Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
Mais filtros

País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Cancer Sci ; 115(2): 465-476, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37991109

RESUMO

NR0B1 is frequently activated in hepatocellular carcinoma (HCC). However, the role of NR0B1 is controversial in HCC. In this study, we observed that NR0B1 was an independent poor prognostic factor, negatively correlated with the overall survival of HCC and the relapse-free survival of patients treated with sorafenib. Meanwhile, NR0B1 promoted the proliferation, migration, and invasion of HCC cells, inhibited sorafenib-induced apoptosis, and elevated the IC50 of sorafenib in HCC cells. NR0B1 was further displayed to increase sorafenib-induced autophagic vesicles and activate Beclin1/LC3-II-dependent autophagy pathway. Finally, NR0B1 was revealed to transcriptionally suppress GSK3ß that restrains AMPK/mTOR-driven autophagy and increases BAX-mediated apoptosis. Collectively, our study uncovered that the ectopic expression of NR0B1 augmented sorafenib-resistance in HCC cells by activating autophagy and inhibiting apoptosis. Our findings supported that NR0B1 was a detrimental factor for HCC prognosis.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Sorafenibe/farmacologia , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Resistencia a Medicamentos Antineoplásicos , Recidiva Local de Neoplasia , Apoptose , Autofagia , Proliferação de Células , Linhagem Celular Tumoral , Receptor Nuclear Órfão DAX-1
2.
Ann Hum Genet ; 88(4): 300-306, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38192234

RESUMO

BACKGROUND: BMPR1A-mediated signaling transduction plays an essential role in intestinal growth. Variations of BMPR1A lead to a rare autosomal dominant inherited juvenile polyposis syndrome (JPS) with high probability of developing into colorectal cancer (CRC). Nonsense and frameshift variations, generating premature termination codons (PTCs), are the most pathogenic variants in the BMPR1A gene. OBJECTIVE: This study aimed to investigate the molecular genetic etiology in a Chinese family with three generations of CRC. METHODS: Pathogenic variants of 18 known CRC susceptibility genes were examined in a Chinese CRC family through multigene panel testing using the next-generation sequencing platform. The candidate gene variant was validated in the family members by Sanger sequencing. Potential biological functions of the gene variant were further investigated in the RKO colon cancer cell line. RESULTS: A novel nonsense variant (c.1114A > T, p.Lys372*) of BMPR1A was identified in the CRC family. This variant generated a PTC at the kinase domain and caused nonsense-mediated mRNA decay. Read-through inducing reagents G418 and PTC124 partially restored BMPR1A expression and its following signaling pathway. CONCLUSION: The identification of the novel BMPR1A variant enriched the genotype-phenotype spectrum of BMPR1A. Meanwhile, our finding also provided support for future PTC-targeting therapy for BMPR1A-mediated JPS and CRC.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas Tipo I , Códon sem Sentido , Linhagem , Adulto , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Povo Asiático/genética , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Linhagem Celular Tumoral , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , População do Leste Asiático , Família , Predisposição Genética para Doença , Degradação do RNAm Mediada por Códon sem Sentido
3.
Ann Hum Genet ; 87(5): 241-247, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37461830

RESUMO

BACKGROUND: The protein encoded by the cartilage oligomeric matrix protein (COMP) gene is a noncollagenous extracellular matrix (ECM) protein that is important for chondrocyte formation and growth. Variations in the COMP gene cause pseudoachondroplasia (PSACH), which is mainly characterized by short-limbed dwarfing in the clinic. AIMS: To characterize the function of a rare pathogenic variant in the COMP gene (c.875G > A, p.Cys292Tyr). MATERIALS & METHODS: We performed 3D structural analysis, in vitro expression analysis, and immunofluorescence to characterize the effects of the variant on protein structure, expression, and cellular localization respectively. RESULTS: Variation modeling showed that the interactions between amino acids were changed after the variation, and there were 31 changes in the secondary structure of mutant COMP (MT-COMP). Western blot showed that the intracellular quantity of MT-COMP was higher than the wild-type COMP (WT-COMP). Cellular immunofluorescence results showed that WT-COMP was less abundant and homogenously distributed in cells, while the MT-COMP accumulated in the cytoplasm. DISCUSSION: Herein, we report a variant of COMP in a Chinese family with PSACH. We have shown that the rare missense variant, COMP c.875G > A, previously reported in ClinVar and identified in our patient, results in excessive accumulation of mutant protein in the cytoplasm, and is therefore pathogenic. CONCLUSION: Through in silico and experimental analyses, we provide evidence that COMP c.875G > A is the likely cause of PSACH in a Chinese family.


Assuntos
Acondroplasia , Humanos , Acondroplasia/genética , Acondroplasia/metabolismo , Acondroplasia/patologia , Proteína de Matriz Oligomérica de Cartilagem/genética , Proteína de Matriz Oligomérica de Cartilagem/metabolismo , Condrócitos/metabolismo , Condrócitos/patologia , Mutação
4.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 40(9): 1160-1164, 2023 Sep 10.
Artigo em Zh | MEDLINE | ID: mdl-37643966

RESUMO

OBJECTIVE: To explore the genetic basis for a patient with early-onset retinitis pigmentosa (RP). METHODS: A patient who had presented at the West China Hospital of Sichuan University on March 10, 2020 was selected as the study subject. The patient and his parents were subjected to whole exome sequencing (WES). Candidate variants were verified by Sanger sequencing and in silico analysis. RESULTS: The patient has featured substantial loss of binocular vision field. Funduscopy revealed characteristic bone spicule-type pigment deposits, as well as attenuated retinal arterioles and pale-appearing optic discs. WES revealed that he has harbored compound missense variants of a RP-associated CRB1 gene, including c.2969T>C (p.Leu990Ser) and c.1816T>C (p.Cys606Arg), which were respectively inherited from his father and mother. Homozygous c.1816T>C (p.Cys606Arg) variant has been identified among RP patients, whilst the c.2969T>C (p.Leu990Ser) variant was unreported previously. Both variants were predicted as likely pathogenic based on the guidelines from the American College of Medical Genetics and Genomics (ACMG). CONCLUSION: The novel compound heterozygous variants of the CRB1 gene probably underlay the early-onset RP in this patient. Above finding has enriched the mutational spectrum of the CRB1 gene.


Assuntos
Genômica , Retinose Pigmentar , Masculino , Feminino , Humanos , China , Homozigoto , Mães , Retinose Pigmentar/genética , Proteínas do Olho/genética , Proteínas de Membrana/genética , Proteínas do Tecido Nervoso/genética
5.
J Hum Genet ; 67(1): 27-34, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34257392

RESUMO

The development of sequencing techniques identified numerous genetic variants, and accurate evaluation of the clinical significance of these variants facilitates the diagnosis of Mendelian diseases. In the present study, 549 rare single- nucleotide variants of uncertain significance were extracted from the ADPKD and ClinVar databases. MaxEntScan scoresplice is an in silico splicing prediction tool that was used to analyze rare PKD1 and PKD2 variants of unknown significance. An in vitro minigene splicing assay was used to verify 37 splicing-altering candidates that were located within seven residues of the splice donor sequence excluding canonical GT dinucleotides or within 21 residues of the acceptor sequence excluding canonical AG dinucleotides of PKD1 and PKD2. We demonstrated that eight PKD1 variants alter RNA splicing and were predicted to be pathogenic.


Assuntos
Mutação Puntual , Splicing de RNA , Canais de Cátion TRPP/genética , Análise Mutacional de DNA , Estudos de Associação Genética/métodos , Predisposição Genética para Doença , Humanos , Fenótipo
6.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 39(5): 522-525, 2022 May 10.
Artigo em Zh | MEDLINE | ID: mdl-35598270

RESUMO

OBJECTIVE: To identify the pathogenic variants from a patient with suspected congenital contractural arachnodactyly, and to explore the possible molecular genetic pathogenesis, so as to provide evidence for clinical diagnosis. METHODS: Whole exome sequencing was performed for the patient. The splicing site variation of candidate pathogenic genes was verified by Sanger sequencing, and the new transcript sequence was determined by RT-PCR and TA-cloning sequencing. RESULTS: The patient carried a heterozygous c.533-1G>C variant of FBN2 gene, which was not reported. The sequencing of mRNA showed that the variant leaded to the disappearance of the canonical splice acceptor site of FBN2 gene and the activation of a cryptic splice acceptor site at c.533-71, resulting in the insertion of 70 bp sequence in the new transcript. It was speculated that the polypeptide encoded by the new transcript changed from valine (Val) to serine (Ser) at amino acid 179, and prematurely terminated after 26 aminoacids. According to the guidelines of American College of Medical Genetics and Genomics, the variant of FBN2 gene c. 533-1G>C was determined as pathogenic (PVS1+PM2+PP3 ). CONCLUSION: A novel splicing variant of FBN2 gene (c.533-1G>C) was identified, which can lead to congenital contractural arachnodactyly.


Assuntos
Aracnodactilia , Contratura , Aracnodactilia/genética , Contratura/genética , Fibrilina-2/genética , Humanos , Mutação , Sítios de Splice de RNA , Sequenciamento do Exoma
7.
Cancer Sci ; 112(2): 691-702, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33185915

RESUMO

Testis-specific protein Y-encoded 1 (TSPY1), a Y chromosome-linked oncogene, is frequently activated in prostate cancers (PCa) and its expression is correlated with the poor prognosis of PCa. However, the cause of the ectopic transcription of TSPY1 in PCa remains unclear. Here, we observed that the methylation status in the CpG islands (CGI) of the TSPY1 promoter was negatively correlated with its expression level in different human samples. The acetyl-histone H4 and trimethylated histone H3-lysine 4, two post-translational modifications of histones occupying the TSPY1 promoter, facilitated the TSPY1 expression in PCa cells. In addition, we found that androgen accelerated the TSPY1 transcription on the condition of hypomethylated of TSPY1-CGI and promoted PCa cell proliferation. Moreover, the binding of androgen receptor (AR) to the TSPY1 promoter, enhancing TSPY1 transcription, was detected in PCa cells. Taken together, our findings identified the regulation of DNA methylation, acting as a primary mechanism, on TSPY1 expression in PCa, and revealed that TSPY1 is an androgen-AR axis-regulated oncogene, suggesting a novel and potential target for PCa therapy.


Assuntos
Proteínas de Ciclo Celular/biossíntese , Metilação de DNA/genética , Regulação Neoplásica da Expressão Gênica/genética , Neoplasias da Próstata/genética , Receptores Androgênicos/metabolismo , Acetilação , Proliferação de Células/genética , Ilhas de CpG/genética , Histonas/metabolismo , Humanos , Masculino , Regiões Promotoras Genéticas/genética , Neoplasias da Próstata/patologia , Processamento de Proteína Pós-Traducional/genética
8.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 38(12): 1241-1244, 2021 Dec 10.
Artigo em Zh | MEDLINE | ID: mdl-34839516

RESUMO

OBJECTIVE: To provide genetic counseling for a couple with recurrent detection of fetal structural abnormality during second trimester pregnancy. METHODS: The fetal tissue and peripheral blood samples of the couple were subjected to G banded chromosomal analysis, copy number variation sequencing (CNV-seq) and fluorescence in situ hybridization (FISH) assays. RESULTS: CNV-seq has detected a 6.59 Mb duplication at 7p22.3-p22.1 and a 3.81 Mb deletion at 4p16.3 in the fetal tissue, though conventional karyotyping results of both parents were normal. FISH has confirmed that the father has harbored a cryptic translocation of t(4;7)(7p+,4q+,4p+,7q+). CONCLUSION: The ultrasonographic abnormality of the fetuses may be attributed to the 7p microduplication and 4p microdeletion derived from the cryptic translocation carried by the father. Reciprocal translocation of tiny chromosomal segments should be suspected for couples with recurrent adverse pregnancies but apparently normal karyotypes.


Assuntos
Transtornos Cromossômicos , Variações do Número de Cópias de DNA , Feminino , Humanos , Hibridização in Situ Fluorescente , Cariotipagem , Gravidez , Translocação Genética
9.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 38(7): 643-646, 2021 Jul 10.
Artigo em Zh | MEDLINE | ID: mdl-34247368

RESUMO

OBJECTIVE: To explore the genetic basis for a couple with recurrent conceptions of fetus with abnormal longbones, and another couple with a history of omphalocele. METHODS: Genomic DNA was extracted from the peripheral blood samples from both couples. All exons and flanking regions were analyzed with next generation sequencing. Candidate variants were verified by Sanger sequencing. RESULTS: Couple one was found to be heterozygous for, a c.997+1G>T splice-site variant and a missence c.871G>A(p.Glu291Lys) variant of the ALPL gene. Both variants were predicted to be pathogenic and may result in reduced function or loss of alkaline phosphatase. For couple two, the wife was found to harbor a novel c.637_652 delins CCC variant of the CDKN1C gene. This deletion-insertion variant resulted in frame-shift and loss of function (p.Ala213Profs*55) of the CDKN1C protein. Maternally inherited CDKN1C LOF variant has been found to underlie Beckwith-Wiedemann syndrome (BWS), which may manifest as omphalocele. CONCLUSION: Dispite the lack the direct proof from the lost fetuses, the variants of ALPL and CDKN1C genes can explain the recurrence of fetal malformations for both couples.


Assuntos
Síndrome de Beckwith-Wiedemann , Feto , Humanos , Mutação
10.
J Cell Mol Med ; 23(7): 4689-4698, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31099187

RESUMO

Circadian rhythms are maintained by series of circadian clock proteins, and post-translation modifications of clock proteins significantly contribute to regulating circadian clock. However, the underlying upstream mechanism of circadian genes that are responsible for circadian rhythms in cancer cells remains unknown. PIWIL1 participates in many physiological processes and current discoveries have shown that PIWIL1 is involved in tumorigenesis in various cancers. Here we report that PIWIL1 can suppress circadian rhythms in cancer cells. Mechanistically, by promoting SRC interacting with PI3K, PIWIL1 can activate PI3K-AKT signalling pathway to phosphorylate and inactivate GSK3ß, repressing GSK3ß-induced phosphorylation and ubiquitination of CLOCK and BMAL1. Simultaneously, together with CLOCK/BMAL1 complex, PIWIL1 can bind with E-BOX region to suppress transcriptional activities of clock-controlled genes promoters. Collectively, our findings first demonstrate that PIWIL1 negatively regulates circadian rhythms via two pathways, providing molecular connection between dysfunction of circadian rhythms and tumorigenesis.


Assuntos
Fatores de Transcrição ARNTL/metabolismo , Proteínas Argonautas/metabolismo , Proteínas CLOCK/metabolismo , Ritmo Circadiano , Glicogênio Sintase Quinase 3 beta/metabolismo , Proteólise , Proteínas Argonautas/química , Linhagem Celular Tumoral , Elementos E-Box/genética , Humanos , Modelos Biológicos , Fosforilação , Ligação Proteica , Domínios Proteicos , Ativação Transcricional/genética , Ubiquitinação , Quinases da Família src/metabolismo
11.
Cancer Sci ; 110(5): 1573-1586, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30815935

RESUMO

The testis-specific protein, Y-linked 1 (TSPY1), a newly recognized cancer/testis antigen, has been suggested to accelerate tumor progression. However, the mechanisms underlying TSPY1 cancer-related function remain limited. By mining the RNA sequencing data of lung and liver tumors from The Cancer Genome Atlas, we found frequent ectopic expression of TSPY1 in lung adenocarcinoma (LUAD) and liver hepatocellular carcinoma (LIHC), and the male-specific protein was associated with higher mortality rate and worse overall survival in patients with LUAD and LIHC. Overexpression of TSPY1 promotes cell proliferation, invasiveness, and cycle transition and inhibits apoptosis, whereas TSPY1 knockdown has the opposite effects on these cancer cell phenotypes. Transcriptomic analysis revealed the involvement of TSPY1 in PI3K/AKT and RAS signaling pathways in both LUAD and LIHC cells, which was further confirmed by the increase in the levels of phosphorylated proteins in the PI3K-AKT and RAS signaling pathways in TSPY1-overexpressing cancer cells, and by the suppression on the activity of these two pathways in TSPY1-knockdown cells. Further investigation identified that TSPY1 could directly bind to the promoter of insulin growth factor binding protein 3 (IGFBP3) to inhibit IGFBP3 expression and that downregulation of IGFBP3 increased the activity of PI3K/AKT/mTOR/BCL2 and RAS/RAF/MEK/ERK/JUN signaling in LUAD and LIHC cells. Taken together, the observations reveal a novel mechanism by which TSPY1 could contribute to the progression of LUAD and LIHC. Our finding is of importance for evaluating the potential of TSPY1 in immunotherapy of male tumor patients with TSPY1 expression.


Assuntos
Carcinoma Hepatocelular/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Neoplasias Hepáticas/genética , Neoplasias Pulmonares/genética , Transdução de Sinais , Células A549 , Carcinoma Hepatocelular/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Progressão da Doença , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Células Hep G2 , Humanos , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Neoplasias Hepáticas/metabolismo , Neoplasias Pulmonares/metabolismo , Masculino , Invasividade Neoplásica , Fosfatidilinositol 3-Quinases/metabolismo , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-akt/metabolismo , Análise de Sequência de RNA , Análise de Sobrevida , Proteínas ras/metabolismo
12.
Biochem Biophys Res Commun ; 516(3): 819-824, 2019 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-31262447

RESUMO

PIWIL2 belongs to the PIWI protein subfamily and is widely expressed in a variety of tumors. Previous studies have shown that PIWIL2 has the characteristics of oncogene. Recently we reported that PIWIL2 suppresses GSK3ß activity to regulate circadian rhythms through SRC-PI3K-AKT pathway. As GSK3ß is a key part of the ß-catenin destruction complex, it plays a vital role in regulating the degradation of ß-catenin. Besides, the activated ß-catenin/CyclinD1 pathway is involved in the proliferation of tumor cells. It is intriguing to investigate whether PIWIL2 regulates ß-catenin and downstream pathway. In this study, we found that PIWIL2 suppressed GSK3ß induced phosphorylation and ubiquitination of ß-catenin, and thus increased ß-catenin accumulation in the nucleus. By up-regulating ß-catenin and CyclinD1, PIWIL2 can promote cell cycle and proliferation in tumor cells. Taken together, our results revealed a novel function of PIWIL2 in regulating ß-catenin/CyclinD1 pathway in tumor cells, providing a new perspective for PIWIL2 as an oncogene.


Assuntos
Proteínas Argonautas/genética , Ciclo Celular/genética , Ciclina D1/genética , Regulação Neoplásica da Expressão Gênica , Processamento de Proteína Pós-Traducional , beta Catenina/genética , Proteínas Argonautas/metabolismo , Núcleo Celular/genética , Núcleo Celular/metabolismo , Proliferação de Células , Ciclina D1/metabolismo , Glicogênio Sintase Quinase 3 beta/genética , Glicogênio Sintase Quinase 3 beta/metabolismo , Células HeLa , Células Hep G2 , Humanos , Fosforilação , Estabilidade Proteica , Transdução de Sinais , Ubiquitinação , beta Catenina/metabolismo
13.
Biol Reprod ; 100(4): 994-1007, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30541061

RESUMO

Some X-linked genes necessary for spermiogenesis are specifically activated in the postmeiotic germ cells. However, the regulatory mechanism about this activation is not clearly understood. Here, we examined the potential mechanism controlling the transcriptional activation of the mouse testis specific gene A8 (Tsga8) gene in round spermatids. We observed that the Tsga8 expression was negatively correlated with the methylation level of the CpG sites in its core promoter. During spermatogenesis, the Tsga8 promoter was methylated in spermatogonia, and then demethylated in spermatocytes. The demethylation status of Tsga8 promoter was maintained through the postmeiotic germ cells, providing a potentially active chromatin for Tsga8 transcription. In vitro investigation showed that the E12 and Spz1 transcription factors can enhance the Tsga8 promoter activity by binding to the unmethylated E-box motif within the Tsga8 promoter. Additionally, the core Tsga8 promoter drove green fluorescent protein (GFP) expression in the germ cells of Tsga8-GFP transgenic mice, and the GFP expression pattern was similar to that of endogenous Tsga8. Moreover, the DNA methylation profile of the Tsga8-promoter-driven transgene was consistent with that of the endogenous Tsga8 promoter, indicating the existence of a similar epigenetic modification for the Tsga8 promoter to ensure its spatiotemporal expression in vivo. Taken together, this study reports the details of a regulatory mechanism that includes DNA methylation and transcription factors to mediate the postmeiotic expression of an X-linked gene.


Assuntos
Desmetilação do DNA , Nucleoproteínas/genética , Espermátides/metabolismo , Ativação Transcricional , Animais , Células Cultivadas , Epigênese Genética/fisiologia , Feminino , Genes Ligados ao Cromossomo X/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células NIH 3T3 , Nucleoproteínas/metabolismo , Regiões Promotoras Genéticas , Espermatogênese/genética , Fatores de Transcrição/fisiologia , Ativação Transcricional/genética
14.
J Biol Chem ; 292(34): 14165-14175, 2017 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-28687631

RESUMO

Circadian clock and Smad2/3/4-mediated Nodal signaling regulate multiple physiological and pathological processes. However, it remains unknown whether Clock directly cross-talks with Nodal signaling and how this would regulate embryonic development. Here we show that Clock1a coordinated mesoderm development and primitive hematopoiesis in zebrafish embryos by directly up-regulating Nodal-Smad3 signaling. We found that Clock1a is expressed both maternally and zygotically throughout early zebrafish development. We also noted that Clock1a alterations produce embryonic defects with shortened body length, lack of the ventral tail fin, or partial defect of the eyes. Clock1a regulates the expression of the mesodermal markers ntl, gsc, and eve1 and of the hematopoietic markers scl, lmo2, and fli1a Biochemical analyses revealed that Clock1a stimulates Nodal signaling by increasing expression of Smad2/3/4. Mechanistically, Clock1a activates the smad3a promoter via its E-box1 element (CAGATG). Taken together, these findings provide mechanistic insight into the role of Clock1a in the regulation of mesoderm development and primitive hematopoiesis via modulation of Nodal-Smad3 signaling and indicate that Smad3a is directly controlled by the circadian clock in zebrafish.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Desenvolvimento Embrionário , Mesoderma/metabolismo , Proteína Nodal/agonistas , Transdução de Sinais , Proteína Smad3/agonistas , Proteínas de Peixe-Zebra/agonistas , Peixe-Zebra , Animais , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/genética , Embrião não Mamífero/anormalidades , Embrião não Mamífero/citologia , Embrião não Mamífero/efeitos dos fármacos , Embrião não Mamífero/metabolismo , Desenvolvimento Embrionário/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento , Células HEK293 , Hematopoese/efeitos dos fármacos , Humanos , Hibridização In Situ , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Mesoderma/anormalidades , Mesoderma/citologia , Mesoderma/efeitos dos fármacos , Microinjeções , Microscopia de Fluorescência , Morfolinos/farmacologia , Mutação , Proteína Nodal/genética , Proteína Nodal/metabolismo , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Elementos de Resposta/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Proteína Smad3/antagonistas & inibidores , Proteína Smad3/genética , Proteína Smad3/metabolismo , Proteínas de Peixe-Zebra/antagonistas & inibidores , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
15.
Hum Mol Genet ; 22(8): 1679-95, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23307928

RESUMO

AZFc deletions cause a significant phenotypic heterogeneity with respect to spermatogenesis; however, the reason for this is poorly understood. Recently, testis-specific protein Y-encoded 1 (TSPY1) copy number variation (CNV) was determined to be a potential genetic modifier of spermatogenesis. We performed a large-scale cohort study to investigate the effect of TSPY1 CNV on spermatogenesis and to elucidate the possible contribution of TSPY1 genetic variation to the phenotypic expression of AZFc deletions. Haplogrouping of the Y-chromosome and quantification of the TSPY1 copy number were performed in 2272 Han Chinese males with different spermatogenic statuses (704 males with the b2/b4 or gr/gr deletion and 1568 non-AZFc-deleted males). Our data revealed that the TSPY1 copy number distributions were significantly different among non-AZFc-deleted males with different spermatogenic phenotypes. Lower sperm production and an elevated risk of spermatogenic failure were observed in males with fewer than 21 TSPY1 copies and in those with more than 55 copies relative to men with 21-35 copies. Similar results were observed in males with the gr/gr deletion. These findings indicate that TSPY1 CNV affects an individual's susceptibility to spermatogenic failure by modulating the efficiency of spermatogenesis and strongly suggest that there is a significant quantity effect of the TSPY1 copy number on the phenotypic expression of the gr/gr deletion. To our knowledge, this CNV is the first independent genetic factor that has been clearly observed to influence the spermatogenic status of gr/gr deletion carriers. A combined genetic analysis of the TSPY1 copy number and the gr/gr deletion could inform the clinical counselling of infertile couples.


Assuntos
Azoospermia/genética , Proteínas de Ciclo Celular/genética , Cromossomos Humanos Y/genética , Variações do Número de Cópias de DNA/genética , Espermatogênese/genética , Adulto , Povo Asiático , China , Deleção Cromossômica , Estudos de Coortes , Haplótipos , Humanos , Masculino , Pessoa de Meia-Idade , Oligospermia/genética , Fenótipo
16.
Biol Reprod ; 92(2): 41, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25505198

RESUMO

Non-obstructive azoospermia (NOA) is a complex, multifactorial disease. Recent genome-wide association studies (GWAS) have identified eight NOA susceptibility loci at genome-wide significance of P < 5.0 × 10(-8) in Han Chinese from southeastern, northern, and central China. To better understand the role of the variants in conferring NOA risk, we selected four GWAS loci (HLA-DRA rs3129878, PRMT6 rs12097821, SOX5 rs10842262, and PEX10 rs2477686) that were reported before 2014 to investigate their association with NOA and their potential effects on sperm production in 1177 Han males from southwest China, including 545 patients with idiopathic NOA and 632 controls with normozoospermia. The results confirmed that the HLA-DRA rs3129878 was an NOA susceptibility locus in the present population. Along with our data, meta-analyses supported the association of the four GWAS-linked loci with NOA, whereas an additive effect of the four loci on NOA susceptibility was not found. Interestingly, the normozoospermic males with the risk genotypes of rs12097821 and rs3129878 + rs10842262 + rs12097821 were observed to have higher total sperm counts relative to non-risk genotypes, suggesting that the risk alleles of the genetic loci may not be via impairing spermatogenic ability to express susceptibility to NOA. These findings may advance our understanding of the role of the NOA susceptibility loci, although the results need to be confirmed in larger samples.


Assuntos
Azoospermia/genética , Loci Gênicos , Infertilidade Masculina/genética , Polimorfismo de Nucleotídeo Único , Espermatogênese/genética , Alelos , China , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla , Genótipo , Humanos , Masculino
17.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 32(2): 192-7, 2015 Apr.
Artigo em Zh | MEDLINE | ID: mdl-25863084

RESUMO

OBJECTIVE: To identify protein-protein interaction partners of PER1 (period circadian protein homolog 1), key component of the molecular oscillation system of the circadian rhythm in tumors using bacterial two-hybrid system technique. METHODS: Human cervical carcinoma cell Hela library was adopted. Recombinant bait plasmid pBT-PER1 and pTRG cDNA plasmid library were cotransformed into the two-hybrid system reporter strain cultured in a special selective medium. Target clones were screened. After isolating the positive clones, the target clones were sequenced and analyzed. RESULTS: Fourteen protein coding genes were identified, 4 of which were found to contain whole coding regions of genes, which included optic atrophy 3 protein (OPA3) associated with mitochondrial dynamics and homo sapiens cutA divalent cation tolerance homolog of E. coli (CUTA) associated with copper metabolism. There were also cellular events related proteins and proteins which are involved in biochemical reaction and signal transduction-related proteins. CONCLUSION: Identification of potential interacting proteins with PER1 in tumors may provide us new insights into the functions of the circadian clock protein PER1 during tumorigenesis.


Assuntos
Neoplasias/metabolismo , Proteínas Circadianas Period/metabolismo , Sequência de Bases , Linhagem Celular Tumoral , Escherichia coli/genética , Escherichia coli/metabolismo , Humanos , Dados de Sequência Molecular , Neoplasias/genética , Proteínas Circadianas Period/genética , Ligação Proteica , Proteínas/genética , Proteínas/metabolismo , Técnicas do Sistema de Duplo-Híbrido
18.
Genet Mol Biol ; 37(4): 708-15, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25505846

RESUMO

The mouse testis-enriched Znf230 gene, which encodes a type of RING finger protein, is present primarily in the nuclei of spermatogonia, the acrosome and the tail of spermatozoa. To investigate the role of Znf230 in spermatogenesis, we generated Znf230-deficient mice by disrupting Znf230 exon-5 and exon-6 using homologous recombination. The homozygous Znf230-knockout (KO) mice did not exhibit Znf230 mRNA expression and Znf230 protein production. Znf230 KO mice exhibited no obvious impairment in body growth or fertility. Male Znf230 KO mice had integral reproductive systems and mature sperm that were regular in number and shape. The developmental stages of male germ cells of Znf230 KO mice were also normal. We further examined variations in the transcriptomes of testicular tissue between Znf230 KO and wild-type mice through microarray analysis. The results showed that the mRNA level of one unclassified transcript 4921513I08Rik was increased and that the mRNA levels of three other transcripts, i.e., 4930448A20Rik, 4931431B13Rik and potassium channel tetramerisation domain containing 14(Kctd14), were reduced more than two-fold in Znf230 KO mice compared with wild-type mice. Using our current examination techniques, these findings suggested that Znf230 deficiency in mice may not affect growth, fertility or spermatogenesis.

19.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 45(1): 1-5, 2014 Jan.
Artigo em Zh | MEDLINE | ID: mdl-24527571

RESUMO

OBJECTIVE: To investigate the temporal and spatial features of mouse Rnf148 gene expression and the function of RING finger domain of Rnf148 protein. METHODS: The whole RNA was extracted from different tissues of adult mice, embryo in four developmental stages, and testes of postnatal mice respectively. RT-PCR and Northern blotting analysis were used to investigate the expression of Rnf148 gene in the above tissues. The in vitro expression vector for GST-Rnf148 fused protein was constructed, which encompassing the entire RING domain of Rnf148 protein. GST-Rnf148 fused protein was expressed in Escherichia coli. BL21(DE3) cells and purified with glutathione-sepharose 4B. In vitro ubiquitination assay was performed to analyze whether GST-Rnf148 fused protein possess the function of E3 ubiquitin ligase. RESULTS: The Mice Rnf148 mRNA expression was only observed in testis, and Northern blotting confirmed that there was only one 1.2 kb mRNA band present in mice testis. Rnf148 mRNA started to appear in the testis of day 21 mice, and then increased dramatically and reached to the highest level in day 25, and continued to express thereafter. GST-Rnf148 fused protein was induced and purified, in vitro ubiquitination reaction showed that the recombinant protein has E3 ubiquitin ligase activity. CONCLUSION: Rnf148 gene is specifically expressed in mice testis.


Assuntos
Testículo/metabolismo , Ubiquitina-Proteína Ligases/genética , Animais , Escherichia coli , Expressão Gênica , Masculino , Camundongos , RNA Mensageiro , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Ubiquitina-Proteína Ligases/metabolismo
20.
Andrology ; 12(3): 561-569, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37594248

RESUMO

BACKGROUND: Azoospermia factor C (AZFc) in the male-specific region of Y-chromosome (MSY) presents wide structure variation mainly due to frequent non-allele homologous recombination, leading to significant copy number variation of the AZFc-linked coding sequences involving in spermatogenesis. A large number of studies had been conducted to investigate the association between AZFc deletions and male infertility in certain Y chromosome genetic backgrounds, however, the influence of primary AZFc duplication on spermatogenesis remained controversial and the cause of the discrepant outcomes is unknown. METHODS: In the present study, a total of 1,102 unrelated Han Chinese males without any detectable AZF deletions were recruited from 2014 to 2019, including 411 controls with normozoospermia and 691 patients with idiopathic spermatogenic failure. Using multiple paralog ratio tests (PRTs), the structure duplications were classified by the copy number of the AZFc-linked amplicons and genes. The Y-chromosome haplogroup (Y-hg) was categorized by genetyping of MSY-linked polymorphism loci. The association of primary AZFc duplication with spermatogenic phenotype was investigated in males with the same Y-hg. RESULTS: Within Y-hg O3* group, the frequency of the gr/gr duplication in patients is significantly higher than that of controls (P = 1.29×10-3 , odds ratio (OR) 7.64, 95% confidence interval (CI) 1.79-32.57). Moreover, Y-hg O3* males with the gr/gr duplication presented a significantly lower sperm production compared with non-AZFc duplicated ones (sperm concentration: P = 1.46×10-3 ; total sperm count: P = 1.82 ×10-3 ). The b2/b3 duplication were identified clustered in Y-hg Cα2*, and the significant difference in the distribution was not observed between patients with spermatogenic failure and controls. CONCLUSION: The results suggest that, in the Han Chinese population, the gr/gr duplication is a predisposing genetic factor for spermatogenic impairment in males harboring Y-hg O3* . Meanwhile, the b2/b3 duplication may be fixed on a yet-unidentified subbranch of Y-hg Cα2* without significantly deleterious effect on spermatogenesis. Our findings provide evidence that the difference in the Y-hg composition may cause the discrepancy on the association of AZFc duplication with spermatogenic failure among the studied populations.


Assuntos
Azoospermia , Infertilidade Masculina , Mercúrio , Humanos , Masculino , Azoospermia/genética , Estudos de Casos e Controles , Variações do Número de Cópias de DNA , Sêmen , Infertilidade Masculina/genética , Cromossomos Humanos Y/genética , Espermatogênese/genética , China , Deleção Cromossômica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA