Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
PLoS Genet ; 18(12): e1010568, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36548300

RESUMEN

H3K9me3-based gene silencing is a conserved strategy for securing cell fate, but the mechanisms controlling lineage-specific installation of this epigenetic mark remain unclear. In Drosophila, H3K9 methylation plays an essential role in securing female germ cell fate by silencing lineage inappropriate phf7 transcription. Thus, phf7 regulation in the female germline provides a powerful system to dissect the molecular mechanism underlying H3K9me3 deposition onto protein coding genes. Here we used genetic studies to identify the essential cis-regulatory elements, finding that the sequences required for H3K9me3 deposition are conserved across Drosophila species. Transposable elements are also silenced by an H3K9me3-mediated mechanism. But our finding that phf7 regulation does not require the dedicated piRNA pathway components, piwi, aub, rhino, panx, and nxf2, indicates that the mechanisms of H3K9me3 recruitment are distinct. Lastly, we discovered that an uncharacterized member of the zinc finger associated domain (ZAD) containing C2H2 zinc finger protein family, IDENTITY CRISIS (IDC; CG4936), is necessary for H3K9me3 deposition onto phf7. Loss of idc in germ cells interferes with phf7 transcriptional regulation and H3K9me3 deposition, resulting in ectopic PHF7 protein expression. IDC's role is likely to be direct, as it localizes to a conserved domain within the phf7 gene. Collectively, our findings support a model in which IDC guides sequence-specific establishment of an H3K9me3 mini domain, thereby preventing accidental female-to-male programming.


Asunto(s)
Proteínas de Drosophila , Drosophila , Animales , Masculino , Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Células Germinativas/metabolismo , Proteínas de Homeodominio/metabolismo , Dedos de Zinc/genética , Femenino
2.
Development ; 147(17)2020 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-32816970

RESUMEN

Maintenance of germ cell sexual identity is essential for reproduction. Entry into the spermatogenesis or oogenesis pathway requires that the appropriate gene network is activated and the antagonist network is silenced. For example, in Drosophila female germ cells, forced expression of the testis-specific PHD finger protein 7 (PHF7) disrupts oogenesis, leading to either an agametic or germ cell tumor phenotype. Here, we show that PHF7-expressing ovarian germ cells inappropriately express hundreds of genes, many of which are male germline genes. We find that the majority of genes under PHF7 control in female germ cells are not under PHF7 control in male germ cells, suggesting that PHF7 is acting in a tissue-specific manner. Remarkably, transcriptional reprogramming includes a positive autoregulatory feedback mechanism in which ectopic PHF7 overcomes its own transcriptional repression through promoter switching. Furthermore, we find that tumorigenic capacity is dependent on the dosage of phf7 This study reveals that ectopic PHF7 in female germ cells leads to a loss of sexual identity and the promotion of a regulatory circuit that is beneficial for tumor initiation and progression.


Asunto(s)
Proteínas de Drosophila/metabolismo , Proteínas de Homeodominio/metabolismo , Neoplasias de Células Germinales y Embrionarias/metabolismo , Oogénesis , Transcripción Genética , Animales , Proteínas de Drosophila/genética , Drosophila melanogaster , Femenino , Proteínas de Homeodominio/genética , Neoplasias de Células Germinales y Embrionarias/genética
3.
Hum Mol Genet ; 27(11): 1913-1926, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29566152

RESUMEN

Primary ovarian insufficiency (POI) is characterized by amenorrhea and loss or dysfunction of ovarian follicles prior to the age of 40. POI has been associated with autosomal recessive mutations in genes involving hormonal signaling and folliculogenesis, however, the genetic etiology of POI most often remains unknown. Here we report MRPS22 homozygous missense variants c.404G>A (p.R135Q) and c.605G>A (p.R202H) identified in four females from two independent consanguineous families as a novel genetic cause of POI in adolescents. Both missense mutations identified in MRPS22 are rare, occurred in highly evolutionarily conserved residues, and are predicted to be deleterious to protein function. In contrast to prior reports of mutations in MRPS22 associated with severe mitochondrial disease, the POI phenotype is far less severe. Consistent with this genotype-phenotype correlation, mitochondrial defects in oxidative phosphorylation or rRNA levels were not detected in fibroblasts derived from the POI patients, suggesting a non-bioenergetic or tissue-specific mitochondrial defect. Furthermore, we demonstrate in a Drosophila model that mRpS22 deficiency specifically in somatic cells of the ovary had no effect on fertility, whereas flies with mRpS22 deficiency specifically in germ cells were infertile and agametic, demonstrating a cell autonomous requirement for mRpS22 in germ cell development. These findings collectively identify that MRPS22, a component of the small mitochondrial ribosome subunit, is critical for ovarian development and may therefore provide insight into the pathophysiology and treatment of ovarian dysfunction.


Asunto(s)
Proteínas de Drosophila/genética , Fertilidad/genética , Proteínas Mitocondriales/genética , Insuficiencia Ovárica Primaria/genética , Proteínas Ribosómicas/genética , Adolescente , Adulto , Amenorrea/genética , Amenorrea/patología , Animales , Modelos Animales de Enfermedad , Drosophila/genética , Femenino , Fertilidad/fisiología , Homocigoto , Humanos , Menopausia Prematura/genética , Mutación Missense/genética , Folículo Ovárico/patología , Insuficiencia Ovárica Primaria/patología , Adulto Joven
4.
Development ; 142(6): 1073-82, 2015 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-25758221

RESUMEN

Adult stem cells maintain tissue homeostasis by balancing self-renewal and differentiation. In Drosophila females, germline stem cells (GSCs) require Sex lethal (Sxl) to exit the stem cell state and to enter the differentiation pathway. Without Sxl GSCs do not differentiate and instead form tumors. Previous studies have shown that these tumors are not caused by a failure in the self-renewal/differentiation switch. Here, we show that Sxl is also necessary for the cell-autonomous maintenance of germ cell female identity and demonstrate that tumors are caused by the acquisition of male characteristics. Germ cells without Sxl protein exhibit a global derepression of testis genes, including Phf7, a male germline sexual identity gene. Phf7 is a key effector of the tumor-forming pathway, as it is both necessary and sufficient for tumor formation. In the absence of Sxl protein, inappropriate Phf7 expression drives tumor formation through a cell-autonomous mechanism that includes sex-inappropriate activation of Jak/Stat signaling. Remarkably, tumor formation requires a novel response to external signals emanating from the GSC niche, highlighting the importance of interactions between mutant cells and the surrounding normal cells that make up the tumor microenvironment. Derepression of testis genes, and inappropriate Phf7 expression, is also observed in germ cell tumors arising from the loss of bag of marbles (bam), demonstrating that maintenance of female sexual identity requires the concerted actions of Sxl and bam. Our work reveals that GSCs must maintain their sexual identity as they are reprogrammed into a differentiated cell, or risk tumorigenesis.


Asunto(s)
Carcinogénesis , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Regulación del Desarrollo de la Expresión Génica/fisiología , Células Germinativas/citología , Oogénesis/fisiología , Proteínas de Unión al ARN/metabolismo , Caracteres Sexuales , Células Madre Adultas/fisiología , Animales , Proteínas de Drosophila/genética , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Técnicas de Silenciamiento del Gen , Células Germinativas/fisiología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Homeostasis/fisiología , Procesamiento de Imagen Asistido por Computador , Masculino , Microscopía Fluorescente , Proteínas de Unión al ARN/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ARN , Testículo/metabolismo
5.
G3 (Bethesda) ; 11(1)2021 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-33561227

RESUMEN

The zinc finger-associated domain (ZAD) is present in over 90 C2H2 zinc finger (ZNF) proteins. Despite their abundance, only a few ZAD-ZNF genes have been characterized to date. Here, we systematically analyze the function of 68 ZAD-ZNF genes in Drosophila female germ cells by performing an in vivo RNA-interference screen. We identified eight ZAD-ZNF genes required for oogenesis, and based on further characterization of the knockdown phenotypes, we uncovered defects broadly consistent with functions in germ cell specification and/or survival, early differentiation, and egg chamber maturation. These results provide a candidate pool for future studies aimed at functionalization of this large but poorly characterized gene family.


Asunto(s)
Dedos de Zinc CYS2-HIS2 , Proteínas de Drosophila , Animales , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Femenino , Células Germinativas/metabolismo , ARN , Dedos de Zinc
6.
Nat Commun ; 9(1): 4155, 2018 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-30297796

RESUMEN

The preservation of germ cell sexual identity is essential for gametogenesis. Here we show that H3K9me3-mediated gene silencing is integral to female fate maintenance in Drosophila germ cells. Germ cell specific loss of the H3K9me3 pathway members, the H3K9 methyltransferase SETDB1, WDE, and HP1a, leads to ectopic expression of genes, many of which are normally expressed in testis. SETDB1 controls the accumulation of H3K9me3 over a subset of these genes without spreading into neighboring loci. At phf7, a regulator of male germ cell sexual fate, the H3K9me3 peak falls over the silenced testis-specific transcription start site. Furthermore, H3K9me3 recruitment to phf7 and repression of testis-specific transcription is dependent on the female sex determination gene Sxl. Thus, female identity is secured by an H3K9me3 epigenetic pathway in which Sxl is the upstream female-specific regulator, SETDB1 is the required chromatin writer, and phf7 is one of the critical SETDB1 target genes.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Células Germinativas/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Animales , Animales Modificados Genéticamente , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Femenino , Gametogénesis/genética , Regulación del Desarrollo de la Expresión Génica , Silenciador del Gen , N-Metiltransferasa de Histona-Lisina/genética , Histonas/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Lisina/metabolismo , Masculino , Metilación , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA