Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Prolif ; 57(4): e13567, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37921559

RESUMO

The successful progression of meiosis prophase I requires integrating information from the structural and molecular levels. In this study, we show that ZFP541 and KCTD19 work in the same genetic pathway to regulate the progression of male meiosis and thus fertility. The Zfp541 and/or Kctd19 knockout male mice show various structural and recombination defects including detached chromosome ends, aberrant localization of chromosome axis components and recombination proteins, and globally altered histone modifications. Further analyses on RNA-seq, ChIP-seq, and ATAC-seq data provide molecular evidence for the above defects and reveal that ZFP541/KCTD19 activates the expression of many genes by repressing several major transcription repressors. More importantly, we reveal an unexpected role of ZFP541/KCTD19 in directly modulating chromatin organization. These results suggest that ZFP541/KCTD19 simultaneously regulates the transcription cascade and chromatin organization to ensure the coordinated progression of multiple events at chromosome structural and biochemical levels during meiosis prophase I.


Assuntos
Cromatina , Fatores de Transcrição , Animais , Camundongos , Masculino , Cromatina/genética , Fatores de Transcrição/metabolismo , Complexo Sinaptonêmico/metabolismo , Processamento de Proteína Pós-Traducional , Meiose , Proteínas Cromossômicas não Histona/metabolismo
2.
Cell Rep ; 42(8): 112953, 2023 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-37542719

RESUMO

Meiotic crossovers are required for the faithful segregation of homologous chromosomes and to promote genetic diversity. However, it is unclear how crossover formation is regulated, especially on the XY chromosomes, which show a homolog only at the tiny pseudoautosomal region. Here, we show that ATF7IP2 is a meiosis-specific ortholog of ATF7IP and a partner of SETDB1. In the absence of ATF7IP2, autosomes show increased axis length and more crossovers; however, many XY chromosomes lose the obligatory crossover, although the overall XY axis length is also increased. Additionally, meiotic DNA double-strand break formation/repair may also be affected by altered histone modifications. Ultimately, spermatogenesis is blocked, and male mice are infertile. These findings suggest that ATF7IP2 constraints autosomal axis length and crossovers on autosomes; meanwhile, it also modulates XY chromosomes to establish meiotic sex chromosome inactivation for cell-cycle progression and to ensure XY crossover formation during spermatogenesis.


Assuntos
Meiose , Cromossomos Sexuais , Fatores de Transcrição , Animais , Masculino , Camundongos , Segregação de Cromossomos , Histona-Lisina N-Metiltransferase/genética , Espermatogênese/genética , Fatores de Transcrição/genética
3.
FASEB J ; 36(6): e22357, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35593531

RESUMO

The reproductive life span of females is largely determined by the number and quality of oocytes. Previously, we identified MEIOK21 as a meiotic recombination regulator required for male fertility. Here, we characterize the important roles of MEIOK21 in regulating female meiosis and oocyte number and quality. MEIOK21 localizes at recombination sites as a component of recombination bridges in oogenesis like in spermatogenesis. Meiok21-/- female mice show subfertility. Consistently, the size of the primordial follicle pool in Meiok21-/- females is only ~40% of wild-type females because a great number of oocytes with defects in meiotic recombination and/or synapsis are eliminated. Furthermore, the numbers of primordial and growing follicles show a more marked decrease in an age-dependent manner compared with wild-type females. Further analysis shows Meiok21-/- oocytes also have reduced rates of germinal vesicle breakdown and the first polar body extrusion when cultured in vitro, indicating poor oocyte quality. Additionally, Meiok21-/- oocytes have more chromosomes bearing a single distally localized crossover (chiasmata), suggesting a possible defect in crossover maturation. Taken together, our findings indicate critical roles for MEIOK21 in ensuring the number and quality of oocytes in the follicles.


Assuntos
Meiose , Oócitos , Animais , Feminino , Recombinação Homóloga , Masculino , Meiose/genética , Camundongos , Oócitos/metabolismo , Oogênese/genética , Folículo Ovariano
4.
Biol Reprod ; 107(1): 275-288, 2022 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-35191959

RESUMO

Meiosis is the foundation of sexual reproduction, and crossover recombination is one hallmark of meiosis. Crossovers establish the physical connections between homolog chromosomes (homologs) for their proper segregation and exchange DNA between homologs to promote genetic diversity in gametes and thus progenies. Aberrant crossover patterns, e.g., absence of the obligatory crossover, are the leading cause of infertility, miscarriage, and congenital disease. Therefore, crossover patterns have to be tightly controlled. During meiosis, loop/axis organized chromosomes provide the structural basis and regulatory machinery for crossover patterning. Accumulating evidence shows that chromosome axis length regulates the numbers and the positions of crossovers. In addition, recent studies suggest that alterations in axis length and the resultant alterations in crossover frequency may contribute to evolutionary adaptation. Here, current advances regarding these issues are reviewed, the possible mechanisms for axis length regulating crossover frequency are discussed, and important issues that need further investigations are suggested.


Assuntos
Segregação de Cromossomos , Recombinação Genética , Cromossomos , Meiose/genética
5.
PeerJ ; 9: e12210, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34631320

RESUMO

BACKGROUND: As a group of membrane-anchored proteins, the proteins containing a disintegrin and metalloprotease domain (ADAMs) control many biological processes, especially for male fertility. Mouse Adam21 was previously found to be specifically expressed in the somatic cells and germ cells of testes, but its functional role during spermatogenesis and male reproductive processes is still unknown. METHODS: Adam21-null mice were created using the CRISPR/Cas9 system. Quantitative real-time PCR was used for analyzing of gene expression. Histological, cytological and immunofluorescence staining were performed to analyze the phenotypes of mouse testis and epididymis. Intracellular lipid droplets (LDs) were detected by Oil red O (ORO) staining and BODIPY staining. Fertility and sperm characteristics were also detected. RESULTS: Here, we successfully generated an Adam21 conventional knockout mouse model via CRISPR/Cas9 technology so that we can explore its potential role in male reproduction. We found that male mice lacking Adam21 have normal fertility without any detectable defects in spermatogenesis or sperm motility. Histological analysis of the seminiferous epithelium showed no obvious spermatogenesis difference between Adam21-null and wild-type mice. Cytological analysis revealed no detectable defects in meiotic progression, neither Sertoli cells nor Leydig cells displayed any defect compared with that of the control mice. All these results suggest that Adam21 might not be essential for male fertility in mice, and its potential function still needs further investigation.

6.
Asian J Androl ; 23(6): 562-571, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33533735

RESUMO

Repairing DNA double-strand breaks (DSBs) with homologous chromosomes as templates is the hallmark of meiosis. The critical outcome of meiotic homologous recombination is crossovers, which ensure faithful chromosome segregation and promote genetic diversity of progenies. Crossover patterns are tightly controlled and exhibit three characteristics: obligatory crossover, crossover interference, and crossover homeostasis. Aberrant crossover patterns are the leading cause of infertility, miscarriage, and congenital disease. Crossover recombination occurs in the context of meiotic chromosomes, and it is tightly integrated with and regulated by meiotic chromosome structure both locally and globally. Meiotic chromosomes are organized in a loop-axis architecture. Diverse evidence shows that chromosome axis length determines crossover frequency. Interestingly, short chromosomes show different crossover patterns compared to long chromosomes. A high frequency of human embryos are aneuploid, primarily derived from female meiosis errors. Dramatically increased aneuploidy in older women is the well-known "maternal age effect." However, a high frequency of aneuploidy also occurs in young women, derived from crossover maturation inefficiency in human females. In addition, frequency of human aneuploidy also shows other age-dependent alterations. Here, current advances in the understanding of these issues are reviewed, regulation of crossover patterns by meiotic chromosomes are discussed, and issues that remain to be investigated are suggested.


Assuntos
Divisão Celular/fisiologia , Segregação de Cromossomos/genética , Meiose/genética , Segregação de Cromossomos/fisiologia , Humanos , Recombinação Genética
7.
Nucleic Acids Res ; 48(12): 6624-6639, 2020 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-32463460

RESUMO

Repair of DNA double-strand breaks (DSBs) with homologous chromosomes is a hallmark of meiosis that is mediated by recombination 'bridges' between homolog axes. This process requires cooperation of DMC1 and RAD51 to promote homology search and strand exchange. The mechanism(s) regulating DMC1/RAD51-ssDNA nucleoprotein filament and the components of 'bridges' remain to be investigated. Here we show that MEIOK21 is a newly identified component of meiotic recombination bridges and is required for efficient formation of DMC1/RAD51 foci. MEIOK21 dynamically localizes on chromosomes from on-axis foci to 'hanging foci', then to 'bridges', and finally to 'fused foci' between homolog axes. Its chromosome localization depends on DSBs. Knockout of Meiok21 decreases the numbers of HSF2BP and DMC1/RAD51 foci, disrupting DSB repair, synapsis and crossover recombination and finally causing male infertility. Therefore, MEIOK21 is a novel recombination factor and probably mediates DMC1/RAD51 recruitment to ssDNA or their stability on chromosomes through physical interaction with HSF2BP.


Assuntos
Proteínas de Ciclo Celular/genética , Proteínas de Ligação a DNA/genética , Recombinação Homóloga/genética , Infertilidade Masculina/genética , Espermatogênese/genética , Animais , Proteínas de Transporte/genética , Pareamento Cromossômico/genética , Cromossomos/genética , Quebras de DNA de Cadeia Dupla , Reparo do DNA/genética , DNA de Cadeia Simples/genética , Técnicas de Inativação de Genes , Proteínas de Choque Térmico/genética , Masculino , Meiose/genética , Camundongos , Camundongos Knockout , Proteínas de Ligação a Fosfato/genética , Rad51 Recombinase/genética
8.
Bioessays ; 41(10): e1800221, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31424607

RESUMO

A striking feature of human female sexual reproduction is the high level of gametes that exhibit an aberrant number of chromosomes (aneuploidy). A high baseline observed in women of prime reproductive age is followed by a dramatic increase in older women. Proper chromosome segregation requires one or more DNA crossovers (COs) between homologous maternal and paternal chromosomes, in combination with cohesion between sister chromatid arms. In human females, CO designations occur normally, according to the dictates of CO interference, giving early CO-fated intermediates. However, ≈25% of these intermediates fail to mature to final CO products. This effect explains the high baseline of aneuploidy and is predicted to synergize with age-dependent cohesion loss to explain the maternal age effect. Here, modern advances in the understanding of crossing over and CO interference are reviewed, the implications of human female CO maturation inefficiency are further discussed, and areas of interest for future studies are suggested.


Assuntos
Aneuploidia , Cromossomos Humanos , Fatores Etários , Segregação de Cromossomos , Feminino , Humanos , Masculino , Meiose , Fatores Sexuais
9.
Cell ; 177(2): 326-338.e16, 2019 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-30879787

RESUMO

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


Assuntos
Troca Genética/genética , Troca Genética/fisiologia , Animais , Núcleo Celular , Segregação de Cromossomos , Cromossomos/genética , Cromossomos/fisiologia , Simulação por Computador , Feminino , Genética Populacional/métodos , Recombinação Homóloga/genética , Humanos , Solanum lycopersicum/genética , Masculino , Meiose/genética , Recombinação Genética/genética , Complexo Sinaptonêmico
10.
Asian J Androl ; 21(2): 183-189, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30333325

RESUMO

Globozoospermia has been reported to be a rare but severe causation of male infertility, which results from the failure of acrosome biogenesis and sperm head shaping. Variants of dpy-19-like 2 (DPY19L2) are highly related to globozoospermia, but related investigations have been mainly performed in patients from Western countries. Here, we performed a screening of DPY19L2 variants in a cohort of Chinese globozoospermic patients and found that five of nine patients carried DPY19L2 deletions and the other four patients contained novel DPY19L2 point mutations, as revealed by whole-exome sequencing. Patient 3 (P3) contained a heterozygous variant (c.2126+5G>A), P6 contained a homozygous nonsense mutation (c.1720C>T, p.Arg574*), P8 contained compound heterozygous variants (c.1182-1184delATC, p.Leu394_Ser395delinsPhe; c.368A>T, p.His123Arg), and P9 contained a heterozygous variant (c.1182-1184delATCTT, frameshift). We also reported intracytoplasmic sperm injection (ICSI) outcomes in the related patients, finding that ICSI followed by assisted oocyte activation (AOA) with calcium ionophore achieved high rates of live births. In summary, the infertility of these patients results from DPY19L2 dysfunction and can be treated by ICSI together with AOA.


Assuntos
Códon sem Sentido , Proteínas de Membrana/genética , Mutação Puntual , Deleção de Sequência , Injeções de Esperma Intracitoplásmicas , Teratozoospermia/genética , Acrossomo , Adulto , China , Feminino , Humanos , Masculino , Gravidez , Resultado da Gravidez , Taxa de Gravidez , Cabeça do Espermatozoide , Sequenciamento do Exoma
11.
Am J Hum Genet ; 103(2): 188-199, 2018 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-30032984

RESUMO

Acephalic spermatozoa syndrome is a severe teratozoospermia that leads to male infertility. Our previous work showed that biallelic SUN5 mutations are responsible for acephalic spermatozoa syndrome in about half of affected individuals, while pathogenic mechanisms in the other individuals remain to be elucidated. Here, we identified a homozygous nonsense mutation in the testis-specific gene PMFBP1 using whole-exome sequencing in a consanguineous family with two infertile brothers with acephalic spermatozoa syndrome. Sanger sequencing of PMFBP1 in ten additional infertile men with acephalic spermatozoa syndrome and without SUN5 mutations revealed two homozygous variants and one compound heterozygous variant. The disruption of Pmfbp1 in male mice led to infertility due to the production of acephalic spermatozoa and the disruption of PMFBP1's cooperation with SUN5 and SPATA6, which plays a role in connecting sperm head to the tail. PMFBP1 mutation-associated male infertility could be successfully overcome by intracytoplasmic sperm injection (ICSI) in both mouse and human. Thus, mutations in PMFBP1 are an important cause of infertility in men with acephalic spermatozoa syndrome.


Assuntos
Infertilidade Masculina/genética , Mutação/genética , Proteínas/genética , Espermatozoides/patologia , Animais , Proteínas do Citoesqueleto/genética , Exoma/genética , Feminino , Homozigoto , Humanos , Masculino , Camundongos , Camundongos Endogâmicos ICR , Injeções de Esperma Intracitoplásmicas/métodos , Síndrome , Sequenciamento do Exoma/métodos
12.
J Cell Biol ; 217(6): 2103-2119, 2018 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-29618492

RESUMO

Testosterone is indispensable for sexual development and maintaining male characteristics, and deficiency of this hormone results in primary or late-onset hypogonadism (LOH). Testosterone is primarily produced in Leydig cells, where autophagy has been reported to be extremely active. However, the functional role of autophagy in testosterone synthesis remains unknown. In this study, we show that steroidogenic cell-specific disruption of autophagy influenced the sexual behavior of aging male mice because of a reduction in serum testosterone, which is similar to the symptoms of LOH. The decline in testosterone was caused mainly by a defect in cholesterol uptake in autophagy-deficient Leydig cells. Further studies revealed that once autophagic flux was disrupted, Na+/H+ exchanger regulatory factor 2 (NHERF2) accumulated in Leydig cells, resulting in the down-regulation of scavenger receptor class B, type I (SR-BI) and eventually leading to insufficient cholesterol supply. Collectively, these results reveal that autophagy promotes cholesterol uptake into Leydig cells by eliminating NHERF2, suggesting that dysfunction of autophagy might be causal in the loss of testosterone production in some patients.


Assuntos
Autofagia , Colesterol/metabolismo , Células Intersticiais do Testículo/citologia , Células Intersticiais do Testículo/metabolismo , Testosterona/biossíntese , Adulto , Sequência de Aminoácidos , Animais , Autofagia/efeitos dos fármacos , Proteínas Relacionadas à Autofagia/metabolismo , Antígenos CD36/metabolismo , Gonadotropina Coriônica/farmacologia , Regulação para Baixo/efeitos dos fármacos , Humanos , Células Intersticiais do Testículo/efeitos dos fármacos , Masculino , Camundongos , Modelos Biológicos , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Comportamento Sexual Animal/efeitos dos fármacos , Trocadores de Sódio-Hidrogênio/química , Trocadores de Sódio-Hidrogênio/metabolismo , Testosterona/sangue , Adulto Jovem
13.
Cell Mol Life Sci ; 75(10): 1707-1722, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29427077

RESUMO

The maternal-to-zygotic transition (MZT) is essential for the developmental control handed from maternal products to newly synthesized zygotic genome in the earliest stages of embryogenesis, including maternal component (mRNAs and proteins) degradation and zygotic genome activation (ZGA). Various protein post-translational modifications have been identified during the MZT, such as phosphorylation, methylation and ubiquitination. Precise post-translational regulation mechanisms are essential for the timely transition of early embryonic development. In this review, we summarize recent progress regarding the molecular mechanisms underlying post-translational regulation of maternal component degradation and ZGA during the MZT and discuss some important issues in the field.


Assuntos
Desenvolvimento Embrionário/genética , RNA Mensageiro Estocado/genética , RNA Mensageiro Estocado/metabolismo , Zigoto/metabolismo , Animais , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Gravidez , Processamento de Proteína Pós-Traducional
14.
J Biol Chem ; 293(7): 2395-2407, 2018 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-29298896

RESUMO

Acephalic spermatozoa syndrome has been reported for many decades; it is characterized by very few intact spermatozoa and tailless sperm heads in the semen and causes severe male infertility. The only gene in which mutations have been found to be associated with this syndrome encodes Sad1 and UNC84 domain-containing 5 (SUN5), a testis-specific nuclear envelope protein. The functional role of SUN5 has been well-studied in mouse models, but the molecular basis for the pathogenic effects of mutations in the human SUN5 gene remains elusive. Here, we report a new SUN5 mutation (c.475C→T; p.Arg159*), and explore the pathogenic effects of all known SUN5 mutations on acephalic spermatozoa syndrome. Using an artificial splicing system, we found that the intronic mutation affects the splicing of SUN5 mRNA, yielding a premature stop codon that results in a truncated SUN5 protein. We also found that SUN5 interacts with the coupling apparatus protein DnaJ heat shock protein family (Hsp40) member B13 (DNAJB13) during spermatogenesis, and the substitutions in the SUN5 SUN domain impair its interaction with DNAJB13. Furthermore, we observed that many SUN5 mutations affect the secondary structure of the protein and influence its folding and cellular localization. In summary, our findings indicate an interaction of SUN5 with DNAJB13 during spermatogenesis, provide mechanistic insights into the functional role of this interaction in sperm head-tail integration, and elucidate the molecular etiology of acephalic spermatozoa syndrome-associated SUN5 mutations.


Assuntos
Proteínas de Membrana/genética , Proteínas/genética , Teratozoospermia/genética , Adulto , Animais , Proteínas Reguladoras de Apoptose , Proteínas de Choque Térmico HSP40/genética , Proteínas de Choque Térmico HSP40/metabolismo , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Humanos , Íntrons , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Chaperonas Moleculares , Mutação Puntual , Ligação Proteica , Proteínas/metabolismo , Splicing de RNA , Espermatogênese , Espermatozoides/citologia , Espermatozoides/metabolismo , Teratozoospermia/metabolismo
15.
Genetics ; 208(3): 1181-1194, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29301906

RESUMO

Meiosis is a specific type of cell division that is essential for sexual reproduction in most eukaryotes. Mitochondria are crucial cellular organelles that play important roles in reproduction, though the detailed mechanism by which the mitochondrial respiratory chain functions during meiosis remains elusive. Here, we show that components of the respiratory chain (Complexes I-V) play essential roles in meiosis initiation during the sporulation of budding yeast, Saccharomyces cerevisiae Any functional defects in the Complex I component Ndi1p resulted in the abolishment of sporulation. Further studies revealed that respiratory deficiency resulted in the failure of premeiotic DNA replication due to insufficient IME1 expression. In addition, respiration promoted the expression of RIM101, whose product inhibits Smp1p, a negative transcriptional regulator of IME1, to promote meiosis initiation. In summary, our studies unveiled the close relationship between mitochondria and sporulation, and uncover a novel meiosis initiation pathway that is regulated by the respiratory chain.


Assuntos
Transporte de Elétrons , Meiose , Saccharomyces cerevisiae/fisiologia , Trifosfato de Adenosina/metabolismo , Complexo I de Transporte de Elétrons/metabolismo , Regulação Fúngica da Expressão Gênica , Modelos Biológicos , Proteínas Repressoras/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Deleção de Sequência , Esporos Fúngicos , Transcrição Gênica
16.
Elife ; 62017 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-28945193

RESUMO

SUN (Sad1 and UNC84 domain containing)-domain proteins are reported to reside on the nuclear membrane playing distinct roles in nuclear dynamics. SUN5 is a new member of the SUN family, with little knowledge regarding its function. Here, we generated Sun5-/- mice and found that male mice were infertile. Most Sun5-null spermatozoa displayed a globozoospermia-like phenotype but they were actually acephalic spermatozoa. Additional studies revealed that SUN5 was located in the neck of the spermatozoa, anchoring sperm head to the tail, and without functional SUN5 the sperm head to tail coupling apparatus was detached from nucleus during spermatid elongation. Finally, we found that healthy heterozygous offspring could be obtained via intracytoplasmic injection of Sun5-mutated sperm heads for both male mice and patients. Our studies reveal the essential role of SUN5 in anchoring sperm head to the tail and provide a promising way to treat this kind of acephalic spermatozoa-associated male infertility.


Assuntos
Proteínas de Membrana/metabolismo , Cabeça do Espermatozoide/fisiologia , Cauda do Espermatozoide/fisiologia , Espermatogênese , Animais , Masculino , Proteínas de Membrana/deficiência , Camundongos Knockout , Membrana Nuclear/metabolismo
17.
EMBO Rep ; 18(2): 205-216, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28073917

RESUMO

The functional role of the ubiquitin-proteasome pathway during maternal-to-zygotic transition (MZT) remains to be elucidated. Here we show that the E3 ubiquitin ligase, Rnf114, is highly expressed in mouse oocytes and that knockdown of Rnf114 inhibits development beyond the two-cell stage. To study the underlying mechanism, we identify its candidate substrates using a 9,000-protein microarray and validate them using an in vitro ubiquitination system. We show that five substrates could be degraded by RNF114-mediated ubiquitination, including TAB1. Furthermore, the degradation of TAB1 in mouse early embryos is required for MZT, most likely because it activates the NF-κB pathway. Taken together, our study uncovers that RNF114-mediated ubiquitination and degradation of TAB1 activate the NF-κB pathway during MZT, and thus directly link maternal clearance to early embryo development.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Herança Materna , Ubiquitina-Proteína Ligases/metabolismo , Zigoto/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Análise por Conglomerados , Desenvolvimento Embrionário/genética , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Camundongos , NF-kappa B/metabolismo , Oócitos/metabolismo , Poliubiquitina/metabolismo , Proteólise , Transdução de Sinais , Especificidade por Substrato , Ubiquitina-Proteína Ligases/genética
18.
Development ; 144(3): 441-451, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-28003215

RESUMO

Sirt1 is a member of the sirtuin family of proteins and has important roles in numerous biological processes. Sirt1-/- mice display an increased frequency of abnormal spermatozoa, but the mechanism of Sirt1 in spermiogenesis remains largely unknown. Here, we report that Sirt1 might be directly involved in spermiogenesis in germ cells but not in steroidogenic cells. Germ cell-specific Sirt1 knockout mice were almost completely infertile; the early mitotic and meiotic progression of germ cells in spermatogenesis were not obviously affected after Sirt1 depletion, but subsequent spermiogenesis was disrupted by a defect in acrosome biogenesis, which resulted in a phenotype similar to that observed in human globozoospermia. In addition, LC3 and Atg7 deacetylation was disrupted in spermatids after knocking out Sirt1, which affected the redistribution of LC3 from the nucleus to the cytoplasm and the activation of autophagy. Furthermore, Sirt1 depletion resulted in the failure of LC3 to be recruited to Golgi apparatus-derived vesicles and in the failure of GOPC and PICK1 to be recruited to nucleus-associated acrosomal vesicles. Taken together, these findings reveal that Sirt1 has a novel physiological function in acrosome biogenesis.


Assuntos
Acrossomo/fisiologia , Sirtuína 1/fisiologia , Espermatogênese/fisiologia , Acrossomo/patologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Autofagia/genética , Autofagia/fisiologia , Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular , Modelos Animais de Doenças , Proteínas da Matriz do Complexo de Golgi , Humanos , Infertilidade Masculina/etiologia , Infertilidade Masculina/genética , Infertilidade Masculina/patologia , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Microscopia Eletrônica de Transmissão , Modelos Biológicos , Proteínas Nucleares/metabolismo , Fenótipo , Sirtuína 1/deficiência , Sirtuína 1/genética , Espermatogênese/genética , Espermatozoides/patologia , Espermatozoides/fisiologia , Esteroides/biossíntese , Teratozoospermia/etiologia , Teratozoospermia/patologia
19.
Open Biol ; 6(8)2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27512143

RESUMO

Cardiovascular disease (CVD) is the leading cause of the death worldwide. An increasing number of studies have found that autophagy is involved in the progression or prevention of CVD. However, the precise mechanism of autophagy in CVD, especially the myocardial ischaemia-reperfusion injury (MI/R injury), is unclear and controversial. Here, we show that the cardiomyocyte-specific disruption of autophagy by conditional knockout of Atg7 leads to severe contractile dysfunction, myofibrillar disarray and vacuolar cardiomyocytes. A negative cytoskeleton organization regulator, CLP36, was found to be accumulated in Atg7-deficient cardiomyocytes. The cardiomyocyte-specific knockout of Atg7 aggravates the MI/R injury with cardiac hypertrophy, contractile dysfunction, myofibrillar disarray and severe cardiac fibrosis, most probably due to CLP36 accumulation in cardiomyocytes. Altogether, this work reveals autophagy may protect cardiomyocytes from the MI/R injury through the clearance of CLP36, and these findings define a novel relationship between autophagy and the regulation of stress fibre in heart.


Assuntos
Proteína 7 Relacionada à Autofagia/genética , Proteínas com Domínio LIM/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , Miócitos Cardíacos/metabolismo , Fatores de Transcrição/metabolismo , Animais , Autofagia , Células Cultivadas , Técnicas de Inativação de Genes , Camundongos , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/patologia , Estresse Oxidativo
20.
Nucleic Acids Res ; 44(20): 9681-9697, 2016 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-27431324

RESUMO

Meiotic recombination is essential for fertility in most sexually reproducing species, but the molecular mechanisms underlying this process remain poorly understood in mammals. Here, we show that RNF20-mediated H2B ubiquitination is required for meiotic recombination. A germ cell-specific knockout of the H2B ubiquitination E3 ligase RNF20 results in complete male infertility. The Stra8-Rnf20-/- spermatocytes arrest at the pachytene stage because of impaired programmed double-strand break (DSB) repair. Further investigations reveal that the depletion of RNF20 in the germ cells affects chromatin relaxation, thus preventing programmed DSB repair factors from being recruited to proper positions on the chromatin. The gametogenetic defects of the H2B ubiquitination deficient cells could be partially rescued by forced chromatin relaxation. Taken together, our results demonstrate that RNF20/Bre1p-mediated H2B ubiquitination regulates meiotic recombination by promoting chromatin relaxation, and suggest an old drug may provide a new way to treat some oligo- or azoospermia patients with chromatin relaxation disorders.


Assuntos
Montagem e Desmontagem da Cromatina , Cromatina/genética , Cromatina/metabolismo , Histonas/metabolismo , Meiose , Recombinação Genética , Proteínas Adaptadoras de Transdução de Sinal/deficiência , Animais , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Feminino , Células Germinativas/metabolismo , Infertilidade Masculina/genética , Masculino , Camundongos , Camundongos Knockout , Estágio Paquíteno/genética , Espermatócitos/metabolismo , Espermatogênese/genética , Ubiquitina-Proteína Ligases/deficiência , Ubiquitinação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...