Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Nucleic Acids Res ; 52(5): 2306-2322, 2024 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-38142439

RESUMEN

Spermatogonial stem cells functionality reside in the slow-cycling and heterogeneous undifferentiated spermatogonia cell population. This pool of cells supports lifelong fertility in adult males by balancing self-renewal and differentiation to produce haploid gametes. However, the molecular mechanisms underpinning long-term stemness of undifferentiated spermatogonia during adulthood remain unclear. Here, we discover that an epigenetic regulator, Polycomb repressive complex 1 (PRC1), shields adult undifferentiated spermatogonia from differentiation, maintains slow cycling, and directs commitment to differentiation during steady-state spermatogenesis in adults. We show that PRC2-mediated H3K27me3 is an epigenetic hallmark of adult undifferentiated spermatogonia. Indeed, spermatogonial differentiation is accompanied by a global loss of H3K27me3. Disruption of PRC1 impairs global H3K27me3 deposition, leading to precocious spermatogonial differentiation. Therefore, PRC1 directs PRC2-H3K27me3 deposition to maintain the self-renewing state of undifferentiated spermatogonia. Importantly, in contrast to its role in other tissue stem cells, PRC1 negatively regulates the cell cycle to maintain slow cycling of undifferentiated spermatogonia. Our findings have implications for how epigenetic regulators can be tuned to regulate the stem cell potential, cell cycle and differentiation to ensure lifelong fertility in adult males.


Asunto(s)
Complejo Represivo Polycomb 1 , Complejo Represivo Polycomb 2 , Espermatogénesis , Células Madre , Animales , Femenino , Masculino , Ratones , Diferenciación Celular , Histonas/metabolismo , Complejo Represivo Polycomb 1/genética , Complejo Represivo Polycomb 1/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Espermatogonias , Células Madre/citología , Células Madre/metabolismo
2.
Nucleic Acids Res ; 51(13): 6668-6683, 2023 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-37283086

RESUMEN

Sperm chromatin retains small amounts of histones, and chromatin states of sperm mirror gene expression programs of the next generation. However, it remains largely unknown how paternal epigenetic information is transmitted through sperm chromatin. Here, we present a novel mouse model of paternal epigenetic inheritance, in which deposition of Polycomb repressive complex 2 (PRC2) mediated-repressive H3K27me3 is attenuated in the paternal germline. By applying modified methods of assisted reproductive technology using testicular sperm, we rescued infertility of mice missing Polycomb protein SCML2, which regulates germline gene expression by establishing H3K27me3 on bivalent promoters with other active marks H3K4me2/3. We profiled epigenomic states (H3K27me3 and H3K4me3) of testicular sperm and epididymal sperm, demonstrating that the epididymal pattern of the sperm epigenome is already established in testicular sperm and that SCML2 is required for this process. In F1 males of X-linked Scml2-knockout mice, which have a wild-type genotype, gene expression is dysregulated in the male germline during spermiogenesis. These dysregulated genes are targets of SCML2-mediated H3K27me3 in F0 sperm. Further, dysregulation of gene expression was observed in the mutant-derived wild-type F1 preimplantation embryos. Together, we present functional evidence that the classic epigenetic regulator Polycomb mediates paternal epigenetic inheritance through sperm chromatin.


Asunto(s)
Cromatina , Epigénesis Genética , Animales , Masculino , Ratones , Cromatina/genética , Epigenómica , Histonas/genética , Histonas/metabolismo , Ratones Noqueados , Proteínas del Grupo Polycomb/genética , Proteínas del Grupo Polycomb/metabolismo , Semen/metabolismo , Espermatozoides/metabolismo
3.
Genes Dev ; 31(16): 1693-1703, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28924034

RESUMEN

During spermatogenesis, a large number of germline genes essential for male fertility are coordinately activated. However, it remains unknown how timely activation of this group of germline genes is accomplished. Here we show that Polycomb-repressive complex 1 (PRC1) directs timely activation of germline genes during spermatogenesis. Inactivation of PRC1 in male germ cells results in the gradual loss of a stem cell population and severe differentiation defects, leading to male infertility. In the stem cell population, RNF2, the dominant catalytic subunit of PRC1, activates transcription of Sall4, which codes for a transcription factor essential for subsequent spermatogenic differentiation. Furthermore, RNF2 and SALL4 together occupy transcription start sites of germline genes in the stem cell population. Once differentiation commences, these germline genes are activated to enable the progression of spermatogenesis. Our study identifies a novel mechanism by which Polycomb directs the developmental process by activating a group of lineage-specific genes.


Asunto(s)
Complejo Represivo Polycomb 1/fisiología , Espermatogénesis/genética , Activación Transcripcional , Animales , Línea Celular , Proteínas de Unión al ADN/metabolismo , Epigénesis Genética , Masculino , Ratones , Ratones Transgénicos , Complejo Represivo Polycomb 1/genética , Complejo Represivo Polycomb 1/metabolismo , Espermatogonias/citología , Espermatogonias/metabolismo , Factores de Transcripción/metabolismo , Sitio de Iniciación de la Transcripción , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
4.
Zoolog Sci ; 39(6): 529-544, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36495488

RESUMEN

Many insects, including ants, are known to respond visually to conspicuous objects. In this study, we compared orientation in an arena containing only a black target beacon as local information in six species of ants of widely varying degree of phylogenic relatedness, foraging strategy, and eye morphology (Aphaenogaster, Brachyponera, Camponotus, Formica, and two Lasius spp.), often found associated in similar urban anthropogenic habitats. Four species of ants displayed orientation toward the beacon, with two orienting toward it directly, while the other two approached it via convoluted paths. The two remaining species did not show any orientation with respect to the beacon. The results did not correlate with morphological parameters of the visual systems and could not be fully interpreted in terms of the species' ecology, although convoluted paths are linked to higher significance of chemical signals. Beacon aiming was shown to be an innate behavior in visually naive Formica workers, which, however, were less strongly attracted to the beacon than older foragers. Thus, despite sharing the same habitats and supposedly having similar neural circuits, even a very simple stimulus-related behavior in the absence of other information can differ widely in ants but is likely an ancestral trait retained especially in species with smaller eyes. The comparative analysis of nervous systems opens the possibility of determining general features of circuits responsible for innate and possibly learned attraction toward particular stimuli.


Asunto(s)
Hormigas , Animales , Hormigas/fisiología , Aprendizaje , Ecosistema
5.
Cell Mol Life Sci ; 79(1): 18, 2021 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-34971404

RESUMEN

In mammalian male meiosis, the heterologous X and Y chromosomes remain unsynapsed and, as a result, are subject to meiotic sex chromosome inactivation (MSCI). MSCI is required for the successful completion of spermatogenesis. Following the initiation of MSCI, the X and Y chromosomes undergo various epigenetic modifications and are transformed into a nuclear body termed the XY body. Here, we review the mechanisms underlying the initiation of two essential, sequential processes in meiotic prophase I: MSCI and XY-body formation. The initiation of MSCI is directed by the action of DNA damage response (DDR) pathways; downstream of the DDR, unique epigenetic states are established, leading to the formation of the XY body. Accumulating evidence suggests that MSCI and subsequent XY-body formation may be driven by phase separation, a physical process that governs the formation of membraneless organelles and other biomolecular condensates. Thus, here we gather literature-based evidence to explore a phase separation hypothesis for the initiation of MSCI and the formation of the XY body.


Asunto(s)
Compensación de Dosificación (Genética) , Meiosis , Modelos Biológicos , Cromosomas Sexuales/metabolismo , Animales , Daño del ADN/genética , Reparación del ADN/genética , Humanos , Meiosis/genética
6.
Proc Natl Acad Sci U S A ; 115(19): 4957-4962, 2018 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-29686098

RESUMEN

Repressive H3K27me3 and active H3K4me2/3 together form bivalent chromatin domains, molecular hallmarks of developmental potential. In the male germline, these domains are thought to persist into sperm to establish totipotency in the next generation. However, it remains unknown how H3K27me3 is established on specific targets in the male germline. Here, we demonstrate that a germline-specific Polycomb protein, SCML2, binds to H3K4me2/3-rich hypomethylated promoters in undifferentiated spermatogonia to facilitate H3K27me3. Thus, SCML2 establishes bivalent domains in the male germline of mice. SCML2 regulates two major classes of bivalent domains: Class I domains are established on developmental regulator genes that are silent throughout spermatogenesis, while class II domains are established on somatic genes silenced during late spermatogenesis. We propose that SCML2-dependent H3K27me3 in the male germline prepares the expression of developmental regulator and somatic genes in embryonic development.


Asunto(s)
Histonas/metabolismo , Proteínas del Grupo Polycomb/metabolismo , Regiones Promotoras Genéticas , Espermatogénesis/fisiología , Espermatogonias/metabolismo , Animales , Desarrollo Embrionario , Regulación del Desarrollo de la Expresión Génica , Histonas/genética , Masculino , Ratones , Ratones Noqueados , Proteínas del Grupo Polycomb/genética , Espermatogonias/citología
7.
PLoS Genet ; 14(2): e1007233, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29462142

RESUMEN

The sex chromosomes are enriched with germline genes that are activated during the late stages of spermatogenesis. Due to meiotic sex chromosome inactivation (MSCI), these sex chromosome-linked genes must escape silencing for activation in spermatids, thereby ensuring their functions for male reproduction. RNF8, a DNA damage response protein, and SCML2, a germline-specific Polycomb protein, are two major, known regulators of this process. Here, we show that RNF8 and SCML2 cooperate to regulate ubiquitination during meiosis, an early step to establish active histone modifications for subsequent gene activation. Double mutants of Rnf8 and Scml2 revealed that RNF8-dependent monoubiquitination of histone H2A at Lysine 119 (H2AK119ub) is deubiquitinated by SCML2, demonstrating interplay between RNF8 and SCML2 in ubiquitin regulation. Additionally, we identify distinct functions of RNF8 and SCML2 in the regulation of ubiquitination: SCML2 deubiquitinates RNF8-independent H2AK119ub but does not deubiquitinate RNF8-dependent polyubiquitination. RNF8-dependent polyubiquitination is required for the establishment of H3K27 acetylation, a marker of active enhancers, while persistent H2AK119ub inhibits establishment of H3K27 acetylation. Following the deposition of H3K27 acetylation, H3K4 dimethylation is established as an active mark on poised promoters. Together, we propose a model whereby regulation of ubiquitin leads to the organization of poised enhancers and promoters during meiosis, which induce subsequent gene activation from the otherwise silent sex chromosomes in postmeiotic spermatids.


Asunto(s)
Histonas/metabolismo , Proteínas del Grupo Polycomb/fisiología , Cromosomas Sexuales/genética , Activación Transcripcional/genética , Ubiquitina-Proteína Ligasas/fisiología , Ubiquitinación/genética , Acetilación , Animales , Femenino , Masculino , Meiosis/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Cromosomas Sexuales/metabolismo , Espermátides/fisiología , Espermatogénesis/genética
8.
J Cell Sci ; 131(17)2018 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-30097555

RESUMEN

Spermatogenesis involves the progressive reorganization of heterochromatin. However, the mechanisms that underlie the dynamic remodeling of heterochromatin remain unknown. Here, we identify SCML2, a germline-specific Polycomb protein, as a critical regulator of heterochromatin organization in spermatogenesis. We show that SCML2 accumulates on pericentromeric heterochromatin (PCH) in male germ cells, where it suppresses PRC1-mediated monoubiquitylation of histone H2A at Lysine 119 (H2AK119ub) and promotes deposition of PRC2-mediated H3K27me3 during meiosis. In postmeiotic spermatids, SCML2 is required for heterochromatin organization, and the loss of SCML2 leads to the formation of ectopic patches of facultative heterochromatin. Our data suggest that, in the absence of SCML2, the ectopic expression of somatic lamins drives this process. Furthermore, the centromere protein CENP-V is a specific marker of PCH in postmeiotic spermatids, and SCML2 is required for CENP-V localization on PCH. Given the essential functions of PRC1 and PRC2 for genome-wide gene expression in spermatogenesis, our data suggest that heterochromatin organization and spermatogenesis-specific gene expression are functionally linked. We propose that SCML2 coordinates the organization of heterochromatin and gene expression through the regulation of Polycomb complexes.


Asunto(s)
Heterocromatina/metabolismo , Proteínas del Grupo Polycomb/metabolismo , Espermátides/metabolismo , Espermatogénesis , Animales , Regulación del Desarrollo de la Expresión Génica , Heterocromatina/genética , Histonas/genética , Histonas/metabolismo , Masculino , Meiosis , Ratones Endogámicos C57BL , Ratones Noqueados , Complejo Represivo Polycomb 1/genética , Complejo Represivo Polycomb 1/metabolismo , Proteínas del Grupo Polycomb/genética , Espermátides/citología
9.
Nucleic Acids Res ; 46(2): 593-608, 2018 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-29126117

RESUMEN

During spermatogenesis, germ cells undergo massive cellular reconstruction and dynamic chromatin remodeling to facilitate highly diverse transcriptomes, which are required for the production of functional sperm. However, it remains unknown how germline chromatin is organized to promote the dynamic, complex transcriptomes of spermatogenesis. Here, using ATAC-seq, we establish the varied landscape of open chromatin during spermatogenesis. We identify the reorganization of accessible chromatin in intergenic and intronic regions during the mitosis-to-meiosis transition. During the transition, mitotic-type open chromatin is closed while the de novo formation of meiotic-type open chromatin takes place. Contrastingly, differentiation processes such as spermatogonial differentiation and the meiosis-to-postmeiosis transition involve chromatin closure without the de novo formation of accessible chromatin. In spermiogenesis, the germline-specific Polycomb protein SCML2 promotes the closure of open chromatin at autosomes for gene suppression. Paradoxically, we identify the massive de novo formation of accessible chromatin when the sex chromosomes undergo meiotic sex chromosome inactivation, and this is also mediated by SCML2. These results reveal meiotic sex chromosome inactivation as an active process for chromatin organization. Together, our results unravel the genome-wide, dynamic reorganization of open chromatin and reveal mechanisms that underlie diverse transcriptomes during spermatogenesis.


Asunto(s)
Cromatina/genética , Espermatogénesis/genética , Espermatozoides/metabolismo , Transcriptoma/genética , Animales , Cromatina/metabolismo , Regulación del Desarrollo de la Expresión Génica , Histonas/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Mitosis/genética , Proteínas del Grupo Polycomb/genética , Proteínas del Grupo Polycomb/metabolismo , Cromosomas Sexuales , Espermatogonias/citología , Espermatogonias/metabolismo , Espermatozoides/citología
10.
Biol Reprod ; 100(2): 409-419, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30137219

RESUMEN

Genes involved in sexual reproduction diverge rapidly as a result of reproductive fitness. Here, we identify a novel protein domain in the germline-specific Polycomb protein SCML2 that is required for the establishment of unique gene expression programs after the mitosis-to-meiosis transition in spermatogenesis. We term this novel domain, which is comprised of rapidly evolved, DNA-binding repeat units of 28 amino acids, the SCML2 DNA-binding (SDB) repeats. These repeats are acquired in a specific subgroup of the rodent lineage, having been subjected to positive selection in the course of evolution. Mouse SCML2 has two DNA-binding domains: one is the SDB repeats and the other is an RNA-binding region, which is conserved in human SCML2. For the recruitment of SCML2 to target loci, the SDB repeats cooperate with the other functional domains of SCML2 to bind chromatin. The cooperative action of these domains enables SCML2 to sense DNA hypomethylation in an in vivo chromatin environment, thereby enabling SCML2 to bind to hypomethylated chromatin. We propose that the rapid evolution of SCML2 is due to reproductive adaptation, which has promoted species-specific gene expression programs in spermatogenesis.


Asunto(s)
Evolución Molecular , Proteínas del Grupo Polycomb/genética , Animales , Línea Celular , Cromatina , Proteínas de Unión al ADN , Regulación de la Expresión Génica , Histonas/genética , Histonas/metabolismo , Ratones , Ratones Noqueados , Filogenia , Unión Proteica , Dominios Proteicos , Proteínas Recombinantes
11.
J Hum Genet ; 59(3): 158-62, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24401909

RESUMEN

Mutations in the gene encoding bilirubin UDP-glucuronosyltransferase (UGT1A1) are known to cause Crigler-Najjar syndrome type II (CN-II). We previously encountered a patient with a nonsense mutation (Q331X) on one allele and with no other mutations in the promoter region or other exons, and proposed that CN-II is inherited as a dominant trait due to the formation of a heterologous subunit structure comprised of the altered UGT1A1 gene product (UGT1A1-p.Q331X) and the intact UGT1A1. Here, we investigated the molecular basis of CN-II in this case by expressing UGT1A1-p.Q331X in cells. UGT1A1-p.Q331X overexpressed in Escherichia coli or mammalian cells directly bound or associated with intact UGT1A1 in vitro or in vivo, respectively. Intact UGT1A1 was observed as a dimer using atomic force microscopy. Fluorescent-tagged UGT1A1-p.Q331X and intact UGT1A1 were colocalized in 293T cells, and fluorescence recovery after photobleaching analysis showed that UGT1A1-p.Q331X was retained in the endoplasmic reticulum (ER) without rapid degradation. These findings support the idea that UGT1A1-p.Q331X and UGT1A1 form a dimer and provide an increased mechanistic understanding of CN-II.


Asunto(s)
Síndrome de Crigler-Najjar/enzimología , Síndrome de Crigler-Najjar/metabolismo , Retículo Endoplásmico/enzimología , Proteínas Mutantes/metabolismo , Glucuronosiltransferasa/química , Glucuronosiltransferasa/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Fotoblanqueo , Unión Proteica , Transporte de Proteínas , Fracciones Subcelulares/metabolismo
12.
Front Mol Neurosci ; 17: 1444629, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39092202

RESUMEN

The Ca2+-dependent activator protein for secretion (CAPS/CADPS) family protein facilitates catecholamine release through the dense-core vesicle exocytosis in model neuroendocrine cell lines. However, it remains unclear if it induces dopamine release in the central neurons. This study aimed to examine the expression and function of CADPS2, one of the two CADPS paralogs, in dopamine neurons of the mouse midbrain. This study shows that CADPS2 was expressed in tyrosine hydroxylase and the vesicular monoamine transporter 2 (VMAT2)-positive dopaminergic neurons of the midbrain samples and primary mesencephalic cell cultures. Subcellular fractions rich in dopamine were collected using immunoaffinity for CADPS2 from midbrain protein extracts. Cell imaging using fluorescent false neurotransmitter FFN511 as a substrate for VMAT2 showed decreased activity-dependent dopamine release in Cadps2-deficient cultures, compared to that in wild-type cultures. These results suggest that CADPS2 is involved in dopamine release from the central neurons, indicating its involvement in the central dopamine pathway.

13.
bioRxiv ; 2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-38979277

RESUMEN

Inter-cellular transmission of mRNA is being explored in mammalian species using immortal cell lines (1-3). Here, we uncover an inter-cellular mRNA transfer phenomenon that allows for the adaptation and reprogramming of human primed pluripotent stem cells (hPSCs). This process is induced by the direct cell contact-mediated coculture with mouse embryonic stem cells (mESCs) under the condition impermissible for human primed PSC culture. Mouse-derived mRNA contents are transmitted into adapted hPSCs only in the coculture. Transfer-specific mRNA analysis show the enrichment for divergent biological pathways involving transcription/translational machinery and stress-coping mechanisms, wherein such transfer is diminished when direct cell contacts are lost. After 5 days of mESC culture, surface marker analysis, and global gene profiling confirmed that mRNA transfer-prone hPSC efficiently gains a naïve-like state. Furthermore, transfer-specific knockdown experiments targeting mouse-specific transcription factor-coding mRNAs in hPSC show that mouse-derived Tfcp2l1, Tfap2c, and Klf4 are indispensable for human naïve-like conversion. Thus, inter-species mRNA transfer triggers cellular reprogramming in mammalian cells. Our results support that episodic mRNA transfer can occur in cell cooperative and competitive processes(4), which provides a fresh perspective on understanding the roles of mRNA mobility for intra- and inter-species cellular communications.

14.
Genes Cells ; 17(9): 790-806, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22897684

RESUMEN

DNA polymerase µ (pol µ) catalyzes nonhomologous end-joining in DNA double-stranded break repair. Pol µ consists of an amino-terminal BRCA1 carboxyl-terminal homology (BRCT) domain and a pol ß-like region, which contains the catalytic site. By DNA cellulose column chromatography, using full-length pol µ and five different deletion mutants, we found that the amino-terminal region has double-stranded DNA (dsDNA)-binding activity. Pol µ without BRCT domain reduces the DNA polymerization activity when compared to full-length pol µ. Observation by atomic force microscopy showed that full-length pol µ binds to the ends and middle part of dsDNA. Pol µ lacking the amino-terminal region or with a mutation within the BRCT domain bound only to DNA ends, whereas the amino-terminal region with the BRCT domain bound to both the ends and the middle part of dsDNA (mpdDNA). Terminal deoxynucleotidyltransferase, which, like pol µ, belongs to the X family DNA polymerases, also bound to mpdDNA through its amino-terminal region.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , ADN Polimerasa Dirigida por ADN/metabolismo , ADN/metabolismo , Polimerizacion , Secuencia de Aminoácidos , Dominio Catalítico , Cromatografía/métodos , Clonación Molecular , ADN/genética , ADN Nucleotidilexotransferasa/genética , ADN Nucleotidilexotransferasa/metabolismo , Proteínas de Unión al ADN/genética , ADN Polimerasa Dirigida por ADN/genética , Activación Enzimática , Biblioteca de Genes , Genoma Humano , Humanos , Microscopía de Fuerza Atómica , Mutación , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína , Relación Estructura-Actividad
15.
Methods Mol Biol ; 2577: 65-81, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36173566

RESUMEN

Accessible chromatin often represents gene regulatory elements, including promoters and enhancers, essential for gene expression. Assay for Transposase Accessible Chromatin sequencing (ATAC-seq) is one of the most popular techniques to investigate chromatin accessibility across the genome. Here we describe, step by step, a series of optimized experimental methods and bioinformatics pipelines for ATAC-seq analysis. As an example, we present an analysis of murine spermatogenic cells: a method to isolate germ cells, a reaction step using Tn5 transposase to insert sequencing adapters into accessible DNA, a library preparation method for high-throughput sequencing, and bioinformatics analysis of sequencing data. Overall, we introduce a framework of ATAC-seq analysis that can be applied to any cell population to identify cell-type-specific gene regulatory elements and their cis-regulatory networks.


Asunto(s)
Secuenciación de Inmunoprecipitación de Cromatina , Cromatina , Animales , Cromatina/genética , ADN/genética , Análisis de Datos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Ratones , Análisis de Secuencia de ADN/métodos , Transposasas/genética , Transposasas/metabolismo
16.
Methods Mol Biol ; 2577: 123-146, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36173570

RESUMEN

Precise regulation of gene expression is integral in development. Emerging studies have highlighted that super-enhancers (SEs), which are clusters of multiple enhancers, play critical roles in regulating cell type-specific gene expression via 3D chromatin, thereby defining the cellular identities of given cells. Here we provide optimized bioinformatics pipelines to identify SEs and 3D chromatin contacts. Our pipelines encompass the processing of chromatin immunoprecipitation sequencing (ChIP-seq) data to identify SEs and the processing of genome-wide chromosome conformation capture (Hi-C) data. We can then infer long-range chromatin contacts between SEs and other genomic regions. This integrative computational approach, which can be applied to CUT&RUN and CUT&Tag, alternative technologies to ChIP-seq, allows us to identify genomic locations of SEs and their 3D genome configuration, whereby multiple SEs act in concert. We show an analysis of mouse spermatogenesis as an example of this application.


Asunto(s)
Cromatina , Elementos de Facilitación Genéticos , Animales , Cromatina/genética , Inmunoprecipitación de Cromatina , Secuenciación de Inmunoprecipitación de Cromatina , Biología Computacional , Ratones
17.
bioRxiv ; 2023 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-37720031

RESUMEN

As transposable elements (TEs) coevolved with the host genome, the host genome exploited TEs as functional regulatory elements. What remains largely unknown are how the activity of TEs, namely, endogenous retroviruses (ERVs), are regulated and how TEs evolved in the germline. Here we show that KRAB domain-containing zinc-finger proteins (KZFPs), which are highly expressed in mitotically dividing spermatogonia, bind to suppressed ERVs that function following entry into meiosis as active enhancers. These features are observed for independently evolved KZFPs and ERVs in mice and humans, i.e., are evolutionarily conserved in mammals. Further, we show that meiotic sex chromosome inactivation (MSCI) antagonizes the coevolution of KZFPs and ERVs in mammals. Our study uncovers a mechanism by which KZFPs regulate ERVs to sculpt germline transcriptomes. We propose that epigenetic programming in the mammalian germline during the mitosis-to-meiosis transition facilitates coevolution of KZFPs and TEs on autosomes and is antagonized by MSCI.

18.
bioRxiv ; 2023 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-38076840

RESUMEN

Spermatogenesis is a unidirectional differentiation process that generates haploid sperm, but how the gene expression program that directs this process is established is largely unknown. Here we determine the high-resolution 3D chromatin architecture of male germ cells during spermatogenesis and show that CTCF-mediated 3D chromatin predetermines the gene expression program required for spermatogenesis. In undifferentiated spermatogonia, CTCF-mediated chromatin contacts on autosomes pre-establish meiosis-specific super-enhancers (SE). These meiotic SE recruit the master transcription factor A-MYB in meiotic spermatocytes, which strengthens their 3D contacts and instructs a burst of meiotic gene expression. We also find that at the mitosis-to-meiosis transition, the germline-specific Polycomb protein SCML2 resolves chromatin loops that are specific to mitotic spermatogonia. Moreover, SCML2 and A-MYB establish the unique 3D chromatin organization of sex chromosomes during meiotic sex chromosome inactivation. We propose that CTCF-mediated 3D chromatin organization enforces epigenetic priming that directs unidirectional differentiation, thereby determining the cellular identity of the male germline.

19.
Cell Death Dis ; 14(8): 501, 2023 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-37542070

RESUMEN

Gonadal sex determination and differentiation are controlled by somatic support cells of testes (Sertoli cells) and ovaries (granulosa cells). In testes, the epigenetic mechanism that maintains chromatin states responsible for suppressing female sexual differentiation remains unclear. Here, we show that Polycomb repressive complex 1 (PRC1) suppresses a female gene regulatory network in postnatal Sertoli cells. We genetically disrupted PRC1 function in embryonic Sertoli cells after sex determination, and we found that PRC1-depleted postnatal Sertoli cells exhibited defective proliferation and cell death, leading to the degeneration of adult testes. In adult Sertoli cells, PRC1 suppressed specific genes required for granulosa cells, thereby inactivating the female gene regulatory network. Chromatin regions associated with female-specific genes were marked by Polycomb-mediated repressive modifications: PRC1-mediated H2AK119ub and PRC2-mediated H3K27me3. Taken together, this study identifies a critical Polycomb-based mechanism that suppresses ovarian differentiation and maintains Sertoli cell fate in adult testes.


Asunto(s)
Histonas , Complejo Represivo Polycomb 1 , Femenino , Masculino , Humanos , Complejo Represivo Polycomb 1/genética , Complejo Represivo Polycomb 1/metabolismo , Histonas/genética , Histonas/metabolismo , Testículo/metabolismo , Redes Reguladoras de Genes , Complejo Represivo Polycomb 2/genética , Complejo Represivo Polycomb 2/metabolismo , Cromatina , Proteínas del Grupo Polycomb/genética , Proteínas del Grupo Polycomb/metabolismo , Diferenciación Celular/genética
20.
Genes Cells ; 16(7): 748-64, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21668587

RESUMEN

Terminal deoxynucleotidyltransferase (TdT) interacting factor 2 (TdIF2) is an acidic protein that binds to TdT. TdIF2 binds to DNA and core histones and contains an acidic-amino acid-rich region in its C-terminus. It has therefore been suggested to function as a histone chaperone within the nucleus. TdIF2 localized within the nucleolus in HEK 293T cells, and its N-terminal (residues 1-234) and C-terminal (residues 606-756) regions were crucial for the nucleolar localization. A chromatin immunoprecipitation (ChIP) assay showed that TdIF2 associated with the promoter of human ribosomal RNA genes (hrDNAP), and an in vitro luciferase assay system showed that it promoted hrDNAP activity. Using the yeast two-hybrid system with TdIF2 as the bait, we isolated the cDNA encoding HIV Tat interactive protein 60 (Tip60), which has histone acetyltransferase (HAT) activity, as a TdIF2-binding protein. TdIF2 bound to Tip60 in vitro and in vivo, inhibited the Tip60 HAT activity in vitro and co-localized with Tip60 within the nucleolus. In addition, TdIF2 promotes upstream binding factor (UBF) acetylation in vivo. Thus, TdIF2 might promote hrDNAP activity by suppressing Tip60's HAT activity and promoting UBF acetylation.


Asunto(s)
Proteínas Portadoras/metabolismo , Nucléolo Celular/metabolismo , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas/genética , ARN Ribosómico/genética , Animales , Proteínas Portadoras/química , Citoplasma/metabolismo , ADN Nucleotidilexotransferasa/metabolismo , Regulación hacia Abajo , Células HEK293 , Histona Acetiltransferasas/metabolismo , Humanos , Lisina Acetiltransferasa 5 , Proteínas Nucleares/química , Unión Proteica , Transporte de Proteínas , Proteínas de Unión al ARN , Alineación de Secuencia , Técnicas del Sistema de Dos Híbridos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA