Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nature ; 612(7941): 725-731, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36517592

RESUMO

Ribosomes are highly sophisticated translation machines that have been demonstrated to be heterogeneous in the regulation of protein synthesis1,2. Male germ cell development involves complex translational regulation during sperm formation3. However, it remains unclear whether translation during sperm formation is performed by a specific ribosome. Here we report a ribosome with a specialized nascent polypeptide exit tunnel, RibosomeST, that is assembled with the male germ-cell-specific protein RPL39L, the paralogue of core ribosome (RibosomeCore) protein RPL39. Deletion of RibosomeST in mice causes defective sperm formation, resulting in substantially reduced fertility. Our comparison of single-particle cryo-electron microscopy structures of ribosomes from mouse kidneys and testes indicates that RibosomeST features a ribosomal polypeptide exit tunnel of distinct size and charge states compared with RibosomeCore. RibosomeST predominantly cotranslationally regulates the folding of a subset of male germ-cell-specific proteins that are essential for the formation of sperm. Moreover, we found that specialized functions of RibosomeST were not replaceable by RibosomeCore. Taken together, identification of this sperm-specific ribosome should greatly expand our understanding of ribosome function and tissue-specific regulation of protein expression pattern in mammals.


Assuntos
Fertilidade , Ribossomos , Espermatozoides , Animais , Masculino , Camundongos , Microscopia Crioeletrônica/métodos , Peptídeos/química , Peptídeos/metabolismo , Biossíntese de Proteínas , Dobramento de Proteína , Ribossomos/metabolismo , Espermatozoides/citologia , Espermatozoides/metabolismo , Fertilidade/fisiologia , Especificidade de Órgãos , Proteínas Ribossômicas , Rim/citologia , Testículo/citologia
2.
Mol Cell ; 80(3): 525-540.e9, 2020 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-33068521

RESUMO

Well-balanced and timed metabolism is essential for making a high-quality egg. However, the metabolic framework that supports oocyte development remains poorly understood. Here, we obtained the temporal metabolome profiles of mouse oocytes during in vivo maturation by isolating large number of cells at key stages. In parallel, quantitative proteomic analyses were conducted to bolster the metabolomic data, synergistically depicting the global metabolic patterns in oocytes. In particular, we discovered the metabolic features during meiotic maturation, such as the fall in polyunsaturated fatty acids (PUFAs) level and the active serine-glycine-one-carbon (SGOC) pathway. Using functional approaches, we further identified the key targets mediating the action of PUFA arachidonic acid (ARA) on meiotic maturation and demonstrated the control of epigenetic marks in maturing oocytes by SGOC network. Our data serve as a broad resource on the dynamics occurring in metabolome and proteome during oocyte maturation.


Assuntos
Meiose/fisiologia , Oócitos/metabolismo , Animais , Epigênese Genética/genética , Ácidos Graxos Insaturados/metabolismo , Feminino , Metaboloma/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Oogênese/genética , Oogênese/fisiologia , Proteoma/metabolismo , Proteômica
3.
Mol Cell Proteomics ; 22(1): 100481, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36496143

RESUMO

Meiotic maturation is an intricate and precisely regulated process orchestrated by various pathways and numerous proteins. However, little is known about the proteome landscape during oocytes maturation. Here, we obtained the temporal proteomic profiles of mouse oocytes during in vivo maturation. We successfully quantified 4694 proteins from 4500 oocytes in three key stages (germinal vesicle, germinal vesicle breakdown, and metaphase II). In particular, we discovered the novel proteomic features during oocyte maturation, such as the active Skp1-Cullin-Fbox pathway and an increase in mRNA decay-related proteins. Using functional approaches, we further identified the key factors controlling the histone acetylation state in oocytes and the vital proteins modulating meiotic cell cycle. Taken together, our data serve as a broad resource on the dynamics occurring in oocyte proteome and provide important knowledge to better understand the molecular mechanisms during germ cell development.


Assuntos
Proteoma , Proteômica , Camundongos , Animais , Proteoma/metabolismo , Oogênese , Oócitos/metabolismo , Núcleo Celular/metabolismo , Meiose
4.
Mol Cell Proteomics ; 22(6): 100564, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37146716

RESUMO

Spermatogenesis defects are important for male infertility; however, the etiology and pathogenesis are still unknown. Herein, we identified two loss-of-function mutations of STK33 in seven individuals with non-obstructive azoospermia. Further functional studies of these frameshift and nonsense mutations revealed that Stk33-/KI male mice were sterile, and Stk33-/KI sperm were abnormal with defects in the mitochondrial sheath, fibrous sheath, outer dense fiber, and axoneme. Stk33KI/KI male mice were subfertile and had oligoasthenozoospermia. Differential phosphoproteomic analysis and in vitro kinase assay identified novel phosphorylation substrates of STK33, fibrous sheath components A-kinase anchoring protein 3 and A-kinase anchoring protein 4, whose expression levels decreased in testis after deletion of Stk33. STK33 regulated the phosphorylation of A-kinase anchoring protein 3/4, affected the assembly of fibrous sheath in the sperm, and played an essential role in spermiogenesis and male infertility.


Assuntos
Proteínas de Ancoragem à Quinase A , Infertilidade Masculina , Humanos , Masculino , Camundongos , Animais , Proteínas de Ancoragem à Quinase A/metabolismo , Sêmen/metabolismo , Espermatozoides/metabolismo , Espermatogênese/fisiologia , Cauda do Espermatozoide/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Infertilidade Masculina/genética , Infertilidade Masculina/metabolismo , Flagelos/metabolismo
5.
J Proteome Res ; 23(6): 2137-2147, 2024 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-38787631

RESUMO

N-glycosylation is one of the most universal and complex protein post-translational modifications (PTMs), and it is involved in many physiological and pathological activities. Owing to the low abundance of N-glycoproteins, enrichment of N-glycopeptides for mass spectrometry analysis usually requires a large amount of peptides. Additionally, oocyte protein N-glycosylation has not been systemically characterized due to the limited sample amount. Here, we developed a glycosylation enrichment method based on lectin and a single-pot, solid-phase-enhanced sample preparation (SP3) technology, termed lectin-based SP3 technology (LectinSP3). LectinSP3 immobilized lectin on the SP3 beads for N-glycopeptide enrichment. It could identify over 1100 N-glycosylation sites and 600 N-glycoproteins from 10 µg of mouse testis peptides. Furthermore, using the LectinSP3 method, we characterized the N-glycoproteome of 1000 mouse oocytes in three replicates and identified a total of 363 N-glycosylation sites from 215 N-glycoproteins. Bioinformatics analysis revealed that these oocyte N-glycoproteins were mainly enriched in cell adhesion, fertilization, and sperm-egg recognition. Overall, the LectinSP3 method has all procedures performed in one tube, using magnetic beads. It is suitable for analysis of a low amount of samples and is expected to be easily adaptable for automation. In addition, our mouse oocyte protein N-glycosylation profiling could help further characterize the regulation of oocyte functions.


Assuntos
Glicopeptídeos , Glicoproteínas , Lectinas , Oócitos , Proteômica , Animais , Oócitos/metabolismo , Camundongos , Glicosilação , Glicoproteínas/metabolismo , Glicoproteínas/química , Glicoproteínas/análise , Lectinas/química , Lectinas/metabolismo , Proteômica/métodos , Feminino , Glicopeptídeos/análise , Glicopeptídeos/química , Processamento de Proteína Pós-Traducional , Masculino , Testículo/metabolismo , Testículo/química , Proteoma/análise , Proteoma/metabolismo
6.
J Biol Chem ; 299(10): 105183, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37611828

RESUMO

Emerging research and clinical evidence suggest that the metabolic activity of oocytes may play a pivotal role in reproductive anomalies. However, the intrinsic mechanisms governing oocyte development regulated by metabolic enzymes remain largely unknown. Our investigation demonstrates that geranylgeranyl diphosphate synthase1 (Ggps1), the crucial enzyme in the mevalonate pathway responsible for synthesizing isoprenoid metabolite geranylgeranyl pyrophosphate from farnesyl pyrophosphate, is essential for oocyte maturation in mice. Our findings reveal that the deletion of Ggps1 that prevents protein prenylation in fully grown oocytes leads to subfertility and offspring metabolic defects without affecting follicle development. Oocytes that lack Ggps1 exhibit disrupted mitochondrial homeostasis and the mitochondrial defects arising from oocytes are inherited by the fetal offspring. Mechanistically, the excessive farnesylation of mitochondrial ribosome protein, Dap3, and decreased levels of small G proteins mediate the mitochondrial dysfunction induced by Ggps1 deficiency. Additionally, a significant reduction in Ggps1 levels in oocytes is accompanied by offspring defects when females are exposed to a high-cholesterol diet. Collectively, this study establishes that mevalonate pathway-protein prenylation is vital for mitochondrial function in oocyte maturation and provides evidence that the disrupted protein prenylation resulting from an imbalance between farnesyl pyrophosphate and geranylgeranyl pyrophosphate is the major mechanism underlying impairment of oocyte quality induced by high cholesterol.

7.
Development ; 148(13)2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-34104941

RESUMO

Zygotic genomic activation (ZGA) is a landmark event in the maternal-to-zygotic transition (MZT), and the regulation of ZGA by maternal factors remains to be elucidated. In this study, the depletion of maternal ring finger protein 114 (RNF114), a ubiquitin E3 ligase, led to developmental arrest of two-cell mouse embryos. Using immunofluorescence and transcriptome analysis, RNF114 was proven to play a crucial role in major ZGA. To study the underlying mechanism, we performed protein profiling in mature oocytes and found a potential substrate for RNF114, chromobox 5 (CBX5), ubiquitylation and degradation of which was regulated by RNF114. The overexpression of CBX5 prevented embryonic development and impeded major ZGA. Furthermore, TAB1 was abnormally accumulated in mutant two-cell embryos, which was consistent with the result of in vitro knockdown of Rnf114. Knockdown of Cbx5 or Tab1 in maternal RNF114-depleted embryos partially rescued developmental arrest and the defect of major ZGA. In summary, our study reveals that maternal RNF114 plays a precise role in degrading some important substrates during the MZT, the misregulation of which may impede the appropriate activation of major ZGA in mouse embryos.


Assuntos
Desenvolvimento Embrionário/fisiologia , Genoma , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Zigoto/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Homólogo 5 da Proteína Cromobox , Proteínas Cromossômicas não Histona/genética , Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Sistema de Sinalização das MAP Quinases/genética , Camundongos , Fatores de Transcrição/metabolismo , Transcriptoma
8.
Brief Bioinform ; 23(4)2022 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-35656712

RESUMO

Multiplexed single-cell proteomes (SCPs) quantification by mass spectrometry greatly improves the SCP coverage. However, it still suffers from a low number of protein identifications and there is much room to boost proteins identification by computational methods. In this study, we present a novel framework DeepSCP, utilizing deep learning to boost SCP coverage. DeepSCP constructs a series of features of peptide-spectrum matches (PSMs) by predicting the retention time based on the multiple SCP sample sets and fragment ion intensities based on deep learning, and predicts PSM labels with an optimized-ensemble learning model. Evaluation of DeepSCP on public and in-house SCP datasets showed superior performances compared with other state-of-the-art methods. DeepSCP identified more confident peptides and proteins by controlling q-value at 0.01 using target-decoy competition method. As a convenient and low-cost computing framework, DeepSCP will help boost single-cell proteome identification and facilitate the future development and application of single-cell proteomics.


Assuntos
Aprendizado Profundo , Proteoma , Peptídeos/química , Proteoma/metabolismo , Proteômica/métodos , Espectrometria de Massas em Tandem/métodos
9.
Mol Cell Proteomics ; 21(8): 100267, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35809850

RESUMO

Oocyte maturation is pertinent to the success of in vitro maturation (IVM), which is used to overcome female infertility, and produced over 5000 live births worldwide. However, the quality of human IVM oocytes has not been investigated at single-cell proteome level. Here, we quantified 2094 proteins in human oocytes during in vitro and in vivo maturation (IVO) by single-cell proteomic analysis and identified 176 differential proteins between IVO and germinal vesicle oocytes and 45 between IVM and IVO oocytes including maternal effect proteins, with potential contribution to the clinically observed decreased fertilization, implantation, and birth rates using human IVM oocytes. IVM and IVO oocytes showed separate clusters in principal component analysis, with higher inter-cell variability among IVM oocytes, and have little correlation between mRNA and protein changes during maturation. The patients with the most aberrantly expressed proteins in IVM oocytes had the lowest level of estradiol per mature follicle on trigger day. Our data provide a rich resource to evaluate effect of IVM on oocyte quality and study mechanism of oocyte maturation.


Assuntos
Técnicas de Maturação in Vitro de Oócitos , Proteômica , Feminino , Humanos , Oócitos , Oogênese , Análise de Célula Única
10.
BMC Biol ; 21(1): 89, 2023 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-37069605

RESUMO

BACKGROUND: Sperm is formed through spermiogenesis, a highly complex process involving chromatin condensation that results in cessation of transcription. mRNAs required for spermiogenesis are transcribed at earlier stages and translated in a delayed fashion during spermatid formation. However, it remains unknown that how these repressed mRNAs are stabilized. RESULTS: Here we report a Miwi-interacting testis-specific and spermiogenic arrest protein, Ck137956, which we rename Tssa. Deletion of Tssa led to male sterility and absence of sperm formation. The spermiogenesis arrested at the round spermatid stage and numerous spermiogenic mRNAs were down-regulated in Tssa-/- mice. Deletion of Tssa disrupted the localization of Miwi to chromatoid body, a specialized assembly of cytoplasmic messenger ribonucleoproteins (mRNPs) foci present in germ cells. We found that Tssa interacted with Miwi in repressed mRNPs and stabilized Miwi-interacting spermiogenesis-essential mRNAs. CONCLUSIONS: Our findings indicate that Tssa is indispensable in male fertility and has critical roles in post-transcriptional regulations by interacting with Miwi during spermiogenesis.


Assuntos
Proteínas Argonautas , Sêmen , Espermatogênese , Animais , Masculino , Camundongos , Fertilidade/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Sêmen/metabolismo , Espermatogênese/genética , Testículo/metabolismo , Proteínas Argonautas/genética
11.
J Biol Chem ; 298(9): 102327, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35931115

RESUMO

Nonobstructive azoospermia (NOA) is the most serious form of spermatogenesis abnormalities in male infertility. Genetic factors are important to consider as elements leading to NOA. Although many pathogenic genes have been reported, the causative genes of NOA for many patients are still unknown. In this study, we found ten point mutations in the gene encoding homeodomain-interacting protein kinase 4 (HIPK4) in patients with NOA, and using in vitro studies, we determined a premature termination point mutation (p. Lys490∗, c.1468A>T) that can cause decreased expression of HIPK4. Our phosphoproteomic analysis of Hipk4-/- testes revealed phosphorylation of multiple proteins regulated by HIPK4 during spermiogenesis. We also confirmed that a substrate of HIPK4 with four downregulated phosphorylation sites matching the xSPx motif is the known manchette-related protein RIMS-binding protein 3, which is required for sperm head morphogenesis. Therefore, we conclude HIPK4 regulates the phosphorylation of manchette protein RIMS-binding protein 3 and plays essential roles in sperm head shaping and male fertility.


Assuntos
Azoospermia , Códon sem Sentido , Proteínas do Citoesqueleto , Proteínas Serina-Treonina Quinases , Cabeça do Espermatozoide , Espermatogênese , Azoospermia/genética , Azoospermia/metabolismo , Proteínas do Citoesqueleto/metabolismo , Humanos , Masculino , Fosforilação , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Cabeça do Espermatozoide/metabolismo , Espermatogênese/genética , Testículo/metabolismo
12.
Cell Mol Life Sci ; 79(8): 467, 2022 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-35930080

RESUMO

Meiosis, a highly conserved process in organisms from fungi to mammals, is subjected to protein phosphorylation regulation. Due to the low abundance of phosphorylation, there is a lack of systemic characterization of phosphorylation regulation of meiosis in mammals. Using the phosphoproteomic approach, we profiled large-scale phosphoproteome of purified primary spermatocytes undergoing meiosis I, and identified 14,660 phosphorylation sites in 4419 phosphoproteins. Kinase-substrate phosphorylation network analysis followed by in vitro meiosis study showed that CDK9 was essential for meiosis progression to metaphase I and had enriched substrate phosphorylation sites in proteins involved in meiotic cell cycle. In addition, histones and epigenetic factors were found to be widely phosphorylated. Among those, HASPIN was found to be essential for male fertility. Haspin knockout led to misalignment of chromosomes, apoptosis of metaphase spermatocytes and a decreased number of sperm by deregulation of H3T3ph, chromosomal passenger complex (CPC) and spindle assembly checkpoint (SAC). The complicated protein phosphorylation and its important regulatory functions in meiosis indicated that in-depth studies of phosphorylation-mediated signaling could help us elucidate the mechanisms of meiosis.


Assuntos
Meiose , Sêmen , Animais , Histonas/metabolismo , Masculino , Mamíferos/metabolismo , Metáfase , Camundongos , Fosforilação , Sêmen/metabolismo , Espermatócitos
13.
Mol Biol Rep ; 49(6): 4469-4478, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35220512

RESUMO

BACKGROUND: Spermatogonial stem cells (SSCs) are unique stem cells that account for the whole reproductive life of males and transmit genetic information to offspring. SSC maintenance is intricate and the underlying mechanisms are largely unclear. Here, we report that SSC maintenance is driven by the plasminogen receptor (PLGRKT). METHODS AND RESULTS: PLGRKT was located in SSCs, and knockdown of PLGRKT expression in cultured neonatal testis and SSCs impaired the proliferation and promoted the apoptosis of cells. PLGRKT interacted with B lymphoma Mo-MLV insertion region 1 (BMI1), and modulated oxidative stress and p16/p19 signaling in SSCs. CONCLUSIONS: We demonstrated that reactive oxygen species (ROS) and p16/p19 signaling are involved in "PLGRKT-BMI1" co-regulation of SSC maintenance in mice.


Assuntos
Plasminogênio , Testículo , Animais , Células Cultivadas , Masculino , Camundongos , Estresse Oxidativo/genética , Plasminogênio/metabolismo , Complexo Repressor Polycomb 1/genética , Complexo Repressor Polycomb 1/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Células-Tronco/metabolismo , Testículo/metabolismo
14.
Proteomics ; 21(15): e2100025, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34050602

RESUMO

A growing body of evidence now supports the fact that protein ubiquitination is an important modification during the regulation of spermatogenesis. However, little is known about the ubiquitome of the testis. In this study, we created a large-scale mouse testis ubiquitome profile using di-glycine remnant antibodies and mass spectrometry and identified a total of 14,219 ubiquitination sites in 4217 proteins. Bioinformatics and phenotypic analyses showed that the ubiquitinated proteins were closely related to meiosis and spermiogenesis. And 512 ubiquitination regulatory enzymes were identified in testis that can exert regulatory functions over ubiquitination: the homologous to E6AP C-terminus (HECT) and multi-subunit RING-finger type E3 ligases were significantly enriched. In addition, we identified 22 new ubiquitination sites on testicular histones and 146 ubiquitinated epigenetic factors, thus demonstrating that ubiquitination plays an important role in epigenetic regulation. Collectively, this in-depth characterization of the ubiquitome in mouse testis could provide a rich resource for further studies of regulatory events at the protein level during spermatogenesis. All MS data are available via ProteomeXchange with the identifier PXD025866.


Assuntos
Epigênese Genética , Testículo , Animais , Masculino , Camundongos , Espermatogênese , Testículo/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Ubiquitinadas/metabolismo , Ubiquitinação
15.
Circulation ; 141(19): 1554-1569, 2020 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-32098494

RESUMO

BACKGROUND: In mammals, regenerative therapy after myocardial infarction is hampered by the limited regenerative capacity of adult heart, whereas a transient regenerative capacity is maintained in the neonatal heart. Systemic phosphorylation signaling analysis on ischemic neonatal myocardium might be helpful to identify key pathways involved in heart regeneration. Our aim was to define the kinase-substrate network in ischemic neonatal myocardium and to identify key pathways involved in heart regeneration after ischemic insult. METHODS: Quantitative phosphoproteomics profiling was performed on infarct border zone of neonatal myocardium, and kinase-substrate network analysis revealed 11 kinases with enriched substrates and upregulated phosphorylation levels, including checkpoint kinase 1 (CHK1) kinase. The effect of CHK1 on cardiac regeneration was tested on Institute of Cancer Research CD1 neonatal and adult mice that underwent apical resection or myocardial infarction. RESULTS: In vitro, CHK1 overexpression promoted whereas CHK1 knockdown blunted cardiomyocyte proliferation. In vivo, inhibition of CHK1 hindered myocardial regeneration on resection border zone in neonatal mice. In adult myocardial infarction mice, CHK1 overexpression on infarct border zone upregulated mammalian target of rapamycin C1/ribosomal protein S6 kinase b-1 pathway, promoted cardiomyocyte proliferation, and improved cardiac function. Inhibiting mammalian target of rapamycin activity by rapamycin blunted the neonatal cardiomyocyte proliferation induced by CHK1 overexpression in vitro. CONCLUSIONS: Our study indicates that phosphoproteome of neonatal regenerative myocardium could help identify important signaling pathways involved in myocardial regeneration. CHK1 is found to be a key signaling responsible for neonatal regeneration. Myocardial overexpression of CHK1 could improve cardiac regeneration in adult hearts by activating the mammalian target of rapamycin C1/ribosomal protein S6 kinase b-1 pathway. Thus, CHK1 might serve as a potential novel target in myocardial repair after myocardial infarction.


Assuntos
Proliferação de Células , Quinase 1 do Ponto de Checagem/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Infarto do Miocárdio/enzimologia , Miocárdio/enzimologia , Proteoma , Regeneração , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Fatores Etários , Animais , Animais Recém-Nascidos , Células Cultivadas , Quinase 1 do Ponto de Checagem/genética , Modelos Animais de Doenças , Camundongos Endogâmicos ICR , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miocárdio/metabolismo , Fosforilação , Transdução de Sinais
16.
Development ; 145(13)2018 07 04.
Artigo em Inglês | MEDLINE | ID: mdl-29866902

RESUMO

Transcription factors of the Sox protein family contain a DNA-binding HMG box and are key regulators of progenitor cell fate. Here, we report that expression of Sox30 is restricted to meiotic spermatocytes and postmeiotic haploids. Sox30 mutant males are sterile owing to spermiogenic arrest at the early round spermatid stage. Specifically, in the absence of Sox30, proacrosomic vesicles fail to form a single acrosomal organelle, and spermatids arrest at step 2-3. Although most Sox30 mutant spermatocytes progress through meiosis, accumulation of diplotene spermatocytes indicates a delayed or impaired transition from meiotic to postmeiotic stages. Transcriptome analysis of isolated stage-specific spermatogenic cells reveals that Sox30 controls a core postmeiotic gene expression program that initiates as early as the late meiotic cell stage. ChIP-seq analysis shows that Sox30 binds to specific DNA sequences in mouse testes, and its genomic occupancy correlates positively with expression of many postmeiotic genes including Tnp1, Hils1, Ccdc54 and Tsks These results define Sox30 as a crucial transcription factor that controls the transition from a late meiotic to a postmeiotic gene expression program and subsequent round spermatid development.


Assuntos
Regulação da Expressão Gênica/fisiologia , Meiose/fisiologia , Fatores de Transcrição SOX/metabolismo , Espermátides/metabolismo , Espermatogênese/fisiologia , Testículo/metabolismo , Iniciação da Transcrição Genética/fisiologia , Animais , Perfilação da Expressão Gênica , Masculino , Camundongos , Elementos de Resposta/fisiologia , Fatores de Transcrição SOX/genética , Espermátides/citologia , Testículo/citologia
17.
Eur J Clin Invest ; 51(1): e13443, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33131070

RESUMO

BACKGROUND: To reveal detailed histopathological changes, virus distributions, immunologic properties and multi-omic features caused by SARS-CoV-2 in the explanted lungs from the world's first successful lung transplantation of a COVID-19 patient. MATERIALS AND METHODS: A total of 36 samples were collected from the lungs. Histopathological features and virus distribution were observed by optical microscope and transmission electron microscope (TEM). Immune cells were detected by flow cytometry and immunohistochemistry. Transcriptome and proteome approaches were used to investigate main biological processes involved in COVID-19-associated pulmonary fibrosis. RESULTS: The histopathological changes of the lung tissues were characterized by extensive pulmonary interstitial fibrosis and haemorrhage. Viral particles were observed in the cytoplasm of macrophages. CD3+ CD4- T cells, neutrophils, NK cells, γ/δ T cells and monocytes, but not B cells, were abundant in the lungs. Higher levels of proinflammatory cytokines iNOS, IL-1ß and IL-6 were in the area of mild fibrosis. Multi-omics analyses revealed a total of 126 out of 20,356 significant different transcription and 114 out of 8,493 protein expression in lung samples with mild and severe fibrosis, most of which were related to fibrosis and inflammation. CONCLUSIONS: Our results provide novel insight that the significant neutrophil/ CD3+ CD4- T cell/ macrophage activation leads to cytokine storm and severe fibrosis in the lungs of COVID-19 patient and may contribute to a better understanding of COVID-19 pathogenesis.


Assuntos
COVID-19/patologia , Hemorragia/patologia , Transplante de Pulmão , Pulmão/patologia , Linfonodos/patologia , Fibrose Pulmonar/patologia , Linfócitos B/patologia , Linfócitos B/ultraestrutura , Linfócitos B/virologia , COVID-19/genética , COVID-19/metabolismo , COVID-19/cirurgia , Cromatografia Líquida , Citometria de Fluxo , Perfilação da Expressão Gênica , Humanos , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Células Matadoras Naturais/patologia , Células Matadoras Naturais/ultraestrutura , Células Matadoras Naturais/virologia , Pulmão/metabolismo , Pulmão/ultraestrutura , Pulmão/virologia , Linfonodos/metabolismo , Linfonodos/ultraestrutura , Linfonodos/virologia , Macrófagos Alveolares/patologia , Macrófagos Alveolares/ultraestrutura , Macrófagos Alveolares/virologia , Masculino , Pessoa de Meia-Idade , Monócitos/patologia , Monócitos/ultraestrutura , Monócitos/virologia , Neutrófilos/patologia , Neutrófilos/ultraestrutura , Neutrófilos/virologia , Óxido Nítrico Sintase Tipo II/metabolismo , Proteômica , Fibrose Pulmonar/genética , Fibrose Pulmonar/metabolismo , Fibrose Pulmonar/cirurgia , RNA-Seq , SARS-CoV-2 , Índice de Gravidade de Doença , Linfócitos T/patologia , Linfócitos T/ultraestrutura , Linfócitos T/virologia , Espectrometria de Massas em Tandem
18.
Nucleic Acids Res ; 47(22): 11755-11770, 2019 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-31724724

RESUMO

During meiosis, telomere attachment to the inner nuclear envelope is required for proper pairing of homologous chromosomes and recombination. Here, we identified F-box protein 47 (FBXO47) as a regulator of the telomeric shelterin complex that is specifically expressed during meiotic prophase I. Knockout of Fbxo47 in mice leads to infertility in males. We found that the Fbxo47 deficient spermatocytes are unable to form a complete synaptonemal complex. FBXO47 interacts with TRF1/2, and the disruption of Fbxo47 destabilizes TRF2, leading to unstable telomere attachment and slow traversing through the bouquet stage. Our findings uncover a novel mechanism of FBXO47 in telomeric shelterin subunit stabilization during meiosis.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Proteínas F-Box/fisiologia , Meiose/fisiologia , Membrana Nuclear/metabolismo , Telômero/metabolismo , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo , Fatores de Transcrição/fisiologia , Animais , Feminino , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Knockout , Membrana Nuclear/genética , Estabilidade Proteica , Espermatócitos/fisiologia
19.
Proc Natl Acad Sci U S A ; 115(23): E5326-E5333, 2018 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-29784807

RESUMO

MTOR (mechanistic target of rapamycin) is a widely recognized integrator of signals and pathways key for cellular metabolism, proliferation, and differentiation. Here we show that conditional knockout (cKO) of Mtor in either primordial or growing oocytes caused infertility but differentially affected oocyte quality, granulosa cell fate, and follicular development. cKO of Mtor in nongrowing primordial oocytes caused defective follicular development leading to progressive degeneration of oocytes and loss of granulosa cell identity coincident with the acquisition of immature Sertoli cell-like characteristics. Although Mtor was deleted at the primordial oocyte stage, DNA damage accumulated in oocytes during their later growth, and there was a marked alteration of the transcriptome in the few oocytes that achieved the fully grown stage. Although oocyte quality and fertility were also compromised when Mtor was deleted after oocytes had begun to grow, these occurred without overtly affecting folliculogenesis or the oocyte transcriptome. Nevertheless, there was a significant change in a cohort of proteins in mature oocytes. In particular, down-regulation of PRC1 (protein regulator of cytokinesis 1) impaired completion of the first meiotic division. Therefore, MTOR-dependent pathways in primordial or growing oocytes differentially affected downstream processes including follicular development, sex-specific identity of early granulosa cells, maintenance of oocyte genome integrity, oocyte gene expression, meiosis, and preimplantation developmental competence.


Assuntos
Células da Granulosa/citologia , Oócitos/citologia , Serina-Treonina Quinases TOR/metabolismo , Animais , Diferenciação Celular/fisiologia , Células Cultivadas , Feminino , Hormônio Foliculoestimulante/sangue , Células da Granulosa/enzimologia , Células da Granulosa/metabolismo , Infertilidade Feminina/genética , Infertilidade Feminina/metabolismo , Infertilidade Feminina/patologia , Meiose/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oócitos/enzimologia , Oócitos/metabolismo , Oogênese , Folículo Ovariano/citologia , Folículo Ovariano/enzimologia , Folículo Ovariano/metabolismo , Serina-Treonina Quinases TOR/genética
20.
J Biol Chem ; 294(25): 9959-9972, 2019 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-31092598

RESUMO

Mesoderm development is a finely tuned process initiated by the differentiation of pluripotent epiblast cells. Serine/threonine kinase 40 (STK40) controls the development of several mesoderm-derived cell types, its overexpression induces differentiation of mouse embryonic stem cells (mESCs) toward the extraembryonic endoderm, and Stk40 knockout (KO) results in multiple organ failure and is lethal at the perinatal stage in mice. However, molecular mechanisms underlying the physiological functions of STK40 in mesoderm differentiation remain elusive. Here, we report that Stk40 ablation impairs mesoderm differentiation both in vitro and in vivo Mechanistically, STK40 interacts with both the E3 ubiquitin ligase mammalian constitutive photomorphogenesis protein 1 (COP1) and the transcriptional regulator proto-oncogene c-Jun (c-JUN), promoting c-JUN protein degradation. Consequently, Stk40 knockout leads to c-JUN protein accumulation, which, in turn, apparently suppresses WNT signaling activity and impairs the mesoderm differentiation process. Overall, this study reveals that STK40, together with COP1, represents a previously unknown regulatory axis that modulates the c-JUN protein level within an appropriate range during mesoderm differentiation from mESCs. Our findings provide critical insights into the molecular mechanisms regulating the c-JUN protein level and may have potential implications for managing cellular disorders arising from c-JUN dysfunction.


Assuntos
Diferenciação Celular , Mesoderma/citologia , Proteínas Nucleares/metabolismo , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Proto-Oncogênicas c-jun/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Proteína Wnt1/metabolismo , Animais , Células Cultivadas , Mesoderma/metabolismo , Camundongos , Camundongos Knockout , Proteínas Nucleares/genética , Proteínas Proto-Oncogênicas c-jun/genética , Ubiquitina-Proteína Ligases/genética , Proteína Wnt1/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA