Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54.270
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 40: 75-94, 2022 04 26.
Article in English | MEDLINE | ID: mdl-34985929

ABSTRACT

Strong epidemiological evidence now exists that sex is an important biologic variable in immunity. Recent studies, for example, have revealed that sex differences are associated with the severity of symptoms and mortality due to coronavirus disease 2019 (COVID-19). Despite this evidence, much remains to be learned about the mechanisms underlying associations between sex differences and immune-mediated conditions. A growing body of experimental data has made significant inroads into understanding sex-influenced immune responses. As physicians seek to provide more targeted patient care, it is critical to understand how sex-defining factors (e.g., chromosomes, gonadal hormones) alter immune responses in health and disease. In this review, we highlight recent insights into sex differences in autoimmunity; virus infection, specifically severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection; and cancer immunotherapy. A deeper understanding of underlying mechanisms will allow the development of a sex-based approach to disease screening and treatment.


Subject(s)
COVID-19 , SARS-CoV-2 , Animals , Female , Humans , Male , Sex Characteristics , Sex Factors
2.
Cell ; 187(6): 1316-1326, 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38490173

ABSTRACT

Understanding sex-related variation in health and illness requires rigorous and precise approaches to revealing underlying mechanisms. A first step is to recognize that sex is not in and of itself a causal mechanism; rather, it is a classification system comprising a set of categories, usually assigned according to a range of varying traits. Moving beyond sex as a system of classification to working with concrete and measurable sex-related variables is necessary for precision. Whether and how these sex-related variables matter-and what patterns of difference they contribute to-will vary in context-specific ways. Second, when researchers incorporate these sex-related variables into research designs, rigorous analytical methods are needed to allow strongly supported conclusions. Third, the interpretation and reporting of sex-related variation require care to ensure that basic and preclinical research advance health equity for all.


Subject(s)
Biomedical Research , Health Equity , Sex , Humans
3.
Cell ; 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39168126

ABSTRACT

Xp11 translocation renal cell carcinoma (tRCC) is a rare, female-predominant cancer driven by a fusion between the transcription factor binding to IGHM enhancer 3 (TFE3) gene on chromosome Xp11.2 and a partner gene on either chromosome X (chrX) or an autosome. It remains unknown what types of rearrangements underlie TFE3 fusions, whether fusions can arise from both the active (chrXa) and inactive X (chrXi) chromosomes, and whether TFE3 fusions from chrXi translocations account for the female predominance of tRCC. To address these questions, we performed haplotype-specific analyses of chrX rearrangements in tRCC whole genomes. We show that TFE3 fusions universally arise as reciprocal translocations and that oncogenic TFE3 fusions can arise from chrXi:autosomal translocations. Female-specific chrXi:autosomal translocations result in a 2:1 female-to-male ratio of TFE3 fusions involving autosomal partner genes and account for the female predominance of tRCC. Our results highlight how X chromosome genetics constrains somatic chrX alterations and underlies cancer sex differences.

4.
Cell ; 186(10): 2044-2061, 2023 05 11.
Article in English | MEDLINE | ID: mdl-37172561

ABSTRACT

Phenotypic sex-based differences exist for many complex traits. In other cases, phenotypes may be similar, but underlying biology may vary. Thus, sex-aware genetic analyses are becoming increasingly important for understanding the mechanisms driving these differences. To this end, we provide a guide outlining the current best practices for testing various models of sex-dependent genetic effects in complex traits and disease conditions, noting that this is an evolving field. Insights from sex-aware analyses will not only teach us about the biology of complex traits but also aid in achieving the goals of precision medicine and health equity for all.


Subject(s)
Models, Genetic , Sex Characteristics , Animals , Female , Male , Multifactorial Inheritance , Phenotype , Quality Control , Genome-Wide Association Study , Guidelines as Topic , Gene-Environment Interaction , Humans
5.
Cell ; 2023 Jun 22.
Article in English | MEDLINE | ID: mdl-37385248

ABSTRACT

Certain cancer types afflict female and male patients disproportionately. The reasons include differences in male/female physiology, effect of sex hormones, risk behavior, environmental exposures, and genetics of the sex chromosomes X and Y. Loss of Y (LOY) is common in peripheral blood cells in aging men, and this phenomenon is associated with several diseases. However, the frequency and role of LOY in tumors is little understood. Here, we present a comprehensive catalog of LOY in >5,000 primary tumors from male patients in the TCGA. We show that LOY rates vary by tumor type and provide evidence for LOY being either a passenger or driver event depending on context. LOY in uveal melanoma specifically is associated with age and survival and is an independent predictor of poor outcome. LOY creates common dependencies on DDX3X and EIF1AX in male cell lines, suggesting that LOY generates unique vulnerabilities that could be therapeutically exploited.

6.
Cell ; 186(13): 2748-2764.e22, 2023 06 22.
Article in English | MEDLINE | ID: mdl-37267948

ABSTRACT

Ferroptosis, a cell death process driven by iron-dependent phospholipid peroxidation, has been implicated in various diseases. There are two major surveillance mechanisms to suppress ferroptosis: one mediated by glutathione peroxidase 4 (GPX4) that catalyzes the reduction of phospholipid peroxides and the other mediated by enzymes, such as FSP1, that produce metabolites with free radical-trapping antioxidant activity. In this study, through a whole-genome CRISPR activation screen, followed by mechanistic investigation, we identified phospholipid-modifying enzymes MBOAT1 and MBOAT2 as ferroptosis suppressors. MBOAT1/2 inhibit ferroptosis by remodeling the cellular phospholipid profile, and strikingly, their ferroptosis surveillance function is independent of GPX4 or FSP1. MBOAT1 and MBOAT2 are transcriptionally upregulated by sex hormone receptors, i.e., estrogen receptor (ER) and androgen receptor (AR), respectively. A combination of ER or AR antagonist with ferroptosis induction significantly inhibited the growth of ER+ breast cancer and AR+ prostate cancer, even when tumors were resistant to single-agent hormonal therapies.


Subject(s)
Ferroptosis , Male , Humans , Phospholipid Hydroperoxide Glutathione Peroxidase , Lipid Peroxidation , Peroxides , Phospholipids
7.
Cell ; 185(10): 1619-1622, 2022 05 12.
Article in English | MEDLINE | ID: mdl-35561661

ABSTRACT

Progress in studying sex as a biological variable (SABV) is slow, and the influence of gendered effects of the social environment on biology is largely unknown. Yet incorporating these concepts into basic science research will enhance our understanding of human health and disease. We provide steps to move this process forward.


Subject(s)
Biomedical Research , Female , Humans , Male , Precision Medicine , Sex Characteristics , Sex Factors , Women's Health
8.
Cell ; 185(4): 654-671.e22, 2022 02 17.
Article in English | MEDLINE | ID: mdl-35065713

ABSTRACT

Sex hormones exert a profound influence on gendered behaviors. How individual sex hormone-responsive neuronal populations regulate diverse sex-typical behaviors is unclear. We performed orthogonal, genetically targeted sequencing of four estrogen receptor 1-expressing (Esr1+) populations and identified 1,415 genes expressed differentially between sexes or estrous states. Unique subsets of these genes were distributed across all 137 transcriptomically defined Esr1+ cell types, including estrous stage-specific ones, that comprise the four populations. We used differentially expressed genes labeling single Esr1+ cell types as entry points to functionally characterize two such cell types, BNSTprTac1/Esr1 and VMHvlCckar/Esr1. We observed that these two cell types, but not the other Esr1+ cell types in these populations, are essential for sex recognition in males and mating in females, respectively. Furthermore, VMHvlCckar/Esr1 cell type projections are distinct from those of other VMHvlEsr1 cell types. Together, projection and functional specialization of dimorphic cell types enables sex hormone-responsive populations to regulate diverse social behaviors.


Subject(s)
Estrous Cycle/genetics , Gene Expression Regulation , Sex Characteristics , Sexual Behavior, Animal/physiology , Aggression , Animals , Aromatase/metabolism , Autistic Disorder/genetics , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Gene Expression Profiling , HEK293 Cells , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neurons/metabolism , Social Behavior
9.
Cell ; 184(7): 1790-1803.e17, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33735607

ABSTRACT

The long non-coding RNA (lncRNA) XIST establishes X chromosome inactivation (XCI) in female cells in early development and thereafter is thought to be largely dispensable. Here, we show XIST is continually required in adult human B cells to silence a subset of X-linked immune genes such as TLR7. XIST-dependent genes lack promoter DNA methylation and require continual XIST-dependent histone deacetylation. XIST RNA-directed proteomics and CRISPRi screen reveal distinctive somatic cell-type-specific XIST complexes and identify TRIM28 that mediates Pol II pausing at promoters of X-linked genes in B cells. Single-cell transcriptome data of female patients with either systemic lupus erythematosus or COVID-19 infection revealed XIST dysregulation, reflected by escape of XIST-dependent genes, in CD11c+ atypical memory B cells (ABCs). XIST inactivation with TLR7 agonism suffices to promote isotype-switched ABCs. These results indicate cell-type-specific diversification and function for lncRNA-protein complexes and suggest expanded roles for XIST in sex-differences in biology and medicine.


Subject(s)
B-Lymphocytes/immunology , COVID-19 , Lupus Erythematosus, Systemic , RNA, Long Noncoding/physiology , Toll-Like Receptor 7/immunology , X Chromosome Inactivation , COVID-19/genetics , COVID-19/immunology , Cell Line , DNA Methylation , Female , Gene Silencing , Humans , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology
10.
Cell ; 183(4): 875-889.e17, 2020 11 12.
Article in English | MEDLINE | ID: mdl-33035453

ABSTRACT

Banyan trees are distinguished by their extraordinary aerial roots. The Ficus genus includes species that have evolved a species-specific mutualism system with wasp pollinators. We sequenced genomes of the Chinese banyan tree, F. microcarpa, and a species lacking aerial roots, F. hispida, and one wasp genome coevolving with F. microcarpa, Eupristina verticillata. Comparative analysis of the two Ficus genomes revealed dynamic karyotype variation associated with adaptive evolution. Copy number expansion of auxin-related genes from duplications and elevated auxin production are associated with aerial root development in F. microcarpa. A male-specific AGAMOUS paralog, FhAG2, was identified as a candidate gene for sex determination in F. hispida. Population genomic analyses of Ficus species revealed genomic signatures of morphological and physiological coadaptation with their pollinators involving terpenoid- and benzenoid-derived compounds. These three genomes offer insights into and genomic resources for investigating the geneses of aerial roots, monoecy and dioecy, and codiversification in a symbiotic system.


Subject(s)
Biological Evolution , Ficus/genetics , Genome, Plant , Pollination/physiology , Trees/genetics , Wasps/physiology , Animals , Chromosomes, Plant/genetics , DNA Transposable Elements/genetics , Female , Gene Expression Profiling , Gene Expression Regulation, Plant , Genes, Plant , Indoleacetic Acids/metabolism , Molecular Sequence Annotation , Phylogeny , Plant Roots/growth & development , Segmental Duplications, Genomic/genetics , Sex Chromosomes/genetics , Volatile Organic Compounds/analysis
11.
Cell ; 177(2): 326-338.e16, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30879787

ABSTRACT

Crossing over is a nearly universal feature of sexual reproduction. Here, analysis of crossover numbers on a per-chromosome and per-nucleus basis reveals a fundamental, evolutionarily conserved feature of meiosis: within individual nuclei, crossover frequencies covary across different chromosomes. This effect results from per-nucleus covariation of chromosome axis lengths. Crossovers can promote evolutionary adaptation. However, the benefit of creating favorable new allelic combinations must outweigh the cost of disrupting existing favorable combinations. Covariation concomitantly increases the frequencies of gametes with especially high, or especially low, numbers of crossovers, and thus might concomitantly enhance the benefits of crossing over while reducing its costs. A four-locus population genetic model suggests that such an effect can pertain in situations where the environment fluctuates: hyper-crossover gametes are advantageous when the environment changes while hypo-crossover gametes are advantageous in periods of environmental stasis. These findings reveal a new feature of the basic meiotic program and suggest a possible adaptive advantage.


Subject(s)
Crossing Over, Genetic/genetics , Crossing Over, Genetic/physiology , Animals , Cell Nucleus , Chromosome Segregation , Chromosomes/genetics , Chromosomes/physiology , Computer Simulation , Female , Genetics, Population/methods , Homologous Recombination/genetics , Humans , Solanum lycopersicum/genetics , Male , Meiosis/genetics , Recombination, Genetic/genetics , Synaptonemal Complex
12.
Cell ; 175(6): 1701-1715.e16, 2018 11 29.
Article in English | MEDLINE | ID: mdl-30449622

ABSTRACT

While many genetic variants have been associated with risk for human diseases, how these variants affect gene expression in various cell types remains largely unknown. To address this gap, the DICE (database of immune cell expression, expression quantitative trait loci [eQTLs], and epigenomics) project was established. Considering all human immune cell types and conditions studied, we identified cis-eQTLs for a total of 12,254 unique genes, which represent 61% of all protein-coding genes expressed in these cell types. Strikingly, a large fraction (41%) of these genes showed a strong cis-association with genotype only in a single cell type. We also found that biological sex is associated with major differences in immune cell gene expression in a highly cell-specific manner. These datasets will help reveal the effects of disease risk-associated genetic polymorphisms on specific immune cell types, providing mechanistic insights into how they might influence pathogenesis (https://dice-database.org).


Subject(s)
Gene Expression Regulation/immunology , Genotype , Polymorphism, Single Nucleotide/immunology , Quantitative Trait Loci/immunology , Sex Characteristics , Adolescent , Adult , Female , Gene Expression Profiling , Genome-Wide Association Study , Humans , Male , Middle Aged
13.
Cell ; 172(3): 500-516.e16, 2018 01 25.
Article in English | MEDLINE | ID: mdl-29275859

ABSTRACT

Microglia are embryonically seeded macrophages that contribute to brain development, homeostasis, and pathologies. It is thus essential to decipher how microglial properties are temporally regulated by intrinsic and extrinsic factors, such as sexual identity and the microbiome. Here, we found that microglia undergo differentiation phases, discernable by transcriptomic signatures and chromatin accessibility landscapes, which can diverge in adult males and females. Remarkably, the absence of microbiome in germ-free mice had a time and sexually dimorphic impact both prenatally and postnatally: microglia were more profoundly perturbed in male embryos and female adults. Antibiotic treatment of adult mice triggered sexually biased microglial responses revealing both acute and long-term effects of microbiota depletion. Finally, human fetal microglia exhibited significant overlap with the murine transcriptomic signature. Our study shows that microglia respond to environmental challenges in a sex- and time-dependent manner from prenatal stages, with major implications for our understanding of microglial contributions to health and disease.


Subject(s)
Germ-Free Life , Microbiota , Microglia/cytology , Prenatal Exposure Delayed Effects/microbiology , Transcriptome , Animals , Brain/cytology , Brain/embryology , Brain/metabolism , Cell Differentiation , Cells, Cultured , Chromatin Assembly and Disassembly , Female , Humans , Male , Mice , Mice, Inbred C57BL , Microglia/metabolism , Pregnancy , Sex Factors
14.
Immunity ; 56(11): 2472-2491, 2023 Nov 14.
Article in English | MEDLINE | ID: mdl-37967530

ABSTRACT

Immune responses to antigens, including innocuous, self, tumor, microbial, and vaccine antigens, differ between males and females. The quest to uncover the mechanisms for biological sex differences in the immune system has intensified, with considerable literature pointing toward sex hormonal influences on immune cell function. Sex steroids, including estrogens, androgens, and progestins, have profound effects on immune function. As such, drastic changes in sex steroid concentrations that occur with aging (e.g., after puberty or during the menopause transition) or pregnancy impact immune responses and the pathogenesis of immune-related diseases. The effect of sex steroids on immunity involves both the concentration of the ligand and the density and distribution of genomic and nongenomic receptors that serve as transcriptional regulators of immune cellular responses to affect autoimmunity, allergy, infectious diseases, cancers, and responses to vaccines. The next frontier will be harnessing these effects of sex steroids to improve therapeutic outcomes.


Subject(s)
Gonadal Steroid Hormones , Neoplasms , Pregnancy , Female , Male , Humans , Estrogens/pharmacology , Estrogens/physiology , Progestins , Androgens/pharmacology , Steroids , Immunity , Sex Characteristics
15.
Immunity ; 56(12): 2773-2789.e8, 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-37992711

ABSTRACT

Although the gut microbiota can influence central nervous system (CNS) autoimmune diseases, the contribution of the intestinal epithelium to CNS autoimmunity is less clear. Here, we showed that intestinal epithelial dopamine D2 receptors (IEC DRD2) promoted sex-specific disease progression in an animal model of multiple sclerosis. Female mice lacking Drd2 selectively in intestinal epithelial cells showed a blunted inflammatory response in the CNS and reduced disease progression. In contrast, overexpression or activation of IEC DRD2 by phenylethylamine administration exacerbated disease severity. This was accompanied by altered lysozyme expression and gut microbiota composition, including reduced abundance of Lactobacillus species. Furthermore, treatment with N2-acetyl-L-lysine, a metabolite derived from Lactobacillus, suppressed microglial activation and neurodegeneration. Taken together, our study indicates that IEC DRD2 hyperactivity impacts gut microbial abundances and increases susceptibility to CNS autoimmune diseases in a female-biased manner, opening up future avenues for sex-specific interventions of CNS autoimmune diseases.


Subject(s)
Autoimmune Diseases of the Nervous System , Multiple Sclerosis , Male , Female , Mice , Animals , Multiple Sclerosis/metabolism , Disease Models, Animal , Signal Transduction , Disease Progression , Receptors, Dopamine
16.
Cell ; 171(2): 287-304.e15, 2017 Oct 05.
Article in English | MEDLINE | ID: mdl-28985561

ABSTRACT

The evolution of land flora transformed the terrestrial environment. Land plants evolved from an ancestral charophycean alga from which they inherited developmental, biochemical, and cell biological attributes. Additional biochemical and physiological adaptations to land, and a life cycle with an alternation between multicellular haploid and diploid generations that facilitated efficient dispersal of desiccation tolerant spores, evolved in the ancestral land plant. We analyzed the genome of the liverwort Marchantia polymorpha, a member of a basal land plant lineage. Relative to charophycean algae, land plant genomes are characterized by genes encoding novel biochemical pathways, new phytohormone signaling pathways (notably auxin), expanded repertoires of signaling pathways, and increased diversity in some transcription factor families. Compared with other sequenced land plants, M. polymorpha exhibits low genetic redundancy in most regulatory pathways, with this portion of its genome resembling that predicted for the ancestral land plant. PAPERCLIP.


Subject(s)
Biological Evolution , Embryophyta/genetics , Genome, Plant , Marchantia/genetics , Adaptation, Biological , Embryophyta/physiology , Gene Expression Regulation, Plant , Marchantia/physiology , Molecular Sequence Annotation , Signal Transduction , Transcription, Genetic
17.
Cell ; 171(5): 1176-1190.e17, 2017 Nov 16.
Article in English | MEDLINE | ID: mdl-29107332

ABSTRACT

The medial amygdala (MeA) plays a critical role in processing species- and sex-specific signals that trigger social and defensive behaviors. However, the principles by which this deep brain structure encodes social information is poorly understood. We used a miniature microscope to image the Ca2+ dynamics of large neural ensembles in awake behaving mice and tracked the responses of MeA neurons over several months. These recordings revealed spatially intermingled subsets of MeA neurons with distinct temporal dynamics. The encoding of social information in the MeA differed between males and females and relied on information from both individual cells and neuronal populations. By performing long-term Ca2+ imaging across different social contexts, we found that sexual experience triggers lasting and sex-specific changes in MeA activity, which, in males, involve signaling by oxytocin. These findings reveal basic principles underlying the brain's representation of social information and its modulation by intrinsic and extrinsic factors.


Subject(s)
Amygdala/physiology , Neurons/cytology , Wakefulness , Amygdala/cytology , Animals , Behavior, Animal , Cues , Endoscopy/methods , Female , Male , Mice , Microscopy/methods , Oxytocin/physiology , Sex Characteristics , Sexual Behavior, Animal , Social Behavior
18.
Cell ; 169(3): 457-469.e13, 2017 04 20.
Article in English | MEDLINE | ID: mdl-28431246

ABSTRACT

Fat metabolism has been linked to fertility and reproductive adaptation in animals and humans, and environmental sex determination potentially plays a role in the process. To investigate the impact of fatty acids (FA) on sex determination and reproductive development, we examined and observed an impact of FA synthesis and mobilization by lipolysis in somatic tissues on oocyte fate in Caenorhabditis elegans. The subsequent genetic analysis identified ACS-4, an acyl-CoA synthetase and its FA-CoA product, as key germline factors that mediate the role of FA in promoting oocyte fate through protein myristoylation. Further tests indicated that ACS-4-dependent protein myristoylation perceives and translates the FA level into regulatory cues that modulate the activities of MPK-1/MAPK and key factors in the germline sex-determination pathway. These findings, including a similar role of ACS-4 in a male/female species, uncover a likely conserved mechanism by which FA, an environmental factor, regulates sex determination and reproductive development.


Subject(s)
Acetate-CoA Ligase/metabolism , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/physiology , Fatty Acids/metabolism , Myristic Acid/metabolism , Protein Processing, Post-Translational , Sex Determination Processes , Acetate-CoA Ligase/genetics , Animals , Caenorhabditis elegans Proteins/genetics , Mutation , Oocytes/metabolism
19.
Genes Dev ; 38(13-14): 585-596, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39048311

ABSTRACT

The X and Y chromosomes play important roles outside of human reproduction; namely, their potential contribution to human sex biases in physiology and disease. While sex biases are often thought to be an effect of hormones and environmental exposures, genes encoded on the sex chromosomes also play a role. Seventeen homologous gene pairs exist on the X and Y chromosomes whose proteins have critical functions in biology, from direct regulation of transcription and translation to intercellular signaling and formation of extracellular structures. In this review, we cover the current understanding of several of these sex chromosome-encoded protein homologs that are involved in transcription and chromatin regulation: SRY/SOX3, ZFX/ZFY, KDM5C/KDM5D, UTX/UTY, and TBL1X/TBL1Y. Their mechanisms of gene regulation are discussed, including any redundancies or divergent roles of the X- and Y-chromosome homologs. Additionally, we discuss associated diseases related to these proteins and any sex biases that exist therein in an effort to drive further research into how these pairs contribute to sexually dimorphic gene regulation in health and disease.


Subject(s)
Gene Expression Regulation , Humans , Gene Expression Regulation/genetics , Animals , Histone Demethylases/metabolism , Histone Demethylases/genetics , Chromosomes, Human, Y/genetics , Chromosomes, Human, X/genetics , Sex Characteristics , Transducin/genetics , Transducin/metabolism , Sex Chromosomes/genetics , Female , Nuclear Proteins , Minor Histocompatibility Antigens
20.
Genes Dev ; 38(3-4): 115-130, 2024 03 22.
Article in English | MEDLINE | ID: mdl-38383062

ABSTRACT

H3K9 trimethylation (H3K9me3) plays emerging roles in gene regulation, beyond its accumulation on pericentric constitutive heterochromatin. It remains a mystery why and how H3K9me3 undergoes dynamic regulation in male meiosis. Here, we identify a novel, critical regulator of H3K9 methylation and spermatogenic heterochromatin organization: the germline-specific protein ATF7IP2 (MCAF2). We show that in male meiosis, ATF7IP2 amasses on autosomal and X-pericentric heterochromatin, spreads through the entirety of the sex chromosomes, and accumulates on thousands of autosomal promoters and retrotransposon loci. On the sex chromosomes, which undergo meiotic sex chromosome inactivation (MSCI), the DNA damage response pathway recruits ATF7IP2 to X-pericentric heterochromatin, where it facilitates the recruitment of SETDB1, a histone methyltransferase that catalyzes H3K9me3. In the absence of ATF7IP2, male germ cells are arrested in meiotic prophase I. Analyses of ATF7IP2-deficient meiosis reveal the protein's essential roles in the maintenance of MSCI, suppression of retrotransposons, and global up-regulation of autosomal genes. We propose that ATF7IP2 is a downstream effector of the DDR pathway in meiosis that coordinates the organization of heterochromatin and gene regulation through the spatial regulation of SETDB1-mediated H3K9me3 deposition.


Subject(s)
Heterochromatin , Histones , Male , Germ Cells/metabolism , Heterochromatin/genetics , Heterochromatin/metabolism , Histones/metabolism , Meiosis/genetics , Methylation , Animals , Mice
SELECTION OF CITATIONS
SEARCH DETAIL