Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 13(1): 22243, 2023 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-38097666

RESUMEN

Pyruvate kinase M2 (PKM2), a subtype of pyruvate kinase, plays a crucial role as a key enzyme in the final step of glycolysis. It is involved in regulating the tumor microenvironment and accelerating tumor progression. However, the relationship between PKM2 expression and the prognosis and immune infiltration remains unclear in lung cancer. In this study, we analyzed PKM2 expression in pan-cancer, and investigated its association with prognosis and immune cell infiltration of lung cancer by using multiple online databases, including Gent2, Tumor Immune Estimation Resource (TIMER), Gene Expression Profiling Interactive Analysis (GEPIA), PrognoScan, Kaplan-Meier plotter, and The Human Protein Atlas (HPA). The results showed that PKM2 expression is elevated in tumor tissues compared with the adjacent normal tissues of most cancers, including lung cancer. Prognostic analysis indicated that high expression of PKM2 was associated with poorer prognosis in overall lung cancer patients, especially in lung adenocarcinoma (LUAD). Notably, PKM2 exhibited a strong correlation with B cells and CD4+ T cells in LUAD; and with B cells, CD8+ T cells, CD4+ cells, and macrophages in lung squamous cell carcinoma (LUSC). Furthermore, PKM2 expression displayed a significant negative correlation with the expression of immune cell markers in both LUAD and LUSC. These findings suggested that PKM2 could serve as a promising prognostic biomarker for lung cancer and provided insights into its essential role in modulating the immune cell infiltration.


Asunto(s)
Adenocarcinoma del Pulmón , Carcinoma de Pulmón de Células no Pequeñas , Carcinoma de Células Escamosas , Neoplasias Pulmonares , Humanos , Adenocarcinoma del Pulmón/genética , Biomarcadores de Tumor/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , Neoplasias Pulmonares/genética , Pronóstico , Piruvato Quinasa/genética , Microambiente Tumoral/genética
2.
Ann Hum Genet ; 87(5): 241-247, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37461830

RESUMEN

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.


Asunto(s)
Acondroplasia , Humanos , Acondroplasia/genética , Acondroplasia/metabolismo , Acondroplasia/patología , Proteína de la Matriz Oligomérica del Cartílago/genética , Proteína de la Matriz Oligomérica del Cartílago/metabolismo , Condrocitos/metabolismo , Condrocitos/patología , Mutación
3.
Genes Genomics ; 44(5): 551-559, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35099763

RESUMEN

BACKGROUND: NPHS2 is the causative gene of nephrotic syndrome type 2 (MIM 600995) which often clinically manifests as steroid-resistant nephrotic syndrome (SRNS). The NPHS2 gene encodes a slit diaphragm (SD) associated protein podocin. OBJECTIVE: This study reported a novel disease-causing mutation of NPHS2 in a Chinese family with SRNS. We also investigated the pathogenic mechanism of the variants in this family. METHOD: A Chinese family with SRNS was recruited. Whole exome sequencing was performed to screen for disease-causing mutation. Sanger sequencing was used to confirm the results. In vitro functional experiments including immunoblotting, co-immunoprecipitation and double immunofluorescence staining were performed to explore the pathogenic mechanisms of mutations. RESULTS: In this family, compound heterozygous mutations of NPHS2 (c.467dupT and c.865A > G) were identified and segregated with the disease. The maternal c.865A > G was a novel variant, leading to amino acid substitution (p.K289E). In vitro functional assays indicated that c.467dupT (p.L156FfsX11) mutant lost interaction with nephrin. Both K289E and L156FfsX11 mutants showed sharply diminished plasma membrane localization. Furthermore, abnormal distribution of podocin mutants also altered the cell membrane localization of nephrin. CONCLUSION: We reported a family with SRNS caused by compound heterozygous mutations of NPHS2 (c.467dupT and c.865A > G). c.865A > G (p.K289E) in NPHS2 was a novel causative variant associated with SRNS. Both variants in this family not only affected the normal cell membrane localization of podocin, but also altered the cell membrane localization of nephrin which is the major architectural protein of SD.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/genética , Síndrome Nefrótico , China , Análisis Mutacional de ADN , Femenino , Humanos , Masculino , Mutación , Síndrome Nefrótico/genética , Esteroides
4.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 39(1): 52-55, 2022 Jan 10.
Artículo en Chino | MEDLINE | ID: mdl-34964967

RESUMEN

OBJECTIVE: To explore the genetic basis for a Chinese patient with retinitis pigmentosa (RP). METHODS: Whole exome sequencing (WES) was carried out to screen potential variant in the proband. Candidate variants were determined by taking consideration of clinical phenotype. Sanger sequencing was used to verify the variant in the proband and his parents. RESULTS: The proband was found to harbor compound heterozygous variants of c.8G>A (p.Cys3Tyr) and c.958_959insA (p.Arg320Glnfs*29) in the C2ORF71 gene, which has derived from his father and mother, respectively. Both variants were unreported previously. Based on the ACMG guidelines, they were predicted to be likely pathogenic and pathogenic, respectively. CONCLUSION: The novel compound heterozygous variants of the C2ORF71 gene probably underlay the pathogenesis of RP in the proband. Above finding has enriched the spectrum of C2ORF71 gene mutations and facilitated genetic counseling for the family.


Asunto(s)
Retinitis Pigmentosa , Pueblo Asiatico/genética , China , Humanos , Mutación , Linaje , Retinitis Pigmentosa/genética , Secuenciación del Exoma
5.
Mol Genet Genomic Med ; 9(10): e1795, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34535971

RESUMEN

BACKGROUND: Cone dystrophy with supernormal rod response (CDSRR) is an autosomal recessive retinal disorder characterized by myopia, dyschromatopsia, nyctalopia, photophobia, and nystagmus. CDSRR is caused by mutations in KCNV2, the gene encoding for an electrically silent Kv subunit (Kvs) named Kv8.2. METHODS: A Chinese CDSRR family was recruited. Complete ophthalmology clinical examinations were performed to clarify the phenotype. Genetic examination was underwent using whole exome sequencing (WES). In addition, a candidate gene was validated by Sanger sequencing. Expression analysis in vitro including immunoblotting, quantitative real-time PCR (qRT-PCR), and co-immunoprecipitation experiments was performed to investigate the pathogenic mechanism of the identified gene variants. RESULTS: WES identified two KCNV2 heterozygous mutations from the proband. Sanger sequencing validated that the patient's parents had, respectively, carried those two mutations. Further in vitro functional experiments indicated that the mutated alleles had led the Kv8.2 proteins to fail in expressing and interacting with the Kv2.1 protein, respectively. CONCLUSIONS: This study expanded the KCNV2 mutation spectrum. It can also be deduced that CDSRR has a broad heterogeneity. It is further confirmed that the inability expression of Kv8.2 proteins and the failure of Kv8.2 proteins to interact with Kv2.1 may have accounted for the etiology of CDSRR based on previous studies and this study.


Asunto(s)
Distrofia del Cono/diagnóstico , Distrofia del Cono/genética , Heterocigoto , Mutación , Canales de Potasio con Entrada de Voltaje/genética , Células Fotorreceptoras Retinianas Bastones/metabolismo , Células Fotorreceptoras Retinianas Bastones/patología , Adulto , China , Análisis Mutacional de ADN , Familia , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Pruebas Genéticas , Humanos , Masculino , Imagen Óptica , Linaje , Fenotipo , Tomografía de Coherencia Óptica , Pruebas de Visión , Secuenciación del Exoma
6.
Cell Death Differ ; 28(6): 1941-1954, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33469229

RESUMEN

Esophageal squamous cell carcinoma (ESCC) is one of the most common malignancies and cause of death from cancer in China. Previous studies showed that autophagy and apoptosis inhibition are critical for the survival of ESCC cells. However, the underlying mechanisms remain to be clarified. Recently, we found that PIWIL2, a novel cancer testis protein, is highly expressed in ESCC and associated with high T-stage and poor 5-year survival rate in patients. Our further study showed that PIWIL2 can directly bind to IKK and promote its phosphorylation, leading to phosphorylation of IκB and subsequently nuclear translocation of NF-κB for apoptosis inhibition. Meanwhile, PIWIL2 competitively inhibits binding of IKK to TSC1, and thus deactivate mTORC1 pathway which suppresses ULK1 phosphorylation and initiation of autophagy. The mouse xenograft model suggested that PIWIL2 can promote ESCC growth in an IKK-dependent manner. This present work firstly revealed that PIWIL2 can play a role in regulating autophagy and apoptosis, and is associated with poor prognosis in ESCC patients, providing novel insights into the roles of PIWIL2 in tumorigenesis.


Asunto(s)
Proteínas Argonautas/metabolismo , Biomarcadores de Tumor/metabolismo , Carcinoma de Células Escamosas de Esófago/genética , Animales , Apoptosis , Autofagia , Línea Celular Tumoral , Proliferación Celular , Humanos , Masculino , Ratones , Ratones Desnudos , Persona de Mediana Edad , Transfección
7.
Genes Genomics ; 42(9): 1067-1074, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32725577

RESUMEN

BACKGROUND: Duchenne muscular dystrophy (DMD) is a fatal X-linked recessive disorder with no effective treatment, which underscores the importance of avoiding the birth of children with DMD by identifying pathogenic mutations and obtaining an accurate prenatal diagnosis. OBJECTIVE: The objective of this study was to analyze the genetic defect of a Chinese family where all male patients have died of DMD. METHODS: Multiplex ligation dependent probe analysis (MLPA) and next-generation sequencing (NGS) were employed to detect DMD mutations. The candidate mutations were then validated by Sanger sequencing. In vitro splicing assay was further conducted to examine the potential effect of the novel DMD splice site mutation on splicing. RESULTS: We found that two rare DMD mutations c.1318G>A and c.6438+2T>G passed from generation to generation among female carriers and they may be used as genetic markers in the Chinese DMD family. In vitro splicing assay further revealed that the novel classical splice site mutation c.6438+2T>G gave rise to a new donor splice site, which resulted in a frame shift of the transcripts and a premature termination at position 2159 in exon 45 (p.Y2144Nfs*16). CONCLUSION: We found that two co-inherited mutations passed from generation to generation in female carriers and they may be used as genetic markers in the Chinese DMD family. Our findings not only expanded the DMD mutation spectrum, but also provided an important basis for identifying of female carriers and avoiding the birth of affected male children in this DMD family.


Asunto(s)
Distrofina/genética , Distrofia Muscular de Duchenne/genética , Adulto , Anciano , Pueblo Asiatico/genética , China , Distrofina/metabolismo , Exones/genética , Familia , Femenino , Marcadores Genéticos/genética , Heterocigoto , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Masculino , Persona de Mediana Edad , Distrofia Muscular de Duchenne/metabolismo , Mutación/genética , Linaje
8.
Biochem Biophys Res Commun ; 516(3): 819-824, 2019 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-31262447

RESUMEN

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.


Asunto(s)
Proteínas Argonautas/genética , Ciclo Celular/genética , Ciclina D1/genética , Regulación Neoplásica de la Expresión Génica , Procesamiento Proteico-Postraduccional , beta Catenina/genética , Proteínas Argonautas/metabolismo , Núcleo Celular/genética , Núcleo Celular/metabolismo , Proliferación Celular , Ciclina D1/metabolismo , Glucógeno Sintasa Quinasa 3 beta/genética , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Células HeLa , Células Hep G2 , Humanos , Fosforilación , Estabilidad Proteica , Transducción de Señal , Ubiquitinación , beta Catenina/metabolismo
9.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 36(6): 598-601, 2019 Jun 10.
Artículo en Chino | MEDLINE | ID: mdl-31055815

RESUMEN

OBJECTIVE: To analyze the clinical characteristics and genetic features of a family affected with isolated proteinuria. METHODS: Clinical data of the family was collected. Mutations of 191 renal disease-related genes in the proband were screened with next generation sequencing (NGS). Sanger sequencing was used to verify suspected mutations in his family members and 100 healthy controls. The impact of the mutation was predicted with online software SIFT. Frequency of the mutation was searched in databases including 1000 Genomic Project, ESP and ExAC. RESULTS: NGS and Sanger sequencing showed that the proband harbored compound heterozygous mutations of ADCK4 gene including c.748C>G (p.Asp250His) and c.1041G>T (p.Cys347*), which were respectively inherited from his mother and father whom were both non-symptomatic. CONCLUSION: The proband may have ADCK4-associated glomerulopathy due to the compound heterozygous mutations of the ADCK4 gene.


Asunto(s)
Secuenciación de Nucleótidos de Alto Rendimiento , Proteinuria , Análisis Mutacional de ADN , Familia , Humanos , Mutación , Proteinuria/genética
10.
J Cell Mol Med ; 23(7): 4689-4698, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31099187

RESUMEN

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.


Asunto(s)
Factores de Transcripción ARNTL/metabolismo , Proteínas Argonautas/metabolismo , Proteínas CLOCK/metabolismo , Ritmo Circadiano , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Proteolisis , Proteínas Argonautas/química , Línea Celular Tumoral , Elementos E-Box/genética , Humanos , Modelos Biológicos , Fosforilación , Unión Proteica , Dominios Proteicos , Activación Transcripcional/genética , Ubiquitinación , Familia-src Quinasas/metabolismo
11.
Cell Death Dis ; 10(2): 62, 2019 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-30683868

RESUMEN

Circadian rhythm is an autoregulatory rhythm, which is sustained by various mechanisms. The nucleocytoplasmic shuttling of BMAL1 is essential for CLOCK translocation between cytoplasm and nucleus and maintenance of the correct pace of the circadian clock. Here we showed that RAE1 and NUP98 can promote the degradation of BMAL1 and CLOCK. Knockdown of RAE1 and NUP98 suppressed BMAL1 shuttling, leading to cytoplasm accumulation of CLOCK. Furthermore, Chip assay showed that knockdown of RAE1 and NUP98 can enhance the interaction between CLOCK: BMAL1 and E-box region in the promoters of Per2 and Cry1 while reducing its transcription activation activity. Our present study firstly revealed that RAE1 and NUP98 are critical regulators for BMAL1 shuttling.


Asunto(s)
Factores de Transcripción ARNTL/metabolismo , Proteínas CLOCK/metabolismo , Proteínas Asociadas a Matriz Nuclear/metabolismo , Proteínas de Complejo Poro Nuclear/metabolismo , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Multimerización de Proteína , Proteolisis , Animales , Relojes Circadianos/fisiología , Ritmo Circadiano/fisiología , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Ratones , Células 3T3 NIH , Proteínas Asociadas a Matriz Nuclear/genética , Proteínas de Complejo Poro Nuclear/genética , Proteínas de Transporte Nucleocitoplasmático/genética , Plásmidos/genética , Activación Transcripcional/genética , Transfección , Ubiquitinación/genética
12.
Biol Reprod ; 100(4): 994-1007, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30541061

RESUMEN

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.


Asunto(s)
Desmetilación del ADN , Nucleoproteínas/genética , Espermátides/metabolismo , Activación Transcripcional , Animales , Células Cultivadas , Epigénesis Genética/fisiología , Femenino , Genes Ligados a X/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células 3T3 NIH , Nucleoproteínas/metabolismo , Regiones Promotoras Genéticas , Espermatogénesis/genética , Factores de Transcripción/fisiología , Activación Transcripcional/genética
13.
Exp Mol Med ; 50(10): 1-15, 2018 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-30287810

RESUMEN

Circadian genes control most of the physiological functions in cancer cells, including cell proliferation, migration, and invasion. The CLOCK and BMAL1 complex plays a central role in circadian rhythms. Previous studies have shown that circadian genes may act as oncogenes or tumor-suppressor genes. In addition, F-actin, regulated by RHOA, has been shown to participate in tumor progression. However, the roles of the CLOCK and BMAL1 genes in the regulation of tumor progression via the RHOA-ROCK-CFL pathway remain largely unclear. Here we first indicate that the rearrangement of F-actin is regulated by CLOCK and BMAL1. We found that CLOCK and BMAL1 can upregulate RHOA expression by inhibiting CUL3-mediated ubiquitination and activate RHOA by reducing the interaction between RHOA and RhoGDI. Consequently, CLOCK and BMAL1 control the expression of the components of the RHOA-ROCK-CFL pathway, which alters the dynamics of F-actin/G-actin turnover and promotes cancer cell proliferation, migration, and invasion. In conclusion, our research proposes a novel insight into the role of CLOCK and BMAL1 in tumor cells.


Asunto(s)
Factores de Transcripción ARNTL/metabolismo , Actinas/metabolismo , Proteínas CLOCK/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Animales , Línea Celular Tumoral , Proteínas Cullin/metabolismo , Humanos , Modelos Biológicos , Estabilidad Proteica , Ubiquitinación
14.
Cell Death Dis ; 9(4): 423, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29555935

RESUMEN

HDAC3 is involved in deacetylation of histone and non-histone proteins, having a key role in the regulation of gene transcription and also in the process of tumorgenesis. However, how HDAC3 is regulated in cancer remains largely unclear. Here, we showed that PIWIL2 can interact with HDAC3, leading to stabilization of HDAC3 from ubiquitin-mediated degradation by competitive association with E3 ubiquitin ligase Siah2. Furthermore, we found that expression of PIWIL2 enhanced HDAC3 activity via CK2α. PIWIL2 facilitated the interaction between HDAC3 and CK2α, thus exhibiting a promotion on the HDAC3 phosphorylation by CK2α. Further work showed that PIWIL2 could promote cell proliferation and suppress cell apoptosis via regulating HDAC3. Our present study firstly revealed that PIWIL2 can play a role in HDAC3-mediated epigenetic regulation on cancer cell proliferation and apoptosis. These findings provide a novel insight into the roles of PIWIL2 in tumorigenesis.


Asunto(s)
Proteínas Argonautas/metabolismo , Histona Desacetilasas/metabolismo , Proteínas Nucleares/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Anticuerpos/inmunología , Apoptosis , Proteínas Argonautas/antagonistas & inhibidores , Proteínas Argonautas/genética , Quinasa de la Caseína II/antagonistas & inhibidores , Quinasa de la Caseína II/genética , Quinasa de la Caseína II/metabolismo , Línea Celular Tumoral , Femenino , Histona Desacetilasas/genética , Humanos , Leupeptinas/farmacología , Proteínas Nucleares/inmunología , Fosforilación , Unión Proteica , Proteolisis/efectos de los fármacos , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Ubiquitina-Proteína Ligasas/inmunología , Ubiquitinación , Neoplasias del Cuello Uterino/metabolismo , Neoplasias del Cuello Uterino/patología
15.
Gene ; 642: 199-204, 2018 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-29126923

RESUMEN

The core circadian gene CLOCK plays an important role in regulating male reproduction. However, the underlying mechanism still remains unclear. In the present study, we executed yeast two-hybrid screening using cDNA fragment of CLOCK PAS A domain as bait, and identified RANBP9 as a novel protein interacting with CLOCK. The interaction between CLOCK and RANBP9 was further validated by in vivo and in vitro assays. Previous studies have confirmed that SF3B3 was a RANBP9 interacting protein. Subsequently, our study also found that CLOCK and SF3B3 can interact with each other by co-immunoprecipitation in mouse testis. In order to dissect the underlying mechanism of CLOCK in spermatogenesis, we also performed RNA-immunoprecipitation followed by high-throughput sequencing (RIP-Seq) in mouse testis. The result of sequence analyses and Gene Ontology enrichment analyses (biological processes) demonstrated that CLOCK can directly bind 186 key mRNA transcripts in mouse spermatogenesis. Taken together, our results firstly showed that CLOCK can interact with RANBP9 and bind with mRNAs, demonstrating that CLOCK is involved in alternative splicing in spermatogenesis. These results reveal a novel mechanism for CLOCK in spermatogenesis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Empalme Alternativo , Proteínas CLOCK/metabolismo , Proteínas del Citoesqueleto/metabolismo , Proteínas Nucleares/metabolismo , Espermatogénesis , Animales , Perfilación de la Expresión Génica , Inmunoprecipitación , Masculino , Ratones , Unión Proteica , ARN Mensajero/metabolismo , Análisis de Secuencia de ARN , Testículo/metabolismo
16.
Biomed Res Int ; 2017: 5809787, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29104872

RESUMEN

Pseudoachondroplasia (PSACH) is an autosomal dominant osteochondrodysplasia caused by mutations in the gene encoding cartilage oligomeric matrix protein (COMP). Accurate clinical diagnosis of PSACH is sometimes difficult. Here, we identified a novel COMP mutation (c.1675G>A, p.Glu559Lys) in a Chinese PSACH family. We detected the plasma levels of COMP and type II collagen (CTX-II) in the four affected individuals. The results showed the levels of plasma COMP significantly decreased and plasma CTX-II significantly increased in the three PSACH patients with COMP mutation. However, both plasma levels of COMP and CTX-II were not to have found significant difference between the presymptomatic carrier and the age-matched subjects. In vitro analysis and immunofluorescence displayed wild type COMP homogenously expressed in cytoplasm, but mutant proteins were irregularly accumulated inside the HEK-293 cells. Western blot revealed that the quantity of the mutant COMP was more compared to wild type COMP in cells after transfection for 12 hours and 24 hours. Subsequently, 3D structural analysis showed three changes have taken place in secondary structure of the mutant COMP. In conclusion, the novel mutation of COMP may result in intracellular accumulation of the mutant protein. Decreased plasma COMP and increased plasma CTX-II may potentially serve as diagnostic markers of PSACH but may not be applicable in the presymptomatic carrier.


Asunto(s)
Acondroplasia/genética , Proteína de la Matriz Oligomérica del Cartílago/genética , Colágeno Tipo II/genética , Fragmentos de Péptidos/genética , Acondroplasia/sangre , Acondroplasia/patología , Biomarcadores/sangre , Proteína de la Matriz Oligomérica del Cartílago/sangre , Proteína de la Matriz Oligomérica del Cartílago/química , Preescolar , China , Condrocitos/metabolismo , Condrocitos/patología , Colágeno Tipo II/sangre , Colágeno Tipo II/química , Femenino , Predisposición Genética a la Enfermedad , Células HEK293 , Humanos , Masculino , Conformación Molecular , Mutación/genética , Linaje , Fragmentos de Péptidos/sangre , Fragmentos de Péptidos/química , Conformación Proteica
17.
Oncotarget ; 8(33): 54913-54924, 2017 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-28903391

RESUMEN

Circadian rhythms are regulated by transcriptional and post-translational feedback loops generated by appropriate functions of clock proteins. Rhythmic degradation of the circadian clock proteins is critical for maintenance of the circadian oscillations. Notably, circadian clock does not work during spermatogenesis and can be disrupted in tumors. However, the underlying mechanism that suppresses circadian rhythms in germ cells and cancer cells remains largely unknown. Here we report that the cancer/testis antigen PIWIL2 can repress circadian rhythms both in the testis and cancer cells. By facilitating SRC binding with PI3K, PIWIL2 activates the PI3K-AKT pathway to phosphorylate and deactivate GSK3ß, suppressing GSK3ß-induced phosphorylation and degradation of circadian protein BMAL1 and CLOCK. Meanwhile, PIWIL2 can bind with E-Box sequences associated with the BMAL1/CLOCK complex to negatively regulate the transcriptional activation activity of promoters of clock-controlled genes. Taken together, our results first described a function for the germline-specific protein PIWIL2 in regulation of the circadian clock, providing a molecular link between spermatogenesis as well as tumorigenesis to the dysfunction of circadian rhythms.

18.
J Biol Chem ; 292(34): 14165-14175, 2017 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-28687631

RESUMEN

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.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Desarrollo Embrionario , Mesodermo/metabolismo , Proteína Nodal/agonistas , Transducción de Señal , Proteína smad3/agonistas , Proteínas de Pez Cebra/agonistas , Pez Cebra , Animales , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Embrión no Mamífero/anomalías , Embrión no Mamífero/citología , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/metabolismo , Desarrollo Embrionario/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica , Células HEK293 , Hematopoyesis/efectos de los fármacos , Humanos , Hibridación in Situ , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Mesodermo/anomalías , Mesodermo/citología , Mesodermo/efectos de los fármacos , Microinyecciones , Microscopía Fluorescente , Morfolinos/farmacología , Mutación , Proteína Nodal/genética , Proteína Nodal/metabolismo , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Elementos de Respuesta/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Proteína smad3/antagonistas & inhibidores , Proteína smad3/genética , Proteína smad3/metabolismo , Proteínas de Pez Cebra/antagonistas & inhibidores , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
19.
Gene ; 626: 89-94, 2017 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-28514642

RESUMEN

11ß-hydroxylase deficiency (11ß-OHD) occurs in about 5-8% of congenital adrenal hyperplasia (CAH). In this study, we identified three CYP11B1 (encoding Cytochrome P450 11B1) heterozygous mutations: c.1358G>C (p.R453Q), c.1229T>G (p.L410R) and c.1231G>T (p.G411C) in a Chinese CAH patient due to classic 11ß-OHD. His parents were healthy and respectively carried the prevalent mutation c.1358G>C (p.R453Q), and the two novel mutations c.1229T>G (p.L410R) and c.1231G>T (p.G411C). In vitro expression studies, immunofluorescence demonstrated that wild type and mutant (L410R and G411C) proteins of CYP11B1 were correctly expressed on the mitochondria, and enzyme activity assay revealed the mutant reduced the 11-hydroxylase activity to 10% (P<0.001) for the conversion of 11ß-deoxycortisol to cortisol. Subsequently, three dimensional homology models for the normal and mutant proteins were built by using the x-ray structure of the human CYP11B2 as a template. Interestingly, in the heme binding site I helix, a change from helix to loop in four amino acide took place in the mutant model. In conclusion, this study expands the spectrum of mutations in CYP11B1 causing to 11ß-OHD and provides evidence for prenatal diagnosis and genetic counseling. In addition, our results confirm the two novel CYP11B1 mutations led to impaired 11-hydroxylase activity in vitro.


Asunto(s)
Hiperplasia Suprarrenal Congénita/genética , Mutación Missense , Esteroide 11-beta-Hidroxilasa/genética , Hiperplasia Suprarrenal Congénita/diagnóstico , Adulto , Sitios de Unión , Cortodoxona/metabolismo , Femenino , Células HEK293 , Hemo/metabolismo , Heterocigoto , Humanos , Masculino , Linaje , Unión Proteica , Esteroide 11-beta-Hidroxilasa/química , Esteroide 11-beta-Hidroxilasa/metabolismo
20.
Sci Rep ; 7: 46376, 2017 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-28393858

RESUMEN

Human PIWIL2, aka HILI, is a member of PIWI protein family and overexpresses in various tumors. However, the underlying mechanisms of HILI in tumorigenesis remain largely unknown. TBCB has a critical role in regulating microtubule dynamics and is overexpressed in many cancers. Here we report that HILI inhibits Gigaxonin-mediated TBCB ubiquitination and degradation by interacting with TBCB, promoting the binding between HSP90 and TBCB, and suppressing the interaction between Gigaxonin and TBCB. Meanwhile, HILI can also reduce phosphorylation level of TBCB induced by PAK1. Our results showed that HILI suppresses microtubule polymerization and promotes cell proliferation, migration and invasion via TBCB for the first time, revealing a novel mechanism for HILI in tumorigenesis.


Asunto(s)
Proteínas Argonautas/metabolismo , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Proteínas del Citoesqueleto/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/metabolismo , Proteínas Argonautas/genética , Proteínas del Citoesqueleto/genética , Proteínas HSP90 de Choque Térmico/metabolismo , Células HeLa , Humanos , Proteínas Asociadas a Microtúbulos/genética , Fosforilación , Unión Proteica , Proteolisis , Tubulina (Proteína)/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...