Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Genes Dev ; 37(21-24): 984-997, 2023 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-37993255

RESUMO

The RING-type E3 ligase has been known for over two decades, yet its diverse modes of action are still the subject of active research. Plant homeodomain (PHD) finger protein 7 (PHF7) is a RING-type E3 ubiquitin ligase responsible for histone ubiquitination. PHF7 comprises three zinc finger domains: an extended PHD (ePHD), a RING domain, and a PHD. While the function of the RING domain is largely understood, the roles of the other two domains in E3 ligase activity remain elusive. Here, we present the crystal structure of PHF7 in complex with the E2 ubiquitin-conjugating enzyme (E2). Our structure shows that E2 is effectively captured between the RING domain and the C-terminal PHD, facilitating E2 recruitment through direct contact. In addition, through in vitro binding and functional assays, we demonstrate that the N-terminal ePHD recognizes the nucleosome via DNA binding, whereas the C-terminal PHD is involved in histone H3 recognition. Our results provide a molecular basis for the E3 ligase activity of PHF7 and uncover the specific yet collaborative contributions of each domain to the PHF7 ubiquitination activity.


Assuntos
Histonas , Ubiquitina-Proteína Ligases , Histonas/metabolismo , Ubiquitinação , Ubiquitina-Proteína Ligases/metabolismo , Proteínas de Ligação a DNA/metabolismo , Dedos de Zinco , Enzimas de Conjugação de Ubiquitina/metabolismo
2.
Biochem Biophys Res Commun ; 704: 149596, 2024 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-38430697

RESUMO

PHD finger protein 7 (Phf7) is a member of the PHF family proteins, which plays important roles in spermiogenesis. Phf7 is expressed in the adult testes and its deficiency causes male infertility. In this study, we tried to find the causal relationship between Phf7 deficiency and reduced growth retardation which were found in null knock-out (Phf7-/-) mice. Phf7-/- mice were born normally in the Mendelian ratio. However, the Phf7-/- males showed decreased body weight gain, bone mineral density, and bone mineral content compared to those in wild-type (WT) mice. Histological analysis for tibia revealed increased number of osteoclast cells in Phf7-/- mice compared with that in WT mice. When we analyzed the expressions for marker genes for the initial stage of osteoclastogenesis, such as receptor activator of nuclear factor kappa B (Rank) in tibia, there was no difference in the mRNA levels between Phf7-/- and WT mice. However, the expression of tartrate-resistant acid phosphatase (Trap), a mature stage marker gene, was significantly higher in Phf7-/- mice than in WT mice. In addition, the levels of testosterone and dihydrotestosterone (DHT), more potent and active form of testosterone, were significantly reduced in the testes of Phf7-/- mice compared to those in WT mice. Furthermore, testicular mRNA levels for steroidogenesis marker genes, namely Star, Cyp11a1, Cyp17a1 and 17ß-hsd, were significantly lower in Phf7-/- mice than in WT mice. In conclusion, these results suggest that Phf7 deficiency reduces the production of male sex hormones and thereby impairs associated bone remodeling.


Assuntos
Hormônios Testiculares , Animais , Masculino , Camundongos , Remodelação Óssea , Osteoclastos/metabolismo , RNA Mensageiro/metabolismo , Hormônios Testiculares/metabolismo , Testosterona/metabolismo
3.
Development ; 146(13)2019 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-31189663

RESUMO

Epigenetic regulation, including histone-to-protamine exchanges, controls spermiogenesis. However, the underlying mechanisms of this regulation are largely unknown. Here, we report that PHF7, a testis-specific PHD and RING finger domain-containing protein, is essential for histone-to-protamine exchange in mice. PHF7 is specifically expressed during spermiogenesis. PHF7 deletion results in male infertility due to aberrant histone retention and impaired protamine replacement in elongated spermatids. Mechanistically, PHF7 can simultaneously bind histone H2A and H3; its PHD domain, a histone code reader, can specifically bind H3K4me3/me2, and its RING domain, a histone writer, can ubiquitylate H2A. Thus, our study reveals that PHF7 is a novel E3 ligase that can specifically ubiquitylate H2A through binding H3K4me3/me2 prior to histone-to-protamine exchange.


Assuntos
Histonas/metabolismo , Protaminas/metabolismo , Espermatogênese/genética , Ubiquitina-Proteína Ligases/fisiologia , Ubiquitinação/genética , Animais , Células Cultivadas , Montagem e Desmontagem da Cromatina/fisiologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Infertilidade Masculina/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais/genética , Testículo/metabolismo , Ubiquitina-Proteína Ligases/genética
4.
Development ; 142(6): 1073-82, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25758221

RESUMO

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.


Assuntos
Carcinogênese , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Células Germinativas/citologia , Oogênese/fisiologia , Proteínas de Ligação a RNA/metabolismo , Caracteres Sexuais , Células-Tronco Adultas/fisiologia , Animais , Proteínas de Drosophila/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento/genética , Técnicas de Silenciamento de Genes , Células Germinativas/fisiologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Homeostase/fisiologia , Processamento de Imagem Assistida por Computador , Masculino , Microscopia de Fluorescência , Proteínas de Ligação a RNA/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de RNA , Testículo/metabolismo
5.
Proc Biol Sci ; 284(1855)2017 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-28539513

RESUMO

PHD finger protein 7 (Phf7) is a male germline specific gene in Drosophila melanogaster that can trigger the male germline sexual fate and regulate spermatogenesis, and its human homologue can rescue fecundity defects in male flies lacking this gene. These findings prompted us to investigate conservation of reproductive strategies through studying the evolutionary origin of this gene. We find that Phf7 is present only in select species including mammals and some insects, whereas the closely related G2/M-phase specific E3 ubiquitin protein ligase (G2e3) is in the genome of most metazoans. Interestingly, phylogenetic analyses showed that vertebrate and insect Phf7 genes did not evolve from a common Phf7 ancestor but rather through independent duplication events from an ancestral G2e3 This is an example of parallel evolution in which a male germline factor evolved at least twice from a pre-existing template to develop new regulatory mechanisms of spermatogenesis.


Assuntos
Proteínas de Drosophila/genética , Evolução Molecular , Proteínas de Homeodomínio/genética , Espermatogênese/genética , Animais , Drosophila melanogaster , Duplicação Gênica , Genes de Insetos , Masculino , Mamíferos , Filogenia
6.
Curr Biol ; 33(9): 1825-1835.e3, 2023 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-37059098

RESUMO

Sexually dimorphic traits in morphologies are widely studied,1,2,3,4 but those in essential molecular pathways remain largely unexplored. Previous work showed substantial sex differences in Drosophila gonadal piRNAs,5 which guide PIWI proteins to silence selfish genetic elements, thereby safeguarding fertility.6,7,8 However, the genetic control mechanisms of piRNA sexual dimorphism remain unknown. Here, we showed that most sex differences in the piRNA program originate from the germ line rather than the gonadal somatic cells. Building on this, we dissected the contribution of sex chromosomes and cellular sexual identity toward the sex-specific germline piRNA program. We found that the presence of the Y chromosome is sufficient to recapitulate some aspects of the male piRNA program in a female cellular environment. Meanwhile, sexual identity controls the sexually divergent piRNA production from X-linked and autosomal loci, revealing a crucial input from sex determination into piRNA biogenesis. Sexual identity regulates piRNA biogenesis through Sxl, and this effect is mediated, in part, through chromatin proteins Phf7 and Kipferl. Together, our work delineated the genetic control of a sex-specific piRNA program, where sex chromosomes and sexual identity collectively sculpt an essential molecular trait.


Assuntos
Proteínas de Drosophila , Drosophila melanogaster , Animais , Feminino , Masculino , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , RNA de Interação com Piwi , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Drosophila/genética , Células Germinativas/metabolismo , Proteínas Argonautas/genética , Proteínas de Ligação a RNA/metabolismo , Proteínas de Homeodomínio/metabolismo
7.
Cell Rep ; 39(1): 110620, 2022 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-35385723

RESUMO

Establishing germ cell sexual identity is critical for development of male and female germline stem cells (GSCs) and production of sperm or eggs. Germ cells depend on signals from the somatic gonad to determine sex, but in organisms such as flies, mice, and humans, the sex chromosome genotype of the germ cells is also important for germline sexual development. How somatic signals and germ-cell-intrinsic cues combine to regulate germline sex determination is thus a key question. We find that JAK/STAT signaling in the GSC niche promotes male identity in germ cells, in part by activating the chromatin reader Phf7. Further, we find that JAK/STAT signaling is blocked in XX (female) germ cells through the action of the sex determination gene Sex lethal to preserve female identity. Thus, an important function of germline sexual identity is to control how GSCs respond to signals in their niche environment.


Assuntos
Proteínas de Drosophila , Células Germinativas , Processos de Determinação Sexual , Células-Tronco , Animais , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Feminino , Células Germinativas/metabolismo , Proteínas de Homeodomínio/metabolismo , Janus Quinases/metabolismo , Masculino , Fatores de Transcrição STAT/metabolismo , Processos de Determinação Sexual/genética , Processos de Determinação Sexual/fisiologia , Transdução de Sinais/fisiologia , Nicho de Células-Tronco , Células-Tronco/metabolismo
8.
Cell Rep ; 32(4): 107950, 2020 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-32726616

RESUMO

Spermatogenesis is a complex process of sperm generation, including mitosis, meiosis, and spermiogenesis. During spermiogenesis, histones in post-meiotic spermatids are removed from chromatin and replaced by protamines. Although histone-to-protamine exchange is important for sperm nuclear condensation, the underlying regulatory mechanism is still poorly understood. Here, we identify PHD finger protein 7 (PHF7) as an E3 ubiquitin ligase for histone H3K14 in post-meiotic spermatids. Generation of Phf7-deficient mice and Phf7 C160A knockin mice with impaired E3 ubiquitin ligase activity reveals defects in histone-to-protamine exchange caused by dysregulation of histone removal factor Bromodomain, testis-specific (BRDT) in early condensing spermatids. Surprisingly, E3 ubiquitin ligase activity of PHF7 on histone ubiquitination leads to stabilization of BRDT by attenuating ubiquitination of BRDT. Collectively, our findings identify PHF7 as a critical factor for sperm chromatin condensation and contribute to mechanistic understanding of fundamental phenomenon of histone-to-protamine exchange and potential for drug development for the male reproduction system.


Assuntos
Espermatogênese/genética , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Acetilação , Animais , Núcleo Celular/metabolismo , Cromatina/metabolismo , Montagem e Desmontagem da Cromatina , Técnicas de Introdução de Genes/métodos , Células HEK293 , Histonas/metabolismo , Humanos , Masculino , Meiose , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Nucleares/metabolismo , Proteínas Nucleares/fisiologia , Protaminas/metabolismo , Espermátides/metabolismo , Espermatogênese/fisiologia , Espermatozoides/metabolismo , Testículo/metabolismo , Ubiquitinação
9.
Genetics ; 206(4): 1939-1949, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28588035

RESUMO

A key aspect of germ cell development is to establish germline sexual identity and initiate a sex-specific developmental program to promote spermatogenesis or oogenesis. Previously, we have identified the histone reader Plant Homeodomain Finger 7 (PHF7) as an important regulator of male germline identity. To understand how PHF7 directs sexual differentiation of the male germline, we investigated the downstream targets of PHF7 by combining transcriptome analyses, which reveal genes regulated by Phf7, with genomic profiling of histone H3K4me2, the chromatin mark that is bound by PHF7. Through these genomic experiments, we identify a novel spermatocyte factor Receptor Accessory Protein Like 1 (REEPL1) that can promote spermatogenesis and whose expression is kept off by PHF7 in the spermatogonial stage. Loss of Reepl1 significantly rescues the spermatogenesis defects in Phf7 mutants, indicating that regulation of Reepl1 is an essential aspect of PHF7 function. Further, increasing REEPL1 expression facilitates spermatogenic differentiation. These results indicate that PHF7 controls spermatogenesis by regulating the expression patterns of important male germline genes.


Assuntos
Proteínas de Drosophila/genética , Proteínas de Homeodomínio/genética , Espermatócitos/metabolismo , Espermatogênese/genética , Animais , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citologia , Drosophila melanogaster/genética , Drosophila melanogaster/crescimento & desenvolvimento , Histonas/metabolismo , Proteínas de Homeodomínio/metabolismo , Masculino , Espermatócitos/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA