Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
J Biol Chem ; 299(9): 105148, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37567474

RESUMEN

Mutations in sterile alpha motif domain and histidine-aspartate domain-containing protein 1 (SAMHD1) are found in a neurodevelopmental disorder, Aicardi-Goutières syndrome, and cancers, and SAMHD1, which is a deoxynucleoside triphosphate (dNTP) triphosphorylase, was identified as a myeloid-specific HIV-1 restriction factor. Here, we characterized the enzymology and structure of an SAMHD1 ortholog of Caenorhabditis elegans, ZK177.8, which also reportedly induces developmental defects upon gene knockdown. We found ZK177.8 protein is a dNTPase allosterically regulated by dGTP. The active site of ZK177.8 recognizes both 2' OH and triphosphate moieties of dNTPs but not base moiety. The dGTP activator induces the formation of the enzymatically active ZK177.8 tetramers, and ZK177.8 protein lowers cellular dNTP levels in a human monocytic cell line. Finally, ZK177.8 tetramers display very similar X-ray crystal structure with human and mouse SAMHD1s except that its lack of the canonical sterile alpha motif domain. This striking conservation in structure, function, and allosteric regulatory mechanism for the hydrolysis of the DNA building blocks supports their host developmental roles.

2.
Cell ; 137(2): 308-20, 2009 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-19379696

RESUMEN

Epigenetic information undergoes extensive reprogramming in the germline between generations. This reprogramming may be essential to establish a developmental ground state in the zygote. We show that mutants in spr-5, the Caenorhabditis elegans ortholog of the H3K4me2 demethylase LSD1/KDM1, exhibit progressive sterility over many generations. This sterility correlates with the misregulation of spermatogenesis-expressed genes and transgenerational accumulation of the histone modification dimethylation of histone H3 on lysine 4 (H3K4me2). This suggests that H3K4me2 can serve as a stable epigenetic memory, and that erasure of H3K4me2 by LSD/KDM1 in the germline prevents the inappropriate transmission of this epigenetic memory from one generation to the next. Thus, our results provide direct mechanistic insights into the processes that are required for epigenetic reprogramming between generations.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/embriología , Caenorhabditis elegans/genética , Epigénesis Genética , Células Germinativas/citología , Células Germinativas/metabolismo , Animales , Caenorhabditis elegans/citología , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Infertilidad , Análisis por Micromatrices , Mutación , Oogénesis , Oxidorreductasas N-Desmetilantes , Espermatogénesis
3.
PLoS Genet ; 13(5): e1006821, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28562665

RESUMEN

[This corrects the article DOI: 10.1371/journal.pgen.1006227.].

4.
Nat Rev Genet ; 14(3): 228-35, 2013 03.
Artículo en Inglés | MEDLINE | ID: mdl-23416892

RESUMEN

Much attention has been given to the idea of transgenerational epigenetic inheritance, but fundamental questions remain regarding how much takes place and the impact that this might have on organisms. We asked five leading researchers in this area--working on a range of model organisms and in human disease--for their views on these topics. Their responses highlight the mixture of excitement and caution that surrounds transgenerational epigenetic inheritance and the wide gulf between species in terms of our knowledge of the mechanisms that may be involved.


Asunto(s)
Epigénesis Genética , Epigenómica , Patrón de Herencia/genética , Animales , Modelos Animales de Enfermedad , Herencia , Humanos
5.
PLoS Genet ; 12(8): e1006227, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27541139

RESUMEN

The elongation phase of transcription by RNA Polymerase II (Pol II) involves numerous events that are tightly coordinated, including RNA processing, histone modification, and chromatin remodeling. RNA splicing factors are associated with elongating Pol II, and the interdependent coupling of splicing and elongation has been documented in several systems. Here we identify a conserved, multi-domain cyclophilin family member, SIG-7, as an essential factor for both normal transcription elongation and co-transcriptional splicing. In embryos depleted for SIG-7, RNA levels for over a thousand zygotically expressed genes are substantially reduced, Pol II becomes significantly reduced at the 3' end of genes, marks of transcription elongation are reduced, and unspliced mRNAs accumulate. Our findings suggest that SIG-7 plays a central role in both Pol II elongation and co-transcriptional splicing and may provide an important link for their coordination and regulation.


Asunto(s)
Proteínas de Caenorhabditis elegans/genética , Ciclofilinas/genética , Desarrollo Embrionario/genética , ARN Polimerasa II/genética , Transcripción Genética , Animales , Caenorhabditis elegans/genética , Caenorhabditis elegans/crecimiento & desarrollo , Ciclofilinas/biosíntesis , Embrión no Mamífero , Regulación del Desarrollo de la Expresión Génica , Intrones/genética , Fosforilación , Interferencia de ARN , Empalme del ARN/genética
6.
Development ; 140(17): 3703-13, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23903194

RESUMEN

RNA polymerase II (Pol II) elongation in metazoans is thought to require phosphorylation of serine 2 (Ser2-P) of the Pol II C-terminal domain (CTD) by the P-TEFb complex, CDK-9/cyclin T. Another Ser2 kinase complex, CDK-12/cyclin K, which requires upstream CDK-9 activity has been identified in Drosophila and human cells. We show that regulation of Ser2-P in C. elegans soma is similar to other metazoan systems, but Ser2-P in the germline is independent of CDK-9, and largely requires only CDK-12. The observed differences are not due to differential tissue expression as both kinases and their cyclin partners are ubiquitously expressed. Surprisingly, loss of CDK-9 from germ cells has little effect on Ser2-P, yet CDK-9 is essential for germline development. By contrast, loss of CDK-12 and Ser2-P specifically from germ cells has little impact on germline development or function, although significant loss of co-transcriptional H3K36 trimethylation is observed. These results show a reduced requirement for Pol II Ser2-P in germline development and suggest that generating Ser2-P is not the essential role of CDK-9 in these cells. Transcriptional elongation in the C. elegans germline thus appears to be uniquely regulated, which may be a novel facet of germline identity.


Asunto(s)
Caenorhabditis elegans/enzimología , Células Germinativas/metabolismo , Factor B de Elongación Transcripcional Positiva/metabolismo , ARN Polimerasa II/metabolismo , Animales , Animales Modificados Genéticamente , Western Blotting , Tamaño de la Nidada , Quinasas Ciclina-Dependientes/metabolismo , Microscopía Fluorescente , Fosforilación , Interferencia de ARN , Reacción en Cadena en Tiempo Real de la Polimerasa , Serina/metabolismo
7.
Nucleic Acids Res ; 42(9): 5567-81, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24682813

RESUMEN

The hermaphrodite germline of Caenorhabditis elegans initially engages in spermatogenesis and then switches to oogenesis during late stages of larval development. TRA-1, a member of the Ci/Gli family of transcriptional repressors, plays an essential role in this switch by repressing genes that promote spermatogenesis. WDR5 proteins are conserved components of histone methyltransferase complexes normally associated with gene activation. However, two C. elegans WDR5 homologs, wdr-5.1 and wdr-5.2 are redundantly required for normal TRA-1 dependent repression, and this function is independent of their roles in histone methylation. Animals lacking wdr-5.1/wdr-5.2 function fail to switch to oogenesis at 25°C, resulting in a masculinization of germline (Mog) phenotype. The Mog phenotype is caused by ectopic expression of fog-3, a direct target of TRA-1 repression. WDR-5.1 associates with the fog-3 promoter and is required for TRA-1 to bind to fog-3 promoter. Other direct targets of TRA-1 are similarly derepressed in the double mutant. These results show that WDR5 plays a novel and important role in stabilizing transcriptional repression during C. elegans sex determination, and provide evidence that this important protein may operate independently of its established role in histone methyltransferase complexes.


Asunto(s)
Proteínas de Caenorhabditis elegans/fisiología , Caenorhabditis elegans/genética , Proteínas de Unión al ADN/fisiología , Factores de Transcripción/fisiología , Transcripción Genética , Animales , Caenorhabditis elegans/citología , Caenorhabditis elegans/crecimiento & desarrollo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Epigénesis Genética , Regulación del Desarrollo de la Expresión Génica , Técnicas de Inactivación de Genes , Histonas/metabolismo , Masculino , Metilación , Oocitos/fisiología , Oogénesis , Procesamiento Proteico-Postraduccional , Procesos de Determinación del Sexo , Espermatogénesis , Espermatozoides/fisiología
8.
PLoS Genet ; 7(3): e1001349, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21455483

RESUMEN

The methylation of lysine 4 of Histone H3 (H3K4me) is an important component of epigenetic regulation. H3K4 methylation is a consequence of transcriptional activity, but also has been shown to contribute to "epigenetic memory"; i.e., it can provide a heritable landmark of previous transcriptional activity that may help promote or maintain such activity in subsequent cell descendants or lineages. A number of multi-protein complexes that control the addition of H3K4me have been described in several organisms. These Set1/MLL or COMPASS complexes often share a common subset of conserved proteins, with other components potentially contributing to tissue-specific or developmental regulation of the methyltransferase activity. Here we show that the normal maintenance of H3K4 di- and tri-methylation in the germ line of Caenorhabditis elegans is dependent on homologs of the Set1/MLL complex components WDR-5.1 and RBBP-5. Different methylation states that are each dependent on wdr-5.1 and rbbp-5 require different methyltransferases. In addition, different subsets of conserved Set1/MLL-like complex components appear to be required for H3K4 methylation in germ cells and somatic lineages at different developmental stages. In adult germ cells, mutations in wdr-5.1 or rbbp-5 dramatically affect both germ line stem cell (GSC) population size and proper germ cell development. RNAi knockdown of RNA Polymerase II does not significantly affect the wdr-5.1-dependent maintenance of H3K4 methylation in either early embryos or adult GSCs, suggesting that the mechanism is not obligately coupled to transcription in these cells. A separate, wdr-5.1-independent mode of H3K4 methylation correlates more directly with transcription in the adult germ line and in embryos. Our results indicate that H3K4 methylation in the germline is regulated by a combination of Set1/MLL component-dependent and -independent modes of epigenetic establishment and maintenance.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans , Epigenómica , Células Germinativas/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Metiltransferasas/metabolismo , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Animales , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Regulación del Desarrollo de la Expresión Génica , Genoma de los Helmintos/genética , Histonas/metabolismo , Metilación , Metiltransferasas/genética , Proteínas Nucleares/metabolismo , Interferencia de ARN , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo , Transcripción Genética
9.
PLoS Genet ; 7(6): e1001391, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21695223

RESUMEN

Epigenetic information, such as parental imprints, can be transmitted with genetic information from parent to offspring through the germ line. Recent reports show that histone modifications can be transmitted through sperm as a component of this information transfer. How the information that is transferred is established in the parent and maintained in the offspring is poorly understood. We previously described a form of imprinted X inactivation in Caenorhabditis elegans where dimethylation on histone 3 at lysine 4 (H3K4me2), a mark of active chromatin, is excluded from the paternal X chromosome (Xp) during spermatogenesis and persists through early cell divisions in the embryo. Based on the observation that the Xp (unlike the maternal X or any autosome) is largely transcriptionally inactive in the paternal germ line, we hypothesized that transcriptional activity in the parent germ line may influence epigenetic information inherited by and maintained in the embryo. We report that chromatin modifications and histone variant patterns assembled in the germ line can be retained in mature gametes. Furthermore, despite extensive chromatin remodeling events at fertilization, the modification patterns arriving with the gametes are largely retained in the early embryo. Using transgenes, we observe that expression in the parental germline correlates with differential chromatin assembly that is replicated and maintained in the early embryo. Expression in the adult germ cells also correlates with more robust expression in the somatic lineages of the offspring. These results suggest that differential expression in the parental germ lines may provide a potential mechanism for the establishment of parent-of-origin epigenomic content. This content can be maintained and may heritably affect gene expression in the offspring.


Asunto(s)
Caenorhabditis elegans/embriología , Caenorhabditis elegans/genética , Embrión no Mamífero/metabolismo , Epigénesis Genética , Células Germinativas/citología , Animales , Caenorhabditis elegans/metabolismo , Cromatina/metabolismo , Metilación de ADN , Regulación del Desarrollo de la Expresión Génica , Células Germinativas/metabolismo , Histonas/genética , Histonas/metabolismo , Masculino
10.
Nat Genet ; 36(1): 100-5, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14702046

RESUMEN

The genetic imprinting of individual loci or whole chromosomes, as in imprinted X-chromosome inactivation in mammals, is established and reset during gametogenesis; defects in this process in the parent can result in disease in the offspring. We describe a sperm-specific chromatin-based imprinting of the X chromosome in the nematode Caenorhabditis elegans that is restricted to histone H3 modifications. The epigenetic imprint is established during spermatogenesis and its stability in the offspring is affected by the presence of a pairing partner during meiosis in the parental germ line. We observed that DNA lacking a pairing partner during meiosis, the normal situation for the X chromosome in males, is targeted for methylation of histone H3 at Lys9 (H3-Lys9) and can be silenced. Targeting unpaired DNA for silencing during meiosis, a potential hallmark of genome defense, could therefore have a conserved role in imprinted X-chromosome inactivation and, ultimately, in sex chromosome evolution.


Asunto(s)
Caenorhabditis elegans/genética , Impresión Genómica , Meiosis , Cromosoma X , Animales , Cromatina , Gametogénesis , Histonas , Masculino , Metilación
11.
PLoS Genet ; 5(8): e1000624, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19714217

RESUMEN

Many organisms have a mechanism for down regulating the expression of non-synapsed chromosomes and chromosomal regions during meiosis. This phenomenon is thought to function in genome defense. During early meiosis in Caenorhabditis elegans, unpaired chromosomes (e.g., the male X chromosome) become enriched for a modification associated with heterochromatin and transcriptional repression, dimethylation of histone H3 on lysine 9 (H3K9me2). This enrichment requires activity of the cellular RNA-directed RNA polymerase, EGO-1. Here we use genetic mutation, RNA interference, immunofluorescence microscopy, fluorescence in situ hybridization, and molecular cloning methods to identify and analyze three additional regulators of meiotic H3K9me2 distribution: CSR-1 (a Piwi/PAZ/Argonaute protein), EKL-1 (a Tudor domain protein), and DRH-3 (a DEAH/D-box helicase). In csr-1, ekl-1, and drh-3 mutant males, we observed a reduction in H3K9me2 accumulation on the unpaired X chromosome and an increase in H3K9me2 accumulation on paired autosomes relative to controls. We observed a similar shift in H3K9me2 pattern in hermaphrodites that carry unpaired chromosomes. Based on several assays, we conclude that ectopic H3K9me2 accumulates on paired and synapsed chromosomes in these mutants. We propose alternative models for how a small RNA-mediated pathway may regulate H3K9me2 accumulation during meiosis. We also describe the germline phenotypes of csr-1, ekl-1, and drh-3 mutants. Our genetic data suggest that these factors, together with EGO-1, participate in a regulatory network to promote diverse aspects of development.


Asunto(s)
Caenorhabditis elegans/metabolismo , Cromosomas/genética , Heterocromatina/metabolismo , Meiosis , ARN Interferente Pequeño/genética , Animales , Caenorhabditis elegans/citología , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Células Germinativas/citología , Células Germinativas/metabolismo , Heterocromatina/genética , Histonas/metabolismo , Masculino , Metilación , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , ARN Polimerasa Dependiente del ARN/genética , ARN Polimerasa Dependiente del ARN/metabolismo
12.
Genetics ; 220(1)2022 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-34788833

RESUMEN

The dynein motor complex is thought to aid in homolog pairing in many organisms by moving chromosomes within the nuclear periphery to promote and test homologous interactions. This precedes synaptonemal complex (SC) formation during homolog synapsis, which stabilizes homolog proximity during recombination. We observed that depletion of the dynein light chain (DLC-1) in Caenorhabditis elegans irreversibly prevents synapsis, causing an increase in off-chromatin formation of SC protein foci with increasing temperature. This requirement for DLC-1 is independent of its function in dynein motors, as SYP protein foci do not form with depletion of other dynein motor components. In contrast to normal SC-related structures, foci formed with DLC-1 depletion are resistant to dissolution with 1,6-hexanediol, similar to aggregates of SC proteins formed in high growth temperatures. Dynein light chains have been shown to act as hub proteins that interact with other proteins through a conserved binding motif. We identified a similar DLC-1 binding motif in the C. elegans SC protein SYP-2, and mutation of the putative motif causes meiosis defects that are exacerbated by elevated temperatures. We propose that DLC-1 acts as a pre-synapsis chaperone-like factor for SYP proteins to help regulate their self-association prior to the signals for SC assembly, a role that is revealed by its increased essentiality at elevated temperatures.


Asunto(s)
Caenorhabditis elegans , Animales
13.
PLoS Genet ; 4(9): e1000187, 2008 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-18787694

RESUMEN

In all eukaryotes, histone variants are incorporated into a subset of nucleosomes to create functionally specialized regions of chromatin. One such variant, H2A.Z, replaces histone H2A and is required for development and viability in all animals tested to date. However, the function of H2A.Z in development remains unclear. Here, we use ChIP-chip, genetic mutation, RNAi, and immunofluorescence microscopy to interrogate the function of H2A.Z (HTZ-1) during embryogenesis in Caenorhabditis elegans, a key model of metazoan development. We find that HTZ-1 is expressed in every cell of the developing embryo and is essential for normal development. The sites of HTZ-1 incorporation during embryogenesis reveal a genome wrought by developmental processes. HTZ-1 is incorporated upstream of 23% of C. elegans genes. While these genes tend to be required for development and occupied by RNA polymerase II, HTZ-1 incorporation does not specify a stereotypic transcription program. The data also provide evidence for unexpectedly widespread independent regulation of genes within operons during development; in 37% of operons, HTZ-1 is incorporated upstream of internally encoded genes. Fewer sites of HTZ-1 incorporation occur on the X chromosome relative to autosomes, which our data suggest is due to a paucity of developmentally important genes on X, rather than a direct function for HTZ-1 in dosage compensation. Our experiments indicate that HTZ-1 functions in establishing or maintaining an essential chromatin state at promoters regulated dynamically during C. elegans embryogenesis.


Asunto(s)
Proteínas de Caenorhabditis elegans/genética , Caenorhabditis elegans/embriología , Caenorhabditis elegans/genética , Genoma de los Helmintos , Histonas/genética , Animales , Caenorhabditis elegans/crecimiento & desarrollo , Proteínas de Caenorhabditis elegans/metabolismo , Compensación de Dosificación (Genética) , Desarrollo Embrionario/genética , Femenino , Técnica del Anticuerpo Fluorescente , Histonas/metabolismo , Modelos Genéticos , Operón/genética , Regiones Promotoras Genéticas , Interferencia de ARN , ARN Polimerasa II/metabolismo , Sitio de Iniciación de la Transcripción , Cromosoma X/metabolismo
14.
Dev Growth Differ ; 52(6): 527-32, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20646025

RESUMEN

Epigenetic mechanisms are thought to help regulate the unique transcription program that is established in germ cell development. During the germline cycle of many organisms, the epigenome undergoes waves of extensive resetting events, while a part of epigenetic modification remains faithful to specific loci. Little is known about the mechanisms underlying these events, how loci are selected for, or avoid, reprogramming, or even why these events are required. In particular, although the significance of genomic imprinting phenomena involving DNA methylation in mammals is now well accepted, the role of histone modification as a transgenerational epigenetic mechanism has been the subject of debate. Such epigenetic mechanisms may help regulate transcription programs and/or the pluripotent status conferred on germ cells, and contribute to germ line continuity across generations. Recent studies provide new evidence for heritability of histone modifications through germ line cells and its potential effects on transcription regulation both in the soma and germ line of subsequent generations. Unraveling transgenerational epigenetic mechanisms involving highly conserved histone modifications in elegant model systems will accelerate the generation of new paradigms and inspire research in a wide variety of fields, including basic developmental studies and clinical stem cell research.


Asunto(s)
Desarrollo Embrionario/fisiología , Epigénesis Genética/genética , Regulación del Desarrollo de la Expresión Génica/genética , Células Germinativas/citología , Histonas/metabolismo , Patrón de Herencia/genética , Modelos Biológicos , Animales , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/metabolismo , Oxidorreductasas N-Desmetilantes/metabolismo
15.
Dev Cell ; 5(5): 747-57, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14602075

RESUMEN

In C. elegans, mRNA production is initially repressed in the embryonic germline by a protein unique to C. elegans germ cells, PIE-1. PIE-1 is degraded upon the birth of the germ cell precursors, Z2 and Z3. We have identified a chromatin-based mechanism that succeeds PIE-1 repression in these cells. A subset of nucleosomal histone modifications, methylated lysine 4 on histone H3 (H3meK4) and acetylated lysine 8 on histone H4 (H4acetylK8), are globally lost and the DNA appears more condensed. This coincides with PIE-1 degradation and requires that germline identity is not disrupted. Drosophila pole cell chromatin also lacks H3meK4, indicating that a unique chromatin architecture is a conserved feature of embryonic germ cells. Regulation of the germline-specific chromatin architecture requires functional nanos activity in both organisms. These results indicate that genome-wide repression via a nanos-regulated, germ cell-specific chromatin organization is a conserved feature of germline maintenance during embryogenesis.


Asunto(s)
Caenorhabditis elegans/genética , Linaje de la Célula , Cromatina/metabolismo , Drosophila melanogaster/genética , Células Germinativas/fisiología , Conformación de Ácido Nucleico , Animales , Blastómeros/citología , Blastómeros/fisiología , Caenorhabditis elegans/citología , Caenorhabditis elegans/embriología , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Drosophila melanogaster/embriología , Histonas/metabolismo , Metilación , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Factores de Transcripción/metabolismo
16.
Curr Biol ; 15(21): 1972-8, 2005 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-16271877

RESUMEN

During meiosis in C. elegans, unpaired chromosomes and chromosomal regions accumulate high levels of histone H3 lysine 9 dimethylation (H3K9me2), a modification associated with facultative heterochromatin assembly and the resulting transcriptional silencing. Meiotic silencing of unpaired DNA may be a widely conserved genome defense mechanism. The mechanisms of meiotic silencing remain unclear, although both transcriptional and posttranscriptional processes are implicated. Cellular RNA-dependent RNA polymerases (RdRPs) function in development and RNA-mediated silencing in many species and in heterochromatin assembly in S. pombe. There are four C. elegans RdRPs, including two with known germline functions. EGO-1 is required for fertility and robust germline RNAi. RRF-3 acts genetically to repress RNAi and is required for normal meiosis and spermatogenesis at elevated temperatures (S. L'Hernault, personal communication). Among C. elegans RdRPs, we find that only EGO-1 is required for H3K9me2 enrichment on unpaired chromosomal regions during meiosis. This H3K9me2 enrichment does not require Dicer or Drosha nuclease or any of several other proteins required for RNAi. ego-1 interacts genetically with him-17, another regulator of chromatin and meiosis, to promote germline development. We conclude that EGO-1 is an essential component of meiotic silencing in C. elegans.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/enzimología , ADN/metabolismo , Silenciador del Gen/fisiología , Heterocromatina/fisiología , Meiosis/fisiología , ARN Polimerasa Dependiente del ARN/metabolismo , Animales , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/fisiología , Proteínas de Ciclo Celular/metabolismo , Técnica del Anticuerpo Fluorescente Indirecta , Heterocromatina/enzimología , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina/metabolismo , Meiosis/genética , Metilación , Modelos Moleculares , Mutación/genética , Proteína Metiltransferasas , ARN Polimerasa Dependiente del ARN/genética , ARN Polimerasa Dependiente del ARN/fisiología
17.
Genetics ; 174(4): 1895-906, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17028322

RESUMEN

In C. elegans, germline blastomeres are initially kept transcriptionally quiescent by the maternally loaded CCCH zinc-finger protein PIE-1. PIE-1 disappears upon the birth of the primordial germ cells Z2 and Z3, yet these cells appear to remain quiescent. We have previously demonstrated that there is a chromatin-based repression that succeeds PIE-1 degradation. The chromatin in Z2/Z3 loses certain histone modifications, including histone H3 lysine 4 dimethylation (H3K4me2), a conserved marker for transcriptionally competent chromatin. We find that mutations in the maternal-effect gene emb-4 cause defects in both PIE-1 degradation and germline-specific chromatin remodeling. emb-4 encodes a highly conserved protein with orthologs in fly, mouse, and human and has a subtle role in Notch signaling. The embryonic phenotype of emb-4 is consistent with a defect in the efficient and timely activation of developmental programs, including germline chromatin remodeling. We also find that, as in early somatic blastomeres, the degradation of PIE-1 in Z2/Z3 is facilitated by zinc-finger-interacting protein ZIF-1, and in the absence of either zif-1 or emb-4, PIE-1 is abnormally retained in Z2/Z3.


Asunto(s)
Caenorhabditis elegans/genética , Linaje de la Célula , Ensamble y Desensamble de Cromatina , Embrión no Mamífero/metabolismo , Células Germinativas/fisiología , Animales , Caenorhabditis elegans/citología , Caenorhabditis elegans/embriología , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Embrión no Mamífero/citología , Desarrollo Embrionario , Regulación del Desarrollo de la Expresión Génica , Histonas/metabolismo , Mutación/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fenotipo
18.
PLoS One ; 12(5): e0177473, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28498828

RESUMEN

The proper regulation of spermatogenesis is crucial to ensure the continued production of sperm and fertility. Here, we investigated the function of the H3K4me2 demethylase KDM1A/LSD1 during spermatogenesis in developing and adult mice. Conditional deletion of Kdm1a in the testis just prior to birth leads to fewer spermatogonia and germ cell loss before 3 weeks of age. These results demonstrate that KDM1A is required for spermatogonial differentiation, as well as germ cell survival, in the developing testis. In addition, inducible deletion of Kdm1a in the adult testis results in the abnormal accumulation of meiotic spermatocytes, as well as apoptosis and progressive germ cell loss. These results demonstrate that KDM1A is also required during adult spermatogenesis. Furthermore, without KDM1A, the stem cell factor OCT4 is ectopically maintained in differentiating germ cells. This requirement for KDM1A is similar to what has been observed in other stem cell populations, suggesting a common function. Taken together, we propose that KDM1A is a key regulator of spermatogenesis and germ cell maintenance in the mouse.


Asunto(s)
Diferenciación Celular/genética , Histona Demetilasas/metabolismo , Espermatogénesis/genética , Espermatogonias/citología , Espermatogonias/metabolismo , Animales , Apoptosis/genética , Apoptosis/fisiología , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Histona Demetilasas/genética , Masculino , Ratones , Espermatozoides/citología , Espermatozoides/metabolismo , Células Madre/citología , Células Madre/metabolismo , Testículo/citología , Testículo/metabolismo
19.
Mech Dev ; 122(5): 707-20, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15817227

RESUMEN

RNA helicase A (RHA) is a multifunctional protein with established roles in chromatin regulation. The protein is conserved in worms, Drosophila, and mammals, but its role in worms has not been previously studied. We found that a deletion mutant lacking rha-1 has a temperature-sensitive defect in germline transcriptional silencing, consistent with RHA-1 having a function in transcription regulation. Transcriptional desilencing in these rha-1(tm329) mutants was associated with a loss of lysine 9 methylation on histone H3 that is normally associated with silenced chromatin. Other histone modifications are also mis-localized in the germ cells in the mutants. These defects in histone modifications suggest that there is a general transcription regulation defect in the mutant worms that results in a temperature-sensitive sterile phenotype. At the restrictive temperature, the extent of germ cell mitoses is reduced, and the mutants are sterile due to defects in meiosis and gametogenesis. Our results suggest that RHA-1 is a conserved transcription regulation protein that controls germline proliferation and development in C. elegans.


Asunto(s)
Autoantígenos/fisiología , Caenorhabditis elegans/fisiología , Regulación del Desarrollo de la Expresión Génica , ARN Helicasas/fisiología , Transcripción Genética , Animales , Muerte Celular , Proliferación Celular , Cromatina/metabolismo , ARN Helicasas DEAD-box , Eliminación de Gen , Silenciador del Gen , Histonas/metabolismo , Inmunohistoquímica , Hibridación in Situ , Lisina/química , Meiosis , Metilación , Mitosis , Modelos Genéticos , Proteínas de Neoplasias , Fenotipo , ARN/química , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Temperatura
20.
FASEB J ; 18(15): 1834-50, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15576487

RESUMEN

ADP-ribosylation factor (Arf) and Arf-like (Arl) proteins are a family of highly conserved 21 kDa GTPases that emerged early in the evolution of eukaryotes. These proteins serve regulatory roles in vesicular traffic, lipid metabolism, microtubule dynamics, development, and likely other cellular processes. We found evidence for the presence of 6 Arf family members in the protist Giardia lamblia and 22 members in mammals. A phylogenetic analysis was performed to delineate the evolutionary relationships among Arf family members and to attempt to organize them by both their evolutionary origins and functions in cells and/or organisms. The approximately 100 protein sequences analyzed from animals, fungi, plants, and protists clustered into 11 groups, including Arfs, nine Arls, and Sar proteins. To begin functional analyses of the family in a metazoan model organism, we examined roles for all three C. elegans Arfs (Arf-1, Arf-3, and Arf-6) and three Arls (Arl-1, Arl-2, and Arl-3) by use of RNA-mediated interference (RNAi). Injection of double-stranded RNA (dsRNA) encoding Arf-1 or Arf-3 into N2 hermaphrodites produced embryonic lethality in their offspring and, later, sterility in the injected animals themselves. Injection of Arl-2 dsRNA resulted in a disorganized germline and sterility in early offspring, with later offspring exhibiting an early embryonic arrest. Thus, of the six Arf family members examined in C. elegans, at least three are required for embryogenesis. These data represent the first analysis of the role(s) of multiple members of this family in the development of a multicellular organism.


Asunto(s)
Factores de Ribosilacion-ADP/clasificación , Factores de Ribosilacion-ADP/fisiología , Caenorhabditis elegans/embriología , Caenorhabditis elegans/enzimología , Filogenia , Factores de Ribosilacion-ADP/genética , Animales , Caenorhabditis elegans/genética , Células Eucariotas/enzimología , Genómica , Proteínas Fluorescentes Verdes/metabolismo , Proteínas de la Membrana/clasificación , Interferencia de ARN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA