rs2074192 CC, CT and TT genotypes. Parameters of cell and organoid growth were measured, together with qPCR, Western Blotting, and ELISA assessments, in all groups from both organoid models. Here, we report that ACE2 knockout results in delayed placental cell growth and increased cell death. ACE2 knockout organoids had lower ACE protein expression, reduced organoid diameters and asymmetrical growth. Placental organoids with the ACE2 rs2074192 TT genotype had significantly higher expression of ACE2 mRNA and ACE2 protein with elevated ACE2:ACE expression ratio and no change in ACE protein. Despite increased expression of ACE2 protein, ACE2 enzyme activity was significantly decreased in ACE2 rs2074192 TT placental organoids. TT organoids also had reduced diameters and asymmetrical growth. Our research provides a new molecular understanding of the role of ACE2 in placental development, with potential implications for pregnancy in the carriage of the ACE2 rs2074192 gene variant.
Collapse
Affiliation(s)
- Anya L Arthurs
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Adelaide, SA, Australia.
| | - Bianca Dietrich
- Placental Development Group, Medical University of Vienna, Vienna, Austria
| | - Martin Knöfler
- Placental Development Group, Medical University of Vienna, Vienna, Austria
| | - Caleb J Lushington
- School of Biomedicine and Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
- Genome Editing Program, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
| | - Paul Q Thomas
- School of Biomedicine and Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
- Genome Editing Program, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
- South Australian Genome Editing (SAGE), South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
| | - Fatwa Adikusuma
- School of Biomedicine and Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
- Genome Editing Program, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
| | - Jessica M Williamson
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Adelaide, SA, Australia
| | - Susan Babikha
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Adelaide, SA, Australia
| | - Tyla Damhuis
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Adelaide, SA, Australia
| | - Tanja Jankovic-Karasoulos
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Adelaide, SA, Australia
| | - Melanie D Smith
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Adelaide, SA, Australia
| | - Kirsty G Pringle
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
- Mothers and Babies Research Program, Hunter Medical Research Institute, New Lambton Heights, Newcastle, NSW, Australia
| | - Claire T Roberts
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Adelaide, SA, Australia
| |
Collapse
|
31
|
Just J, Ridder LOR, Johannsen EB, Jensen JMB, Petersen MS, Christensen HV, Kjærgaard K, Redder J, Chang S, Stochholm K, Skakkebæk A, Gravholt CH. Elevated levels of neutrophils with a pro-inflammatory profile in Turner syndrome across karyotypes. NPJ Genom Med 2025; 10:9. [PMID: 39915521 PMCID: PMC11803089 DOI: 10.1038/s41525-025-00467-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 01/21/2025] [Indexed: 02/09/2025] Open
Abstract
Turner syndrome (TS) presents with multiple karyotypes, including 45,X monosomy and variants such as isochromosomes and mosaicism, and is characterized by several co-morbidities, including metabolic conditions and autoimmunity. Here, we investigated the genomic landscapes across a range of karyotypes. We show that TS have a common autosomal methylome and transcriptome, despite distinct karyotypic variations. All TS individuals lacked the X chromosome p-arm, and XIST expression from the q-arm did not affect the autosomal transcriptome or methylome, highlighting the critical role of the missing p-arm with its pseudoautosomal region 1. Furthermore, we show increased levels of neutrophils and increased neutrophil activation. The increase in neutrophils was linked to TS clinical traits and to increased expression of the X-Y homologous gene TBL1X, suggesting a genetic basis, which may lead to neutrophil-driven inflammatory stress in TS. Identifying TS individuals with increased neutrophil activation could potentially mitigate the progression towards more severe metabolic issues.
Collapse
Affiliation(s)
- Jesper Just
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark.
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| | - Lukas Ochsner Reynaud Ridder
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark.
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
- Department of Endocrinology, Aarhus University Hospital, Aarhus, Denmark.
| | - Emma Bruun Johannsen
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Jens Magnus Bernth Jensen
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
| | | | | | - Kenneth Kjærgaard
- Department of Data and Data Utilization, Central Denmark Region, Denmark
| | - Jacob Redder
- Department of Data and Data Utilization, Central Denmark Region, Denmark
| | - Simon Chang
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Endocrinology, Aarhus University Hospital, Aarhus, Denmark
| | - Kirstine Stochholm
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Endocrinology, Aarhus University Hospital, Aarhus, Denmark
| | - Anne Skakkebæk
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Clinical Genetics, Aarhus University Hospital, Aarhus, Denmark
| | - Claus Højbjerg Gravholt
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark.
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
- Department of Endocrinology, Aarhus University Hospital, Aarhus, Denmark.
| |
Collapse
|
32
|
Zhang T, Liu Y, Liu F, Guo K, Tang R, Ye J, Xue L, Su Z, Wu ZB. X-linked ubiquitin-specific peptidase 11 (USP11) increases susceptibility to Cushing's disease in women. Acta Neuropathol Commun 2025; 13:22. [PMID: 39910602 DOI: 10.1186/s40478-025-01938-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 01/25/2025] [Indexed: 02/07/2025] Open
Abstract
The incidence of pituitary adrenocorticotropic hormone (ACTH)-secreting PitNETs, commonly known as ACTH PitNETs, is significantly higher in females; however, the underlying causes for this gender disparity remain unclear. In this study, we analyzed the expression of deubiquitinating enzymes in functioning ACTH PitNETs from both male and female subjects using RNA sequencing and identified USP11 as a potential susceptibility factor contributing to the higher prevalence of these PitNETs in females. Further investigation revealed that USP11 expression is markedly elevated in female functioning ACTH PitNETs, with levels significantly higher than those observed in male PitNETs and normal pituitary tissue. Experimental data indicate that USP11 promotes the transcription of proopiomelanocortin (POMC) and the secretion of ACTH. In contrast, knockdown of USP11 leads to a substantial reduction in both POMC transcription and ACTH secretion, as demonstrated in both in vitro and in vivo models. Mechanistically, we found that USP11 facilitates the deubiquitination of the key transcription factor TPIT in functioning ACTH PitNETs, enhancing its protein stability and thereby promoting POMC transcription and ACTH secretion. Additionally, virtual screening identified Lomitapide and Nicergoline as potential inhibitors of USP11, reducing POMC expression and ACTH secretion. Thus, USP11 emerges as a potential therapeutic target, and drugs aimed at inhibiting its function could benefit women with Cushing's disease.
Collapse
Affiliation(s)
- Tao Zhang
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- Department of Neurosurgery, Center of Pituitary Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yanting Liu
- Department of Neurosurgery, Center of Pituitary Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Fang Liu
- Department of Neurosurgery, Center of Pituitary Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Kaiyu Guo
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- Department of Neurosurgery, Center of Pituitary Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Runhua Tang
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Jingwei Ye
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- Department of Neurosurgery, Center of Pituitary Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Li Xue
- Department of Neurosurgery, Center of Pituitary Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Department of Neurosurgery, Center for Immune-Related Diseases, Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Center for Brain Science and Brain-Inspired Technology, Shanghai, 200022, China
| | - Zhipeng Su
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| | - Zhe Bao Wu
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
- Department of Neurosurgery, Center of Pituitary Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Department of Neurosurgery, Center for Immune-Related Diseases, Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
33
|
Sierra I, Toothacre NE, van der Weide RH, Lovell CD, Nguyen SC, Jordan Barnett R, Cook AL, Ryu HS, Pyfrom S, Wang H, Beiting D, Philips-Cremins JE, Joyce EF, Anguera MC. B cell stimulation changes the structure and higher-order organization of the inactive X chromosome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.30.635789. [PMID: 39975382 PMCID: PMC11838360 DOI: 10.1101/2025.01.30.635789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
X Chromosome Inactivation (XCI) equalizes X-linked gene expression between sexes. B cells exhibit dynamic XCI, with Xist RNA/heterochromatic marks absent on the inactive X (Xi) in naive B cells but returning following mitogenic stimulation. The impact of dynamic XCI on Xi structure and maintenance was previously unknown. Here, we find dosage compensation of the Xi with state-specific XCI escape genes in naive and in vitro activated B cells. Allele-specific OligoPaints indicate similar Xi and Xa territories in B cells that are less compact than in fibroblasts. Allele-specific Hi-C reveals a lack of TAD-like structures on the Xi of naive B cells, and stimulation-induced alterations in TAD-like boundary strength independent of gene expression. Notably, Xist deletion in B cells changes TAD boundaries and large-scale Xi compaction. Altogether, our results uncover B cell-specific Xi plasticity which could underlie sex-biased biological mechanisms.
Collapse
|
34
|
Lu Y, Qin M, Qi X, Yang M, Zhai F, Zhang J, Yan Z, Yan L, Qiao J, Yuan P. Sex differences in human pre-gastrulation embryos. SCIENCE CHINA. LIFE SCIENCES 2025; 68:397-415. [PMID: 39327393 DOI: 10.1007/s11427-024-2721-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 09/02/2024] [Indexed: 09/28/2024]
Abstract
Human fetuses exhibit notable sex differences in growth rate and response to the intrauterine environment, yet their origins and underlying mechanisms remain uncertain. Here, we conduct a detailed investigation of sex differences in human pre-gastrulation embryos. The lower methylation and incomplete inactivation of the X chromosome in females, as well as the sex-specific cell-cell communication patterns, contribute to sex-differential transcription. Male trophectoderm is more inclined toward syncytiotrophoblast differentiation and exhibits a stronger hormone secretion capacity, while female trophectoderm tends to retain cytotrophoblast program with stronger mitochondrial function as well as higher vasculogenesis and immunotolerance signals. Male primitive endoderm initiates the anterior visceral endoderm transcriptional program earlier than females. The cell cycle activities of the epiblast and primitive endoderm are higher in males compared to females, while the situation is opposite in the trophectoderm. In conclusion, our study provides in-depth insights into the sex differences in human pre-gastrulation embryos and contributes to unraveling the origins of the sex differences in human fetal development.
Collapse
Affiliation(s)
- Yongjie Lu
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, 100191, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
| | - Meng Qin
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, 100191, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
| | - Xintong Qi
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, 100191, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Ming Yang
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, 100191, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
| | - Fan Zhai
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, 100191, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
| | - Jiaqi Zhang
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, 100191, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
| | - Zhiqiang Yan
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, 100191, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China
| | - Liying Yan
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China.
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, 100191, China.
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China.
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China.
| | - Jie Qiao
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China.
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, 100191, China.
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China.
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China.
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China.
| | - Peng Yuan
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China.
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, 100191, China.
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China.
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China.
| |
Collapse
|
35
|
Buffet A, Filser M, Bruel A, Dard R, Quibel T, Dubucs C, Kwon T, Le Tanno P, Thevenon J, Ziegler A, Allard L, Guigonis V, Roux JJ, Heidet L, Rougeulle C, Boyer O, Vargas-Poussou R, Hureaux M. X-linked transient antenatal Bartter syndrome related to MAGED2 gene: Enriching the phenotypic description and pathophysiologic investigation. Genet Med 2025; 27:101217. [PMID: 39036894 DOI: 10.1016/j.gim.2024.101217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 07/12/2024] [Accepted: 07/12/2024] [Indexed: 07/23/2024] Open
Abstract
PURPOSE Transient Bartter syndrome related to pathogenic variants of MAGED2 is the most recently described antenatal Bartter syndrome. Despite its transient nature, it is the most severe form of Bartter syndrome in the perinatal period. Our aim was to describe 14 new cases and to try to explain the incomplete penetrance in women. METHODS We report on 14 new cases, including 3 females, and review the 40 cases described to date. We tested the hypothesis that MAGED2 is transcriptionally regulated by differential methylation of its CpG-rich promotor by pyrosequencing of DNA samples extracted from fetal and adult leukocytes and kidney samples. RESULTS Analysis of the data from 54 symptomatic patients showed spontaneous resolution of symptoms in 27% of cases, persistent complications in 41% of cases, and fatality in 32% of cases. Clinical anomalies were reported in 76% of patients, mostly renal anomalies (52%), cardiovascular anomalies (29%), and dysmorphic features (13%). A developmental delay was reported in 24% of patients. Variants were found in all regions of the gene. Methylation analysis of the MAGED2 CpG-rich promotor showed a correlation with gender, independent of age, tissue or presence of symptoms, excluding a role for this mechanism in the incomplete penetrance in women. CONCLUSION This work enriches the phenotypic and genetic description of this recently described disease and deepens our understanding of the pathophysiological role and regulation of MAGED2. Finally, by describing the wide range of outcomes in patients, this work opens the discussion on genetic counseling offered to families.
Collapse
Affiliation(s)
- Alexandre Buffet
- Département de Médecine Génomique des Tumeurs et Cancers, Hôpital Européen Georges Pompidou, Fédération de Génétique et de Médecine Génomique Assistance Publique-Hôpitaux de Paris Centre Université Paris Cité, Paris, France; Université Paris Cité, Inserm, PARCC, Paris, France
| | - Mathilde Filser
- Département de Médecine Génomique des Tumeurs et Cancers, Hôpital Européen Georges Pompidou, Fédération de Génétique et de Médecine Génomique Assistance Publique-Hôpitaux de Paris Centre Université Paris Cité, Paris, France
| | - Alexandra Bruel
- Service de Pédiatrie, Centre Hospitalier Universitaire de Nantes, Nantes, France
| | - Rodolphe Dard
- Service de Génétique médicale, Centre Hospitalier Intercommunal de Poissy, Poissy, France
| | - Thibaud Quibel
- Service d'Obstétrique, Centre Hospitalier Intercommunal de Poissy, Poissy, France
| | - Charlotte Dubucs
- Service de Génétique médicale, Oncopole Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Theresa Kwon
- Service de Néphrologie Pédiatrique, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Pauline Le Tanno
- Unité de génétique clinique, Centre Hospitalier Universitaire de Grenoble site Nord - Hôpital Couple-Enfant - Université Grenoble Alpes, La Tranche, France
| | - Julien Thevenon
- Unité de génétique clinique, Centre Hospitalier Universitaire de Grenoble site Nord - Hôpital Couple-Enfant - Université Grenoble Alpes, La Tranche, France
| | - Alban Ziegler
- Service de Génétique Médicale, Centre Hospitalier Universitaire d'Angers, Angers, France; Service de Génétique, CRMR AnDDI-Rares, CHU Reims, Reims, France
| | - Lise Allard
- Service de néphrologie pédiatrique, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Vincent Guigonis
- Service de Néphrologie Pédiatrique, Centre Hospitalier Universitaire de Limoges, Limoges, France
| | - Jean-Jacques Roux
- Laboratoire d'Anatomie et cytologie pathologique, Centre Hospitalier Métropole Savoie, Chambéry, France
| | - Laurence Heidet
- Service de Néphrologie Pédiatrique, Centre de Référence des Maladies Rénales Héréditaires de l'Enfant et de l'Adulte (MARHEA), Hôpital Necker Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris; Laboratoire des Maladies Rénales Héréditaires, Inserm UMR 1163, Institut Imagine, Université de Paris-Cité, Paris, France
| | - Claire Rougeulle
- Université Paris Cité, CNRS, Epigenetics and Cell Fate, Paris, France
| | - Olivia Boyer
- Université Paris Cité, Inserm, PARCC, Paris, France; Service de Médecine Génomique, Hôpital Européen Georges Pompidou, Fédération de Génétique et de Médecine Génomique Assistance Publique-Hôpitaux de Paris Centre Université Paris Cité, Paris, France
| | - Rosa Vargas-Poussou
- Service de Médecine Génomique, Hôpital Européen Georges Pompidou, Fédération de Génétique et de Médecine Génomique Assistance Publique-Hôpitaux de Paris Centre Université Paris Cité, Paris, France
| | - Marguerite Hureaux
- Université Paris Cité, Inserm, PARCC, Paris, France; Service de Médecine Génomique, Hôpital Européen Georges Pompidou, Fédération de Génétique et de Médecine Génomique Assistance Publique-Hôpitaux de Paris Centre Université Paris Cité, Paris, France; Centre de Recherche Cardio-vasculaire de Paris, UMR970, Paris, France.
| |
Collapse
|
36
|
Snell DM, Turner JMA. X chromosome passed from mother to daughter influences brain ageing. Nature 2025; 638:45-47. [PMID: 39843690 DOI: 10.1038/d41586-025-00079-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
|
37
|
Baek IC, Sim SY, Suh BK, Kim TG, Cho WK. Assessment of XCI skewing and demonstration of XCI escape region based on single-cell RNA sequencing: comparison between female Grave's disease and control. BMC Mol Cell Biol 2025; 26:8. [PMID: 39891056 PMCID: PMC11786500 DOI: 10.1186/s12860-025-00533-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 01/27/2025] [Indexed: 02/03/2025] Open
Abstract
BACKGROUND The reactivation and loss of mosaicism hypothesis due to X chromosome inactivation (XCI) skewing and escape could influence gender differences in autoimmune diseases. XCI selectively inactivates one of the two X chromosomes in females. METHODS To estimate XCI skewing and the occurrence of XCI escape, we conducted a normal female (NF) without a history of autoimmune thyroid disease (AITD) and a patient with Grave's disease (GD) based on a thyroid diagnosis. After single-cell RNA sequencing, heterozygous variants were converted and transformed. XCI skewing was calculated using the formula and the skewing degree was defined. NF/GD genes were compared using correction methods. Positions are heterozygous within a single cell as indicated by a unique barcode. RESULTS XCI skewing showed 45.8%/48.9% relatively random, 29.4%/27.0% skewing, 24.6%/23.7% severe skewing, and 0.2%/0.4% extreme severe skewing. 24.8%/24.1% in NF/GD exhibited severe skewing or higher. A total of 13 genes were significantly associated with XCI skewing ratios in NF/GD cells. In total, 371/250 nucleotide positions with only one barcode (representing a unique cell) were identified for XCI escape. A total of 143/52 nucleotide positions spanned 20/6 genes, and 12/1 genes were identified as XCI escapes. CONCLUSIONS These results could aid in understanding the immunogenetics of gender differences in various autoimmune disease pathophysiologies.
Collapse
Affiliation(s)
- In-Cheol Baek
- Catholic Hematopoietic Stem Cell Bank, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Soo Yeun Sim
- Department of Pediatrics, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Byung-Kyu Suh
- Department of Pediatrics, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Tai-Gyu Kim
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Won Kyoung Cho
- Department of Pediatrics, College of Medicine, St. Vincent's Hospital, The Catholic University of Korea, 93, Jungbu-daero, Paldal-gu, Suwon-si, Seoul, Gyeonggi-do, 16247, Republic of Korea.
| |
Collapse
|
38
|
Toothacre NE, Rodríguez-Acevedo KL, Wiggins KJ, Scharer CD, Anguera MC. Xist RNA Dependent and Independent Mechanisms Regulate Dynamic X Chromosome Inactivation in B Lymphocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.27.635124. [PMID: 39975415 PMCID: PMC11838359 DOI: 10.1101/2025.01.27.635124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
X-Chromosome Inactivation (XCI) involves epigenetic pathways to equalize X-linked gene expression between female and male mammals. XCI is dynamic in female B cells, as cytological enrichment of Xist RNA and heterochromatic marks on the inactive X-chromosome (Xi) are absent in naïve B cells yet return following mitogenic stimulation. Here, we asked whether any heterochromatic histone marks are present on the Xi in naïve B cells, and whether Xist RNA is required for their deposition and retention following stimulation. We find that the Xi in naïve B cells is depleted for H2AK119Ub and H3K9me3 but enriched for DNA methylation and H3K27me3, which maintain an Xist RNA-dependent epigenetic memory of XCI. Upon stimulation, Xist-independent H3K27me3 and Xist-dependent H2AK119Ub modifications accumulate across the Xi with temporal and spatial specificity. Our findings reveal the importance of Xist RNA, H3K27me3, and H2AK119Ub marks for the epigenetic integrity of X-linked genes across the Xi following female B cell stimulation.
Collapse
|
39
|
Hromić-Jahjefendić A, Aljabali AAA. Analysis of the immune response in COVID-19. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2025; 213:31-71. [PMID: 40246347 DOI: 10.1016/bs.pmbts.2024.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
The COVID-19 pandemic, instigated by the novel coronavirus SARS-CoV-2, has emerged as a significant global health challenge, demanding a profound grasp of the immune response. The innate immune system, a multifaceted network encompassing pattern recognition receptors (PRRs) and effector cells, assumes a pivotal function in detecting and countering this viral assailant. Toll-like receptors (TLRs), situated on immune cell surfaces and within endosomes, play a central role in recognizing SARS-CoV-2. TLR-2 and TLR-4 discern specific viral constituents, such as the spike (S) protein, setting off inflammatory signaling cascades and catalyzing the generation of type I interferons. Intracellular PRRs, including the RIG-I-like receptors (RLRs), RIG-I and MDA5, detect viral RNA within the cytoplasm of infected cells, provoking antiviral responses by initiating the synthesis of type I interferons. The equilibrium between interferons and pro-inflammatory cytokines dictates the outcomes of the disease. Interferons play an indispensable role in governing viral replication, while unregulated cytokine production can result in tissue harm and inflammation. This intricate dynamic underpins therapeutic strategies aimed at regulating immune responses in individuals grappling with COVID-19. Natural killer (NK) cells, with their capacity to recognize infected cells through the "missing self" phenomenon and activating receptors, make significant contributions to the defense against SARS-CoV-2. NK cells play a pivotal role in eliminating infected cells and boosting immune responses through antibody-dependent cell-mediated cytotoxicity (ADCC). In conclusion, comprehending the interplay among PRRs, interferons, and NK cells within innate immunity is paramount for discerning and combatting SARS-CoV-2. This comprehension illuminates therapeutic interventions and vaccine development, casting light on our endeavors to confront this worldwide health crisis.
Collapse
Affiliation(s)
- Altijana Hromić-Jahjefendić
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Sarajevo, Bosnia and Herzegovina.
| | - Alaa A A Aljabali
- Faculty of Pharmacy, Department of Pharmaceutics and Pharmaceutical Technology, Yarmouk University, Irbid, Jordan
| |
Collapse
|
40
|
Jiang Z, Sullivan PF, Li T, Zhao B, Wang X, Luo T, Huang S, Guan PY, Chen J, Yang Y, Stein JL, Li Y, Liu D, Sun L, Zhu H. The X chromosome's influences on the human brain. SCIENCE ADVANCES 2025; 11:eadq5360. [PMID: 39854466 PMCID: PMC11759047 DOI: 10.1126/sciadv.adq5360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 12/23/2024] [Indexed: 01/26/2025]
Abstract
Genes on the X chromosome are extensively expressed in the human brain. However, little is known for the X chromosome's impact on the brain anatomy, microstructure, and functional networks. We examined 1045 complex brain imaging traits from 38,529 participants in the UK Biobank. We unveiled potential autosome-X chromosome interactions while proposing an atlas outlining dosage compensation for brain imaging traits. Through extensive association studies, we identified 72 genome-wide significant trait-locus pairs (including 29 new associations) that share genetic architectures with brain-related disorders, notably schizophrenia. Furthermore, we found unique sex-specific associations and assessed variations in genetic effects between sexes. Our research offers critical insights into the X chromosome's role in the human brain, underscoring its contribution to the differences observed in brain structure and functionality between sexes.
Collapse
Affiliation(s)
- Zhiwen Jiang
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Patrick F. Sullivan
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Tengfei Li
- Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Biomedical Research Imaging Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Bingxin Zhao
- Department of Statistics and Data Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xifeng Wang
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Tianyou Luo
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Shuai Huang
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Peter Y. Guan
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jie Chen
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yue Yang
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jason L. Stein
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yun Li
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Dajiang Liu
- Department of Public Health Sciences, Penn State University, Hershey, PA 17033, USA
- Department of Biochemistry and Molecular Biology, Penn State University, Hershey, PA 17033, USA
| | - Lei Sun
- Department of Statistical Sciences, University of Toronto, Toronto, ON M5G 1Z5, Canada
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada
| | - Hongtu Zhu
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Biomedical Research Imaging Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Statistics and Operations Research, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
41
|
Bobotis BC, Khakpour M, Braniff O, de Andrade EG, Gargus M, Allen M, Carrier M, Baillargeon J, Rangachari M, Tremblay MÈ. Sex chromosomes and sex hormones differently shape microglial properties during normal physiological conditions in the adult mouse hippocampus. J Neuroinflammation 2025; 22:18. [PMID: 39856696 PMCID: PMC11762133 DOI: 10.1186/s12974-025-03341-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
The brain presents various structural and functional sex differences, for which multiple factors are attributed: genetic, epigenetic, metabolic, and hormonal. While biological sex is determined by both sex chromosomes and sex hormones, little is known about how these two factors interact to establish this dimorphism. Sex differences in the brain also affect its resident immune cells, microglia, which actively survey the brain parenchyma and interact with sex hormones throughout life. However, microglial differences in density and distribution, morphology and ultrastructural patterns in physiological conditions during adulthood are largely unknown. Here, we investigated these aforementioned properties of microglia using the Four Core Genotypes (FCG) model, which allows for an independent assessment of gonadal hormones and sex chromosomal effects in four conditions: FCG XX and Tg XY- (both ovaries); Tg XXSry and Tg XYSry (both testes). We also compared the FCG results with XX and XY wild-type (WT) mice. In adult mice, we focused our investigation on the ventral hippocampus across different layers: CA1 stratum radiatum (Rad) and CA1 stratum lacunosum-moleculare (LMol), as well as the dentate gyrus polymorphic layer (PoDG). Double immunostaining for Iba1 and TMEM119 revealed that microglial density is influenced by both sex chromosomes and sex hormones. We show in the Rad and LMol that microglia are denser in FCG XX compared to Tg XYSry mice, however, microglia were densest in WT XX mice. In the PoDG, ovarian animals had increased microglial density compared to testes animals. Additionally, microglial morphology was modulated by a complex interaction between hormones and chromosomes, affecting both their cellular soma and arborization across the hippocampal layers. Moreover, ultrastructural analysis showed that microglia in WT animals make overall more contacts with pre- and post-synaptic elements than in FCG animals. Lastly, microglial markers of cellular stress, including mitochondrion elongation, and dilation of the endoplasmic reticulum and Golgi apparatus, were mostly chromosomally driven. Overall, we characterized different aspects of microglial properties during normal physiological conditions that were found to be shaped by sex chromosomes and sex hormones, shading more light onto how sex differences affect the brain immunity at steady-state.
Collapse
Affiliation(s)
- Bianca Caroline Bobotis
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada
| | - Mohammadparsa Khakpour
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada
| | - Olivia Braniff
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada
| | | | - Makenna Gargus
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada
| | - Micah Allen
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada
| | - Micaël Carrier
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec City, QC, Canada
- Département de psychiatrie et de neurosciences, Faculté de médecine, Université Laval, Québec City, QC, Canada
| | - Joanie Baillargeon
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec City, QC, Canada
| | - Manu Rangachari
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec City, QC, Canada
- Département de médecine moléculaire, Faculté de médecine, Université Laval, Québec City, QC, Canada
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada.
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec City, QC, Canada.
- Département de médecine moléculaire, Faculté de médecine, Université Laval, Québec City, QC, Canada.
- Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada.
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada.
- Institute on Aging and Lifelong Health (IALH), University of Victoria, Victoria, BC, Canada.
| |
Collapse
|
42
|
Sekino Y, Nakahara H, Ikeda K, Kobatake K, Kohada Y, Tasaka R, Takemoto K, Miyamoto S, Kitano H, Goto K, Goriki A, Hieda K, Hinata N. The Gender-Biased Differential Effect of KDM6A Mutation on Immune Therapy in Urothelial Carcinoma: A Public Database Study. Cancers (Basel) 2025; 17:356. [PMID: 39941725 PMCID: PMC11816370 DOI: 10.3390/cancers17030356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 01/16/2025] [Accepted: 01/19/2025] [Indexed: 02/16/2025] Open
Abstract
Background/Objectives: It is said that genes that escape from X chromosome inactivation (XCI) contribute to gender differences. We analyzed the prognostic role of these genes and identified a gender-biased difference in prognosis according to KDM6A mutation in the immune therapy cohort (IMvigor 210). We also investigate the gender-biased differential effect of KDM6A mutation in several public databases of urothelial carcinoma (UC). Methods: We used AACR GENIE, The Cancer Genome Atlas, International Cancer Genome Consortium, several public databases related to immune therapy, chemotherapy, and BCG treatment. We studied the gender-biased prognostic role of KDM6A mutation in several cohorts and the association between KDM6A mutation and immune-related fractions according to gender. Results: The expression of KDM6A was higher in females than in males in several cohorts. Mutation of KDM6A was observed in about 20-25% of the patients. The rate of KDM6A mutation was higher in females than in males in several cohorts. Kaplan-Meier analysis revealed a gender-biased difference in prognosis between patients with KDM6A mutations and those with the wild-type KDM6A in several cohorts, including the immune therapy cohort. The rate of immune-inflamed type was higher in males than in females in the patients with KDM6A mutation in the IMvigor 210 and UC-GENOME studies. Single-sample Gene Set Enrichment Analysis showed that CD8+ cells and type 1 IFN response fractions and APC co-inhibition fraction were higher in the male than female patients with KDM6A mutation. Similar findings were observed in other immune-related studies (UC-GENOME). Conclusions: The effect of KDM6A mutation on immune therapy varied according to gender, and the status of KDM6A mutation may be a promising biomarker in immune therapy in UC.
Collapse
Affiliation(s)
- Yohei Sekino
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan; (K.I.); (K.K.); (Y.K.); (R.T.); (K.T.); (S.M.); (H.K.); (K.G.); (A.G.); (K.H.); (N.H.)
| | - Hikaru Nakahara
- Department of Clinical and Molecular Genetics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan;
| | - Kenichiro Ikeda
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan; (K.I.); (K.K.); (Y.K.); (R.T.); (K.T.); (S.M.); (H.K.); (K.G.); (A.G.); (K.H.); (N.H.)
| | - Kohei Kobatake
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan; (K.I.); (K.K.); (Y.K.); (R.T.); (K.T.); (S.M.); (H.K.); (K.G.); (A.G.); (K.H.); (N.H.)
| | - Yuki Kohada
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan; (K.I.); (K.K.); (Y.K.); (R.T.); (K.T.); (S.M.); (H.K.); (K.G.); (A.G.); (K.H.); (N.H.)
| | - Ryo Tasaka
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan; (K.I.); (K.K.); (Y.K.); (R.T.); (K.T.); (S.M.); (H.K.); (K.G.); (A.G.); (K.H.); (N.H.)
| | - Kenshiro Takemoto
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan; (K.I.); (K.K.); (Y.K.); (R.T.); (K.T.); (S.M.); (H.K.); (K.G.); (A.G.); (K.H.); (N.H.)
| | - Shunsuke Miyamoto
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan; (K.I.); (K.K.); (Y.K.); (R.T.); (K.T.); (S.M.); (H.K.); (K.G.); (A.G.); (K.H.); (N.H.)
| | - Hiroyuki Kitano
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan; (K.I.); (K.K.); (Y.K.); (R.T.); (K.T.); (S.M.); (H.K.); (K.G.); (A.G.); (K.H.); (N.H.)
| | - Keisuke Goto
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan; (K.I.); (K.K.); (Y.K.); (R.T.); (K.T.); (S.M.); (H.K.); (K.G.); (A.G.); (K.H.); (N.H.)
| | - Akihiro Goriki
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan; (K.I.); (K.K.); (Y.K.); (R.T.); (K.T.); (S.M.); (H.K.); (K.G.); (A.G.); (K.H.); (N.H.)
| | - Keisuke Hieda
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan; (K.I.); (K.K.); (Y.K.); (R.T.); (K.T.); (S.M.); (H.K.); (K.G.); (A.G.); (K.H.); (N.H.)
| | - Nobuyuki Hinata
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan; (K.I.); (K.K.); (Y.K.); (R.T.); (K.T.); (S.M.); (H.K.); (K.G.); (A.G.); (K.H.); (N.H.)
| |
Collapse
|
43
|
Rengarajan S, Derks J, Bellott DW, Slavov N, Page DC. Post-transcriptional cross- and auto-regulation buffer expression of the human RNA helicases DDX3X and DDX3Y. Genome Res 2025; 35:20-30. [PMID: 39794123 PMCID: PMC11789639 DOI: 10.1101/gr.279707.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 11/26/2024] [Indexed: 01/13/2025]
Abstract
The Y-linked gene DDX3Y and its X-linked homolog DDX3X survived the evolution of the human sex chromosomes from ordinary autosomes. DDX3X encodes a multifunctional RNA helicase, with mutations causing developmental disorders and cancers. We find that, among X-linked genes with surviving Y homologs, DDX3X is extraordinarily dosage sensitive. Studying cells of individuals with sex chromosome aneuploidy, we observe that when the number of Y Chromosomes increases, DDX3X transcript levels fall; conversely, when the number of X Chromosomes increases, DDX3Y transcript levels fall. In 46,XY cells, CRISPRi knockdown of either DDX3X or DDX3Y causes transcript levels of the homologous gene to rise. In 46,XX cells, chemical inhibition of DDX3X protein activity elicits an increase in DDX3X transcript levels. Thus, perturbation of either DDX3X or DDX3Y expression is buffered: by negative cross-regulation of DDX3X and DDX3Y in 46,XY cells and by negative auto-regulation of DDX3X in 46,XX cells. DDX3X-DDX3Y cross-regulation is mediated through mRNA destabilization-as shown by metabolic labeling of newly transcribed RNA-and buffers total levels of DDX3X and DDX3Y protein in human cells. We infer that post-transcriptional auto-regulation of the ancestral (autosomal) DDX3X gene transmuted into auto- and cross-regulation of DDX3X and DDX3Y as these sex-linked genes evolved from ordinary alleles of their autosomal precursor.
Collapse
Affiliation(s)
- Shruthi Rengarajan
- Whitehead Institute, Cambridge, Massachusetts 02142, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Jason Derks
- Departments of Bioengineering, Biology, Chemistry, and Chemical Biology, Single Cell Proteomics Center, and Barnett Institute, Northeastern University, Boston, Massachusetts 02115, USA
| | | | - Nikolai Slavov
- Departments of Bioengineering, Biology, Chemistry, and Chemical Biology, Single Cell Proteomics Center, and Barnett Institute, Northeastern University, Boston, Massachusetts 02115, USA
| | - David C Page
- Whitehead Institute, Cambridge, Massachusetts 02142, USA;
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
- Howard Hughes Medical Institute, Whitehead Institute, Cambridge, Massachusetts 02142, USA
| |
Collapse
|
44
|
Fang H, Jiang L, da Veiga Leprevost F, Jian R, Chan J, Glinos D, Lappalainen T, Nesvizhskii AI, Reiner AP, Consortium GTE, Snyder MP, Tang H. Regulation of protein abundance in normal human tissues. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.01.10.25320181. [PMID: 39867362 PMCID: PMC11759590 DOI: 10.1101/2025.01.10.25320181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
We report a systematic quantification of 10,841 unique proteins from over 700 GTEx samples, representing five human tissues. Sex, age and genetic factors are associated with variation in protein abundance. In total, 1981 cis-protein quantitative trait loci (cis-pQTL) are identified, of which a majority of protein targets have not been assayed in the recent plasma-based proteogenomic studies. Integrating transcriptomic information from matching tissues delineates concordant as well as discordant expression patterns at RNA and protein levels. Juxtaposition of data from different tissues indicates both shared and tissue-specific genetic architecture that underlie protein abundance. Complementing genomic annotation, RNA-based eQTL studies, as well as the recent establishment of plasma-based proteogenomic characterization, tissue-pQTLs shed light on biology underlying genotype-phenotype association of complex traits and diseases.
Collapse
|
45
|
Wu Y, Yan Y, Qi J, Liu Y, Wang T, Chen H, Guan X, Zheng C, Zeng P. Mendelian randomization and genetic pleiotropy analysis for the connection between inflammatory bowel disease and Alzheimer's disease. Prog Neuropsychopharmacol Biol Psychiatry 2025; 136:111203. [PMID: 39579960 DOI: 10.1016/j.pnpbp.2024.111203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/18/2024] [Accepted: 11/19/2024] [Indexed: 11/25/2024]
Abstract
BACKGROUND The gut-microbiome-brain axis (GMBA) implies the connection between inflammatory bowel disease (IBD) and Alzheimer's disease (AD). We aimed to comprehensively explore the relation between IBD (and its subtypes) and AD, early-onset AD (EOAD) and late-onset AD (LOAD) from a genetic pleiotropy perspective. METHODS Relying on summary statistics (N = 472,868 for AD, 185,204 for EOAD, 191,061 for LOAD, 59,957 for IBD, 45,975 for CD, and 40,266 for UC), we first performed Mendelian Randomization to examine the causal association between IBD and AD by leveraging vertical pleiotropy. Then, we estimated global and local genetic correlations, followed by cross-trait association analysis to identify SNPs and genes with horizontal pleiotropy. Particularly, we utilized multi-trait colocalization analysis to assess the role of microbes in the common genetic etiology underlying the two types of diseases. Finally, we conducted functional enrichment analysis for pleiotropic genes. RESULTS We discovered suggestively causal relations between IBD (and its subtypes) and EOAD (ORIBD = 1.06 [1.01-1.11], ORCD = 1.05 [1.01-1.10], ORUC = 1.08 [1.01-1.15]) as well as between UC and LOAD (OR = 1.04 [1.01-1.08]), and discovered 44 local regions showing suggestively significant genetic correlations between IBD (and its subtypes) and AD (and EODA and LOAD). We further detected substantial genetic overlap, as characterized by 182 AD-associated, 3 EOAD-associated and 51 LOAD-associated pleiotropic SNPs as well as 291 pleiotropic genes. Pleiotropic genes more likely enriched in the GMBA-relevant tissues such as brain, intestine and esophagus. Moreover, we identified three microorganisms related to these disease pairs, including the Catenibacterium, Clostridia, and Prevotella species. CONCLUSION The suggestively causal associations and shared genetic basis between IBD and its subtypes with AD, EOAD and LOAD may commonly drive their co-occurrence, and gut microbes might partly explain the shared genetic etiology. Further studies are warranted to elaborate the possibly biological mechanisms underlying the two types of diseases.
Collapse
Affiliation(s)
- Yuxuan Wu
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Yu Yan
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Jike Qi
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Yuxin Liu
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Ting Wang
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Hao Chen
- Department of Neurology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou 221004, China
| | - Xinying Guan
- Department of Neurology, Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, Jiangsu 222002, China
| | - Chu Zheng
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; Jiangsu Engineering Research Center of Biological Data Mining and Healthcare Transformation, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China.
| | - Ping Zeng
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; Jiangsu Engineering Research Center of Biological Data Mining and Healthcare Transformation, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China.
| |
Collapse
|
46
|
Chen Q, Xu L, Lu C, Xue Y, Gong X, Shi Y, Wang C, Yu L. Prognostic significance of CDK1 expression in diffuse large B-Cell lymphoma. BMC Cancer 2025; 25:20. [PMID: 39773464 PMCID: PMC11705832 DOI: 10.1186/s12885-024-13388-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 12/23/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Diffuse large B-cell lymphoma (DLBCL) is the most common lymphoma in adult, characterized by uncontrolled cell proliferation and strong aggressiveness. Previous studies have found that cyclin-dependent kinase 1(CDK1) are related to tumor growth and metastasis. However, the role of CDK1 in DLBCL is exclusive. This study investigated the clinical implications and expression of CDK1 in DLBCL. METHODS Gene expression data for healthy subjects were sourced from the Genotype-Tissue Expression repository. Clinical details and survival statistics of patients with DLBCL were obtained from the Gene Expression Omnibus archive (GSE10846). Patients were categorized based on CDK1 expression levels, and differences in clinical outcomes between the groups were examined. Univariate and multivariate Cox regression analyses were used to ascertain whether CDK1 expression independently predicted DLBCL prognosis. The protein expression of CDK1 was gauged by immunohistochemistry. Additionally, we investigated the effect of CDK1 inhibition on DLBCL cell growth and death using the Cell Counting Kit-8 and flow cytometry. RESULTS In the control group, CDK1 expression was predominantly observed in the hematopoietic and reproductive systems. CDK1 levels in patients with DLBCL were notably elevated compared with those in controls. Significant differences were noted in the lactate dehydrogenase ratio and overall survival based on CDK1 expression. Statistical analyses confirmed that CDK1 was an independent predictor of DLBCL outcomes. Elevated CDK1 protein levels were observed in a significant number of DLBCL samples, in contrast to normal lymph node samples from individuals without lymphoma. The inhibitor Ro-3306 curtails DLBCL cell growth and enhances cell death in vitro. CONCLUSIONS Elevated CDK1 levels are correlated with poor prognosis in patients with DLBCL.
Collapse
MESH Headings
- Humans
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/metabolism
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lymphoma, Large B-Cell, Diffuse/mortality
- CDC2 Protein Kinase/metabolism
- CDC2 Protein Kinase/genetics
- Prognosis
- Male
- Female
- Middle Aged
- Aged
- Adult
- Biomarkers, Tumor/metabolism
- Biomarkers, Tumor/genetics
- Cell Proliferation
- Gene Expression Regulation, Neoplastic
- Cell Line, Tumor
Collapse
Affiliation(s)
- Qiuni Chen
- Department of Hematology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, 223300, Jiangsu Province, PR China
- Key Laboratory of Hematology, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, PR China
| | - Lei Xu
- Department of Hematology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, 223300, Jiangsu Province, PR China
- Key Laboratory of Hematology, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, PR China
| | - Chuanyang Lu
- Department of Hematology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, 223300, Jiangsu Province, PR China
- Key Laboratory of Hematology, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, PR China
| | - Yujie Xue
- Department of Pathology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, 223300, Jiangsu Province, PR China
| | - Xue Gong
- Department of Pathology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, 223300, Jiangsu Province, PR China
| | - Yuye Shi
- Department of Hematology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, 223300, Jiangsu Province, PR China
- Key Laboratory of Hematology, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, PR China
| | - Chunling Wang
- Department of Hematology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, 223300, Jiangsu Province, PR China.
- Key Laboratory of Hematology, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, PR China.
| | - Liang Yu
- Department of Hematology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, 223300, Jiangsu Province, PR China.
- Key Laboratory of Hematology, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, PR China.
| |
Collapse
|
47
|
Ogata T, Hattori A, Fukami M. SHOX and sex difference in height: a hypothesis. Endocr J 2025; 72:37-42. [PMID: 38987196 PMCID: PMC11778355 DOI: 10.1507/endocrj.ej24-0249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 06/17/2024] [Indexed: 07/12/2024] Open
Abstract
The mean height is taller in males than in females, except for early teens. In this regard, previous studies have revealed that (1) distribution of the mean adult heights in subjects with disorders accompanied by discordance between sex chromosome complement and bioactive sex steroids and in control subjects (the British height standards) indicates that, of the ~12.5 cm of sex difference in the mean adult height, ~9 cm is accounted for by the difference in the sex chromosome complement and the remaining ~3.5 cm is explained by the dimorphism in sex steroids (primarily due to the growth-promoting effect of gonadal androgens); (2) according to the infancy-childhood-puberty growth model, the sex difference in the childhood growth function produces height differences of ~1 cm in childhood and 8-10 cm at 18-20 years of age, whereas the sex difference in the pubertal growth function yields height difference of ~4.5 cm at 18-20 years of age; and (3) SHOX expression and methylation analyses using knee cartilage tissues and cultured chondrocytes have shown lower SHOX expression levels in female samples than in male samples and methylation patterns consistent with partial spreading of X-inactivation affecting SHOX in female samples. These findings suggest that small but persistent sex difference in SHOX expression dosage leads to the variation in the sex steroid independent childhood growth function, thereby yielding the sex difference in height which remains small in childhood but becomes obvious in adulthood.
Collapse
Affiliation(s)
- Tsutomu Ogata
- Departments of Pediatrics and Biochemistry, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan
- Department of Pediatrics, Hamamatsu Medical Center, Hamamatsu 432-8580, Japan
| | - Atsushi Hattori
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| | - Maki Fukami
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| |
Collapse
|
48
|
Lin J, Zhang J, Ma L, Fang H, Ma R, Groneck C, Filippova GN, Deng X, Kinoshita C, Young JE, Ma W, Disteche CM, Berletch JB. KDM6A facilitates Xist upregulation at the onset of X inactivation. Biol Sex Differ 2025; 16:1. [PMID: 39754175 PMCID: PMC11699772 DOI: 10.1186/s13293-024-00683-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 12/09/2024] [Indexed: 01/06/2025] Open
Abstract
BACKGROUND X chromosome inactivation (XCI) is a female-specific process in which one X chromosome is silenced to balance X-linked gene expression between the sexes. XCI is initiated in early development by upregulation of the lncRNA Xist on the future inactive X (Xi). A subset of X-linked genes escape silencing and thus have higher expression in females, suggesting female-specific functions. One of these genes is the highly conserved gene Kdm6a, which encodes a histone demethylase that removes methyl groups at H3K27 to facilitate gene expression. KDM6A mutations have been implicated in congenital disorders such as Kabuki Syndrome, as well as in sex differences in development and cancer. METHODS Kdm6a was knocked out (KO) using CRISPR/Cas9 gene editing in hybrid female mouse embryonic stem (ES) cells derived either from a 129 × Mus castaneus (cast) cross or a BL6 x cast cross. In one of the lines a transcriptional stop signal inserted in Tsix results in completely skewed X silencing upon differentiation. The effects of both homozygous and heterozygous Kdm6a KO on Xist expression during the onset of XCI were measured by RT-PCR and RNA-FISH. Changes in gene expression and in H3K27me3 enrichment were investigated using allele-specific RNA-seq and Cut&Run, respectively. KDM6A binding to the Xist gene was characterized by Cut&Run. RESULTS We observed impaired upregulation of Xist and reduced coating of the Xi during early stages of differentiation in Kdm6a KO cells, both homozygous and heterozygous, suggesting a threshold effect of KDM6A. This was associated with aberrant overexpression of genes from the Xi after differentiation, indicating loss of X inactivation potency. Consistent with KDM6A having a direct role in Xist regulation, we found that the histone demethylase binds to the Xist promoter and KO cells show an increase in H3K27me3 at Xist, consistent with reduced expression. CONCLUSIONS These results reveal a novel female-specific role for the X-linked histone demethylase, KDM6A in the initiation of XCI through histone demethylase-dependent activation of Xist during early differentiation. X chromosome inactivation is a female-specific mechanism that evolved to balance sex-linked gene dosage between females (XX) and males (XY) by silencing one X chromosome in females. X inactivation begins with the upregulation of the long noncoding RNA Xist on the future inactive X chromosome. While most genes become silenced on the inactive X chromosome some genes escape inactivation and thus have higher expression in females compared to males, suggesting that escape genes may have female-specific functions. One such gene encodes the histone demethylase KDM6A which function is to turn on gene expression by removing repressive histone modifications. In this study, we investigated the role of KDM6A in the regulation of Xist expression during the onset of X inactivation. We found that KDM6A binds to the Xist gene to remove repressive histone marks and facilitate its expression in early development. Indeed, depletion of KDM6A prevents upregulation of Xist due to abnormal persistence of repressive histone modifications. In turn, this results in aberrant overexpression of genes from the inactive X chromosome. Our findings point to a novel mechanism of Xist regulation during the initiation of X inactivation, which may lead to new avenues of treatment to alleviate congenital disorders such as Kabuki syndrome and sex-biased immune disorders where X-linked gene dosage is perturbed.
Collapse
Affiliation(s)
- Josephine Lin
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Jinli Zhang
- Department of Statistics, University of California Riverside, Riverside, CA, 92521, USA
| | - Li Ma
- Department of Microbiology, Immunology & Cell Biology, University of West Virginia, Morgantown, WV, 26506, USA
| | - He Fang
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Rui Ma
- Department of Statistics, University of California Riverside, Riverside, CA, 92521, USA
| | - Camille Groneck
- Department of Biochemistry, University of California Riverside, Riverside, CA, 92521, USA
| | - Galina N Filippova
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Xinxian Deng
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Chizuru Kinoshita
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Jessica E Young
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Wenxiu Ma
- Department of Statistics, University of California Riverside, Riverside, CA, 92521, USA.
| | - Christine M Disteche
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Washington, Seattle, WA, 98195, USA.
- Department of Medicine, School of Medicine, University of Washington, Seattle, WA, 98195, USA.
| | - Joel B Berletch
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
49
|
Seto M, Clifton M, Gomez ML, Coughlan G, Gifford KA, Jefferson AL, De Jager PL, Bennett DA, Wang Y, Barnes LL, Schneider JA, Hohman TJ, Buckley RF, Dumitrescu L. Sex-specific Associations of Gene Expression with Alzheimer's Disease Neuropathology and Ante-mortem Cognitive Performance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.02.631098. [PMID: 39803447 PMCID: PMC11722314 DOI: 10.1101/2025.01.02.631098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/21/2025]
Abstract
The biological mechanisms underlying women's increased Alzheimer's disease (AD) prevalence remain undefined. Previous case/control studies have identified sex-biased molecular pathways, but sex-specific relationships between gene expression and AD endophenotypes, particularly sex chromosomes, are underexplored. With bulk transcriptomic data across 3 brain regions from 767 decedents, we investigated sex-specific associations between gene expression and post-mortem β-amyloid and tau as well as antemortem longitudinal cognition. Of 23,118 significant gene associations, 10% were significant in one sex and not the other (sex-specific). Most sex-specific gene associations were identified in females (73%) and associated with tau tangles and longitudinal cognition (90%). Four X-linked genes, MCF2, HDAC8, FTX, and SLC10A3, demonstrated significant sex differences in their associations with AD endophenotypes (i.e., significant sex x gene interaction). Our results also uncovered sex-specific biological pathways, including a female-specific role of neuroinflammation and neuronal development, reinforcing the potential for sex-aware analyses to enhance precision medicine approaches in AD.
Collapse
Affiliation(s)
- Mabel Seto
- Department of Neurology, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
- Center for Alzheimer's Research and Treatment, Department of Neurology, Brigham and Women's Hospital/Harvard Medical School, Boston, MA, USA
| | - Michelle Clifton
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Melisa Lara Gomez
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Gillian Coughlan
- Department of Neurology, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
- Center for Alzheimer's Research and Treatment, Department of Neurology, Brigham and Women's Hospital/Harvard Medical School, Boston, MA, USA
| | - Katherine A. Gifford
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Angela L. Jefferson
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Philip L. De Jager
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Medical Center, New York, NY, USA
- Cell Circuits Program, Broad Institute, Cambridge, MA, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Yanling Wang
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Lisa L. Barnes
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Julie A Schneider
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Timothy J. Hohman
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Rachel F. Buckley
- Department of Neurology, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
- Center for Alzheimer's Research and Treatment, Department of Neurology, Brigham and Women's Hospital/Harvard Medical School, Boston, MA, USA
- Melbourne School of Psychological Sciences, University of Melbourne, Melbourne, Australia
| | - Logan Dumitrescu
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
50
|
Mosti F, Hoye ML, Escobar-Tomlienovich CF, Silver DL. Multi-modal investigation reveals pathogenic features of diverse DDX3X missense mutations. PLoS Genet 2025; 21:e1011555. [PMID: 39836689 PMCID: PMC11771946 DOI: 10.1371/journal.pgen.1011555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 01/27/2025] [Accepted: 12/27/2024] [Indexed: 01/23/2025] Open
Abstract
De novo mutations in the RNA binding protein DDX3X cause neurodevelopmental disorders including DDX3X syndrome and autism spectrum disorder. Amongst ~200 mutations identified to date, half are missense. While DDX3X loss of function is known to impair neural cell fate, how the landscape of missense mutations impacts neurodevelopment is almost entirely unknown. Here, we integrate transcriptomics, proteomics, and live imaging to demonstrate clinically diverse DDX3X missense mutations perturb neural development via distinct cellular and molecular mechanisms. Using mouse primary neural progenitors, we investigate four recurrently mutated DDX3X missense variants, spanning clinically severe (2) to mild (2). While clinically severe mutations impair neurogenesis, mild mutations have only a modest impact on cell fate. Moreover, expression of severe mutations leads to profound neuronal death. Using a proximity labeling screen in neural progenitors, we discover DDX3X missense variants have unique protein interactors. We observe notable overlap amongst severe mutations, suggesting common mechanisms underlying altered cell fate and survival. Transcriptomic analysis and subsequent cellular investigation highlights new pathways associated with DDX3X missense variants, including upregulated DNA Damage Response. Notably, clinically severe mutations exhibit excessive DNA damage in neurons, associated with increased cytoplasmic DNA:RNA hybrids and formation of stress granules. These findings highlight aberrant RNA metabolism and DNA damage in DDX3X-mediated neuronal cell death. In sum our findings reveal new mechanisms by which clinically distinct DDX3X missense mutations differentially impair neurodevelopment.
Collapse
Affiliation(s)
- Federica Mosti
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Neurobiology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Mariah L. Hoye
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Carla F. Escobar-Tomlienovich
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Neurobiology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Debra L. Silver
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Neurobiology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Cell Biology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Duke Institute for Brain Sciences and Duke Regeneration Center, Duke University School of Medicine, Durham, North Carolina, United States of America
| |
Collapse
|