Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 158
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 49(1): 66-79.e5, 2018 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-29980436

RESUMEN

Genetic mutations of CARD14 (encoding CARMA2) are observed in psoriasis patients. Here we showed that Card14E138A/+ and Card14ΔQ136/+ mice developed spontaneous psoriasis-like skin inflammation, which resulted from constitutively activated CARMA2 via self-aggregation leading to the enhanced activation of the IL-23-IL-17A cytokine axis. Card14-/- mice displayed attenuated skin inflammation in the imiquimod-induced psoriasis model due to impaired IL-17A signaling in keratinocytes. CARMA2, mainly expressed in keratinocytes, associates with the ACT1-TRAF6 signaling complex and mediates IL-17A-induced NF-κB and MAPK signaling pathway activation, which leads to expression of pro-inflammatory factors. Thus, CARMA2 serves as a key mediator of IL-17A signaling and its constitutive activation in keratinocytes leads to the onset of psoriasis, which indicates an important role of NF-κB activation in keratinocytes in psoriatic initiation.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD/genética , Proteínas Adaptadoras de Señalización CARD/metabolismo , Dermatitis/genética , Mutación con Ganancia de Función , Guanilato-Quinasas/genética , Guanilato-Quinasas/metabolismo , Interleucina-17/metabolismo , Queratinocitos/metabolismo , Psoriasis/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Proteínas Adaptadoras de Señalización CARD/química , Proteínas Adaptadoras de Señalización CARD/deficiencia , Línea Celular , Citocinas/genética , Citocinas/metabolismo , Dermatitis/fisiopatología , Regulación de la Expresión Génica/efectos de los fármacos , Guanilato-Quinasas/química , Guanilato-Quinasas/deficiencia , Células HEK293 , Humanos , Imiquimod , Queratinocitos/patología , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Psoriasis/inducido químicamente , Psoriasis/fisiopatología , Transducción de Señal , Subgrupos de Linfocitos T/metabolismo , Factor 6 Asociado a Receptor de TNF/metabolismo
2.
Mol Cell ; 70(3): 395-407.e4, 2018 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-29727616

RESUMEN

Telomeres and telomere-binding proteins form complex secondary nucleoprotein structures that are critical for genome integrity but can present serious challenges during telomere DNA replication. It remains unclear how telomere replication stress is resolved during S phase. Here, we show that the BUB3-BUB1 complex, a component in spindle assembly checkpoint, binds to telomeres during S phase and promotes telomere DNA replication. Loss of the BUB3-BUB1 complex results in telomere replication defects, including fragile and shortened telomeres. We also demonstrate that the telomere-binding ability of BUB3 and kinase activity of BUB1 are indispensable to BUB3-BUB1 function at telomeres. TRF2 targets BUB1-BUB3 to telomeres, and BUB1 can directly phosphorylate TRF1 and promote TRF1 recruitment of BLM helicase to overcome replication stress. Our findings have uncovered previously unknown roles for the BUB3-BUB1 complex in S phase and shed light on how proteins from diverse pathways function coordinately to ensure proper telomere replication and maintenance.


Asunto(s)
Proteínas de Ciclo Celular/genética , Replicación del ADN/genética , Proteínas de Unión a Poli-ADP-Ribosa/genética , Proteínas Serina-Treonina Quinasas/genética , Telómero/genética , Línea Celular , Línea Celular Tumoral , ADN Helicasas/genética , Células HEK293 , Células HeLa , Humanos , Puntos de Control de la Fase M del Ciclo Celular/genética , Fase S/genética , Huso Acromático/genética , Proteínas de Unión a Telómeros/genética
3.
Nat Immunol ; 13(4): 387-95, 2012 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-22388039

RESUMEN

Stringent control of the type I interferon signaling pathway is important for maintaining host immune responses and homeostasis, yet the molecular mechanisms responsible for its tight regulation are still poorly understood. Here we report that the pattern-recognition receptor NLRP4 regulated the activation of type I interferon mediated by double-stranded RNA or DNA by targeting the kinase TBK1 for degradation. NLRP4 recruited the E3 ubiquitin ligase DTX4 to TBK1 for Lys48 (K48)-linked polyubiquitination at Lys670, which led to degradation of TBK1. Knockdown of either DTX4 or NLRP4 abrogated K48-linked ubiquitination and degradation of TBK1 and enhanced the phosphorylation of TBK1 and the transcription factor IRF3. Our results identify a previously unrecognized role for NLRP4 in the regulation of type I interferon signaling and provide molecular insight into the mechanisms by which NLRP4-DTX4 targets TBK1 for degradation.


Asunto(s)
Interferón Tipo I/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Represoras/metabolismo , Transducción de Señal/inmunología , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Línea Celular , Técnica del Anticuerpo Fluorescente , Técnicas de Silenciamiento del Gen , Humanos , Inmunidad Innata/inmunología , Immunoblotting , Inmunoprecipitación , Interferón Tipo I/inmunología , Fosforilación , Proteínas Serina-Treonina Quinasas/inmunología , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Represoras/inmunología , Transfección , Ubiquitina-Proteína Ligasas/inmunología , Ubiquitinación
4.
J Med Virol ; 95(6): e28826, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37254821

RESUMEN

The mechanistic understanding of virus infection and inflammation in many diseases is incomplete. Normally, messenger RNA (mRNA) tails of replication-dependent histones (RDH) that safeguard naked nuclear DNAs are protected by a specialized stem-loop instead of polyadenylation. Here, we showed that infection by various RNA viruses (including severe acute respiratory syndrome coronavirus 2) induced aberrant polyadenylation of RDH mRNAs (pARDH) that resulted in inflammation or cellular senescence, based on which we constructed a pARDH inflammation score (pARIS). We further investigated pARIS elevation in various disease conditions, including different types of virus infection, cancer, and cellular senescence. Notably, we found that pARIS was positively correlated with coronavirus disease 2019 severity in specific immune cell types. We also detected a subset of HIV-1 elite controllers characterized by pARDH "flipping" potentially mediated by HuR. Importantly, pARIS was positively associated with transcription of endogenous retrovirus but negatively associated with most immune cell infiltration in tumors of various cancer types. Finally, we identified and experimentally verified two pARIS regulators, ADAR1 and ZKSCAN1, which was first linked to inflammation. The ZKSCAN1 was known as a transcription factor but instead was shown to regulate pARIS as a novel RNA binding protein. Both regulators were upregulated under most infection and inflammation conditions. In conclusion, we unraveled a potential antiviral mechanism underlying various types of virus infections and cancers.


Asunto(s)
COVID-19 , Neoplasias , Humanos , Histonas , Poliadenilación , ARN Mensajero/metabolismo , Inflamación , Neoplasias/genética
5.
J Med Virol ; 95(8): e28980, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37522289

RESUMEN

Hepatitis B virus (HBV) infection and integration are important for hepatocellular carcinoma (HCC) initiation and progression, while disease mechanisms are still largely elusive. Here, we combined bulk and single-cell sequencing technologies to tackle the disease mechanisms of HBV-related HCC. We observed high HBV mutation rate and diversity only in tumors without HBV integration. We identified human somatic risk loci for HBV integration (VIMs). Transcription factors (TFs) enriched in VIMs were involved in DNA repair and androgen receptor (AR) signaling. Aberration of AR signaling was further observed by single-cell regulon analysis in HBV-infected hepatocytes, which showed remarkable interactions between AR and the complement system that, together with the X-linked ZXDB regulon that contains albumin (ALB), probably contribute to HCC male predominance. Complement system dysregulation caused by HBV infection was further confirmed by analyses of single-cell copy numbers and cell-cell communications. Finally, HBV infection-associated immune cells presented critical defects, including TXNIP in T cells, TYROBP in NK cells, and the X-linked TIMP1 in monocytes. We further experimentally validated our findings in multiple independent patient cohorts. Collectively, our work shed light on the pathogenesis of HBV-related HCC and other liver diseases that affect billions of people worldwide.


Asunto(s)
Carcinoma Hepatocelular , Hepatitis B Crónica , Hepatitis B , Neoplasias Hepáticas , Masculino , Humanos , Femenino , Virus de la Hepatitis B/genética , Neoplasias Hepáticas/patología , Multiómica , Hepatitis B/complicaciones , Hepatitis B/genética , Hepatitis B/patología , Integración Viral
6.
Cell ; 133(7): 1162-74, 2008 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-18585351

RESUMEN

Pluripotency is a unique biological state that allows cells to differentiate into any tissue type. Here we describe a candidate pluripotency factor, Ronin, that possesses a THAP domain, which is associated with sequence-specific DNA binding and epigenetic silencing of gene expression. Ronin is expressed primarily during the earliest stages of murine embryonic development, and its deficiency in mice produces periimplantational lethality and defects in the inner cell mass. Conditional knockout of Ronin prevents the growth of ES cells while forced expression of Ronin allows ES cells to proliferate without differentiation under conditions that normally do not promote self-renewal. Ectopic expression also partly compensates for the effects of Oct4 knockdown. We demonstrate that Ronin binds directly to HCF-1, a key transcriptional regulator. Our findings identify Ronin as an essential factor underlying embryogenesis and ES cell pluripotency. Its association with HCF-1 suggests an epigenetic mechanism of gene repression in pluripotent cells.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Desarrollo Embrionario , Células Madre Embrionarias/citología , Células Madre Pluripotentes/citología , Animales , Diferenciación Celular , Línea Celular , Proteínas de Unión al ADN/genética , Implantación del Embrión , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Expresión Génica , Genes Letales , Factor C1 de la Célula Huésped/metabolismo , Ratones , Proteínas Represoras , Técnicas del Sistema de Dos Híbridos
7.
Genes Dev ; 29(2): 157-70, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25547115

RESUMEN

PTEN [phosphatidylinositol (3,4,5)-trisphosphate phosphatase and tensin homolog deleted from chromosome 10], a phosphatase and critical tumor suppressor, is regulated by numerous post-translational modifications, including phosphorylation, ubiquitination, acetylation, and SUMOylation, which affect PTEN localization and protein stability. Here we report ADP-ribosylation as a new post-translational modification of PTEN. We identified PTEN as a novel substrate of tankyrases, which are members of the poly(ADP-ribose) polymerases (PARPs). We showed that tankyrases interact with and ribosylate PTEN, which promotes the recognition of PTEN by a PAR-binding E3 ubiquitin ligase, RNF146, leading to PTEN ubiquitination and degradation. Double knockdown of tankyrase1/2 stabilized PTEN, resulting in the subsequent down-regulation of AKT phosphorylation and thus suppressed cell proliferation and glycolysis in vitro and tumor growth in vivo. Furthermore, tankyrases were up-regulated and negatively correlated with PTEN expression in human colon carcinomas. Together, our study revealed a new regulation of PTEN and highlighted a role for tankyrases in the PTEN-AKT pathway that can be explored further for cancer treatment.


Asunto(s)
Neoplasias del Colon/fisiopatología , Neoplasias Colorrectales/fisiopatología , Fosfohidrolasa PTEN/metabolismo , Poli Adenosina Difosfato Ribosa/metabolismo , Tanquirasas/metabolismo , Línea Celular Tumoral , Proliferación Celular , Regulación Neoplásica de la Expresión Génica/genética , Técnicas de Silenciamiento del Gen , Glucólisis , Células HCT116 , Células HEK293 , Células HT29 , Humanos , Proteína Oncogénica v-akt/metabolismo , Fosfohidrolasa PTEN/genética , Fosforilación , Procesamiento Proteico-Postraduccional , Ubiquitinación
8.
Int J Cancer ; 150(9): 1504-1511, 2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-34985769

RESUMEN

As one of the hallmarks of cancer, gene fusions play an important role in tumorigenesis, and have been established as biomarkers and therapeutic targets. Although recent years have witnessed the development of gene fusion databases, a tool with interactive analytic functions remains lacking. Here, we introduce fusion profiling interactive analysis (FPIA), a web server to perform interactive and customizable analysis on fusion genes. With this platform, researchers can easily explore fusion-associated biological and molecular differences including gene expression, tumor purity and ploidy, mutation, copy number variations, protein expression, immune cell infiltration, stemness, telomere length, microsatellite instability, survival and novel peptides based on 33 cancer types from The Cancer Genome Atlas (TCGA) data. Currently, it contains 31 633 fusion events from 6910 patients. FPIA complements the existing gene fusion annotation databases with its multiomics analytic capacity, integrated analysis features, customized analysis selection and user-friendly design. The comprehensive data analyses by FPIA will greatly facilitate data mining, hypothesis generation and therapeutic target discovery. FPIA is available at http://bioinfo-sysu.com/fpia.


Asunto(s)
Variaciones en el Número de Copia de ADN , Neoplasias , Minería de Datos , Bases de Datos Genéticas , Perfilación de la Expresión Génica , Fusión Génica , Humanos , Neoplasias/patología
9.
EMBO Rep ; 21(4): e49076, 2020 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-32096305

RESUMEN

Repressor/activator protein 1 (RAP1) is a highly evolutionarily conserved protein found at telomeres. Although yeast Rap1 is a key telomere capping protein preventing non-homologous end joining (NHEJ) and consequently telomere fusions, its role at mammalian telomeres in vivo is still controversial. Here, we demonstrate that RAP1 is required to protect telomeres in replicative senescent human cells. Downregulation of RAP1 in these cells, but not in young or dividing pre-senescent cells, leads to telomere uncapping and fusions. The anti-fusion effect of RAP1 was further explored in a HeLa cell line where RAP1 expression was depleted through an inducible CRISPR/Cas9 strategy. Depletion of RAP1 in these cells gives rise to telomere fusions only when telomerase is inhibited. We further show that the fusions triggered by RAP1 loss are dependent upon DNA ligase IV. We conclude that human RAP1 is specifically involved in protecting critically short telomeres. This has important implications for the functions of telomeres in senescent cells.


Asunto(s)
Telómero , Factor de Transcripción AP-1 , Animales , Senescencia Celular/genética , Daño del ADN , Células HeLa , Humanos , Telómero/genética , Proteínas de Unión a Telómeros/genética
10.
Mol Cell ; 54(6): 960-974, 2014 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-24857548

RESUMEN

Many mutant p53 proteins (mutp53s) exert oncogenic gain-of-function (GOF) properties, but the mechanisms mediating these functions remain poorly defined. We show here that GOF mutp53s inhibit AMP-activated protein kinase (AMPK) signaling in head and neck cancer cells. Conversely, downregulation of GOF mutp53s enhances AMPK activation under energy stress, decreasing the activity of the anabolic factors acetyl-CoA carboxylase and ribosomal protein S6 and inhibiting aerobic glycolytic potential and invasive cell growth. Under conditions of energy stress, GOF mutp53s, but not wild-type p53, preferentially bind to the AMPKα subunit and inhibit AMPK activation. Given the importance of AMPK as an energy sensor and tumor suppressor that inhibits anabolic metabolism, our findings reveal that direct inhibition of AMPK activation is an important mechanism through which mutp53s can gain oncogenic function.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Carcinoma de Células Escamosas/genética , Metabolismo Energético/genética , Neoplasias de Cabeza y Cuello/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Acetil-CoA Carboxilasa/metabolismo , Animales , Antimetabolitos Antineoplásicos/farmacología , Movimiento Celular/genética , Proliferación Celular , Activación Enzimática/genética , Fluorouracilo/farmacología , Humanos , Ratones , Ratones Desnudos , Invasividad Neoplásica/genética , Trasplante de Neoplasias , Unión Proteica/genética , Interferencia de ARN , ARN Interferente Pequeño , Proteína S6 Ribosómica/metabolismo , Transducción de Señal/genética , Esferoides Celulares/citología , Carcinoma de Células Escamosas de Cabeza y Cuello , Trasplante Heterólogo , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/genética
11.
Nucleic Acids Res ; 48(11): 6019-6031, 2020 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-32379321

RESUMEN

ALT tumor cells often contain abundant DNA damage foci at telomeres and rely on the alternative lengthening of telomeres (ALT) mechanism to maintain their telomeres. How the telomere chromatin is regulated and maintained in these cells remains largely unknown. In this study, we present evidence that heterochromatin protein 1 binding protein 3 (HP1BP3) can localize to telomeres and is particularly enriched on telomeres in ALT cells. HP1BP3 inhibition led to preferential growth inhibition of ALT cells, which was accompanied by telomere chromatin decompaction, increased presence of C-circles, more pronounced ALT-associated phenotypes and elongated telomeres. Furthermore, HP1BP3 appeared to participate in regulating telomere histone H3K9me3 epigenetic marks. Taken together, our data suggest that HP1BP3 functions on telomeres to maintain telomere chromatin and represents a novel target for inhibiting ALT cancer cells.


Asunto(s)
Proliferación Celular , Ensamble y Desensamble de Cromatina , Heterocromatina/metabolismo , Histonas/metabolismo , Telómero/metabolismo , Línea Celular Tumoral , Daño del ADN , Proteínas de Unión al ADN , Eucromatina/genética , Eucromatina/metabolismo , Técnicas de Silenciamiento del Gen , Heterocromatina/genética , Código de Histonas , Histonas/química , Humanos , Metilación , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/deficiencia , Proteínas Nucleares/metabolismo , Multimerización de Proteína , Homeostasis del Telómero
12.
Biochem Biophys Res Commun ; 574: 63-69, 2021 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-34438348

RESUMEN

Human extended pluripotent stem (hEPS) cell is a newly established human embryonic stem cell (hESC) line with the capacity of chimerizing both embryonic and extraembryonic tissues compared with primed hESCs which are inefficient to contribute to the inner cell mass (ICM). The molecular mechanism underlying the pluripotency of hEPS cells is still not clear. We conducted RNA-seq and ATAC-seq analysis to investigate the differential expression profiling and genomic chromatin accessibility features. According to our data, more than 2000 genes were specially up-regulated in hEPS cells. Furthermore, the open chromatin regions in these two human embryonic stem cell lines were quite different. In hEPS cells, transcriptional factors binding motifs associated with pluripotency maintenance were enriched in chromatin accessible regions. Integrating the results from ATAC-seq and RNA-seq, we identified new regulatory features which were important for pluripotency maintenance and cell development in hEPS cells. Together, these results provided a new perspective on the understanding of molecular features of hESCs in different pluripotent states and a novel resource for further studies on regenerative medicine by using hEPS cells.


Asunto(s)
Cromatina/metabolismo , Células Madre Pluripotentes/metabolismo , Células Cultivadas , Humanos
13.
Nat Immunol ; 10(11): 1208-14, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19767757

RESUMEN

In response to invading microorganisms, macrophages engage in phagocytosis and rapidly release reactive oxygen species (ROS), which serve an important microbicidal function. However, how phagocytosis induces ROS production remains largely unknown. CARD9, a caspase-recruitment domain (CARD)-containing protein, is important for resistance to fungal and bacterial infection. The mechanism of CARD9-mediated bacterial clearance is still mostly unknown. Here we show that CARD9 is required for killing intracellular bacteria in macrophages. CARD9 associated with the GDP-dissociation inhibitor LyGDI in phagosomes after bacterial and fungal infection and binding of CARD9 suppressed LyGDI-mediated inhibition of the GTPase Rac1, thereby leading to ROS production and bacterial killing in macrophages. Thus, our studies identify a key pathway that leads to microbe-elicited ROS production.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/inmunología , Macrófagos/inmunología , Neuropéptidos/inmunología , Fagosomas/inmunología , Proteínas/inmunología , Especies Reactivas de Oxígeno/inmunología , Proteínas de Unión al GTP rac/inmunología , Animales , Proteínas Adaptadoras de Señalización CARD , Candida albicans/inmunología , Línea Celular , Inhibidores de Disociación de Guanina Nucleótido/inmunología , Inhibidores de Disociación de Guanina Nucleótido/metabolismo , Inmunidad Innata , Listeria monocytogenes/inmunología , Macrófagos/microbiología , Ratones , Ratones Noqueados , Neuropéptidos/metabolismo , Fagosomas/microbiología , Proteínas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Proteína de Unión al GTP rac1 , Inhibidores de la Disociación del Nucleótido Guanina rho-Específico
14.
Nucleic Acids Res ; 47(1): 391-405, 2019 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-30371886

RESUMEN

In human cells, telomeres are elongated by the telomerase complex that contains the reverse transcriptase hTERT and RNA template TERC/hTR. Poly(A)-specific ribonuclease (PARN) is known to trim hTR precursors by removing poly(A) tails. However, the precise mechanism of hTR 3' maturation remains largely unknown. Target of Egr1 (TOE1) is an Asp-Glu-Asp-Asp (DEDD) domain containing deadenylase that is mutated in the human disease Pontocerebella Hypoplasia Type 7 (PCH7) and implicated in snRNA and hTR processing. We have previously found TOE1 to localize specifically in Cajal bodies, where telomerase RNP complex assembly takes place. In this study, we showed that TOE1 could interact with hTR and the telomerase complex. TOE1-deficient cells accumulated hTR precursors, including oligoadenylated and 3'-extended forms, which was accompanied by impaired telomerase activity and shortened telomeres. Telomerase activity in TOE1-deficient cells could be rescued by wild-type TOE1 but not the catalytically inactive mutant. Our results suggest that hTR 3' end processing likely involves multiple exonucleases that work in parallel and/or sequentially, where TOE1 may function non-redundantly as a 3'-to-5' exonuclease in conjunction with PARN. Our study highlights a mechanistic link between TOE1 mutation, improper hTR processing and telomere dysfunction in diseases such as PCH7.


Asunto(s)
Enfermedades Cerebelosas/genética , Proteínas Nucleares/genética , Homeostasis del Telómero/genética , Telómero/genética , Enfermedades Cerebelosas/patología , Exonucleasas/genética , Exorribonucleasas/genética , Células HeLa , Humanos , Mutación , ARN/genética , Telomerasa/genética
15.
Stem Cells ; 37(6): 743-753, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30801858

RESUMEN

The scaffold protein Symplekin (Sympk) is involved in cytoplasmic RNA polyadenylation, transcriptional modulation, and the regulation of epithelial differentiation and proliferation via tight junctions. It is highly expressed in embryonic stem cells (ESCs), in which its role remains unknown. In this study, we found Sympk overexpression in mouse ESCs significantly increased colony formation, and Sympk deletion via CRISPR/Cas9 decreased colony formation. Sympk promoted ESC growth and its overexpression sustained ESC pluripotency, as assessed by teratoma and chimeric mouse formation. Genomic stability was preserved in these cells after long-term passage. The domain of unknown function 3453 (DUF3453) in Sympk was required for its interaction with the key pluripotent factor Oct4, and its depletion led to impaired colony formation. Sympk activated proliferation-related genes and suppressed differentiation-related genes. Our results indicate that Sympk interacts with Oct4 to promote self-renewal and pluripotency in ESCs and preserves genome integrity; accordingly, it has potential value for stem cell therapies. Stem Cells 2019;37:743-753.


Asunto(s)
Proteínas del Citoesqueleto/genética , Regulación del Desarrollo de la Expresión Génica , Genoma , Proteínas de la Membrana/genética , Células Madre Embrionarias de Ratones/metabolismo , Proteínas Nucleares/genética , Factor 3 de Transcripción de Unión a Octámeros/genética , Células Madre Pluripotentes/metabolismo , Animales , Sistemas CRISPR-Cas , Diferenciación Celular , Línea Celular , Proliferación Celular , Proteínas del Citoesqueleto/deficiencia , Eliminación de Gen , Perfilación de la Expresión Génica , Genes Reporteros , Inestabilidad Genómica , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteínas de la Membrana/deficiencia , Ratones , Células Madre Embrionarias de Ratones/citología , Proteínas Nucleares/deficiencia , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Células Madre Pluripotentes/citología , Transducción de Señal , Teratoma/genética , Teratoma/metabolismo , Teratoma/patología , Uniones Estrechas/metabolismo
16.
Immunity ; 34(6): 843-53, 2011 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-21703539

RESUMEN

Tight regulation of NF-κB signaling is essential for innate and adaptive immune responses, yet the molecular mechanisms responsible for its negative regulation are not completely understood. Here, we report that NLRX1, a NOD-like receptor family member, negatively regulates Toll-like receptor-mediated NF-κB activation. NLRX1 interacts with TRAF6 or IκB kinase (IKK) in an activation signal-dependent fashion. Upon LPS stimulation, NLRX1 is rapidly ubiquitinated, disassociates from TRAF6, and then binds to the IKK complex, resulting in inhibition of IKKα and IKKß phosphorylation and NF-κB activation. Knockdown of NLRX1 in various cell types markedly enhances IKK phosphorylation and the production of NF-κB-responsive cytokines after LPS stimulation. We further provide in vivo evidence that NLRX1 knockdown in mice markedly enhances susceptibility to LPS-induced septic shock and plasma IL-6 level. Our study identifies a previously unrecognized role for NLRX1 in the negative regulation of TLR-induced NF-κB activation by dynamically interacting with TRAF6 and the IKK complex.


Asunto(s)
Quinasa I-kappa B/inmunología , Proteínas Mitocondriales/inmunología , Transducción de Señal , Factor 6 Asociado a Receptor de TNF/inmunología , Receptores Toll-Like/inmunología , Animales , Línea Celular , Citocinas/inmunología , Humanos , Quinasa I-kappa B/metabolismo , Lipopolisacáridos/inmunología , Ratones , Proteínas Mitocondriales/metabolismo , FN-kappa B/inmunología , FN-kappa B/metabolismo , Fosforilación , Unión Proteica , Factor 6 Asociado a Receptor de TNF/metabolismo , Ubiquitinación
17.
Mol Cell ; 47(6): 839-50, 2012 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-22885005

RESUMEN

Both mitochondria, which are metabolic powerhouses, and telomeres, which help maintain genomic stability, have been implicated in cancer and aging. However, the signaling events that connect these two cellular structures remain poorly understood. Here, we report that the canonical telomeric protein TIN2 is also a regulator of metabolism. TIN2 is recruited to telomeres and associates with multiple telomere regulators including TPP1. TPP1 interacts with TIN2 N terminus, which contains overlapping mitochondrial and telomeric targeting sequences, and controls TIN2 localization. We have found that TIN2 is posttranslationally processed in mitochondria and regulates mitochondrial oxidative phosphorylation. Reducing TIN2 expression by RNAi knockdown inhibited glycolysis and reactive oxygen species (ROS) production and enhanced ATP levels and oxygen consumption in cancer cells. These results suggest a link between telomeric proteins and metabolic control, providing an additional mechanism by which telomeric proteins regulate cancer and aging.


Asunto(s)
Mitocondrias/metabolismo , Proteínas de Unión a Telómeros/metabolismo , Telómero/metabolismo , Adenosina Trifosfato/metabolismo , Línea Celular Tumoral , Núcleo Celular/metabolismo , Glucólisis/genética , Humanos , Fosforilación Oxidativa , Consumo de Oxígeno , Unión Proteica , Procesamiento Proteico-Postraduccional , Señales de Clasificación de Proteína , Interferencia de ARN , ARN Citoplasmático Pequeño , Especies Reactivas de Oxígeno/metabolismo , Complejo Shelterina , Proteínas de Unión a Telómeros/química , Proteínas de Unión a Telómeros/genética
18.
Mol Cancer ; 18(1): 106, 2019 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-31179925

RESUMEN

BACKGROUND: Cancer cells become immortalized through telomere maintenance mechanisms, such as telomerase reverse transcriptase (TERT) activation. In addition to maintaining telomere length, TERT activates manifold cell survival signaling pathways. However, telomerase-associated gene signatures in cancer remain elusive. METHODS: We performed a systematic analysis of TERT high (TERThigh) and low (TERTlow) cancers using multidimensional data from The Cancer Genome Atlas (TCGA). Multidimensional data were analyzed by propensity score matching weight algorithm. Coexpression networks were constructed by weight gene coexpression network analysis (WGCNA). Random forest classifiers were generated to identify cancer subtypes. RESULTS: The TERThigh-specific mRNA expression signature is associated with cell cycle-related coexpression modules across cancer types. Experimental screening of hub genes in the cell cycle module suggested TPX2 and EXO1 as potential regulators of telomerase activity and cell survival. MiRNA analysis revealed that the TERThigh-specific miR-17-92 cluster can target biological processes enriched in TERTlow cancer and that its expression is negatively correlated with the tumor/normal telomere length ratio. Intriguingly, TERThigh cancers tend to have mutations in extracellular matrix organization genes and amplify MAPK signaling. By mining the clinical actionable gene database, we uncovered a number of TERThigh-specific somatic mutations, amplifications and high expression genes containing therapeutic targets. Finally, a random forest classifier integrating telomerase-associated multi-omics signatures identifies two cancer subtypes showed profound differences in telomerase activity and patient survival. CONCLUSIONS: In summary, our results depict a telomerase-associated molecular landscape in cancers and provide therapeutic opportunities for cancer treatment.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Redes Reguladoras de Genes , MicroARNs/genética , Neoplasias/genética , Telomerasa/metabolismo , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Enzimas Reparadoras del ADN/genética , Exodesoxirribonucleasas/genética , Amplificación de Genes , Regulación Neoplásica de la Expresión Génica , Células Hep G2 , Humanos , Proteínas Asociadas a Microtúbulos/genética , Mutación , Neoplasias/enzimología , Regiones Promotoras Genéticas , Puntaje de Propensión
19.
EMBO Rep ; 18(8): 1412-1428, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28615293

RESUMEN

Repetitive DNA is prone to replication fork stalling, which can lead to genome instability. Here, we find that replication fork stalling at telomeres leads to the formation of t-circle-tails, a new extrachromosomal structure that consists of circular telomeric DNA with a single-stranded tail. Structurally, the t-circle-tail resembles cyclized leading or lagging replication intermediates that are excised from the genome by topoisomerase II-mediated cleavage. We also show that the DNA damage repair machinery NHEJ is required for the formation of t-circle-tails and for the resolution of stalled replication forks, suggesting that NHEJ, which is normally constitutively suppressed at telomeres, is activated in the context of replication stress. Inhibition of NHEJ or knockout of DNA-PKcs impairs telomere replication, leading to multiple-telomere sites (MTS) and telomere shortening. Collectively, our results support a "looping-out" mechanism, in which the stalled replication fork is cut out and cyclized to form t-circle-tails, and broken DNA is religated. The telomere loss induced by replication stress may serve as a new factor that drives replicative senescence and cell aging.


Asunto(s)
Replicación del ADN , Acortamiento del Telómero , Telómero/fisiología , Senescencia Celular , Reparación del ADN por Unión de Extremidades , ADN-Topoisomerasas de Tipo II/genética , ADN-Topoisomerasas de Tipo II/metabolismo , ADN Circular/química , ADN Circular/metabolismo , ADN de Cadena Simple/química , ADN de Cadena Simple/metabolismo , Inestabilidad Genómica , Humanos , Conformación de Ácido Nucleico , Telómero/genética
20.
Macromol Rapid Commun ; 40(5): e1800068, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29708298

RESUMEN

Delivery of CRISPR (clustered regularly interspaced short palindromic repeats)/CRISPR-associated protein-9 (Cas9) represents a major hurdle for successful clinical translation of genome editing tools. Owing to the large size of plasmids that encode Cas9 and single-guide RNA (sgRNA), genome editing efficiency mediated by current delivery carriers is still unsatisfactory to meet the requirement for its real applications. Herein, cationic polymer polyethyleneimine-ß-cyclodextrin (PC), known to be efficient for small plasmid transfection, is reported to likewise mediate efficient delivery of plasmid encoding Cas9 and sgRNA. Whereas PC can condense and encapsulate large plasmids at high N/P ratio, the delivery of plasmid results in efficient editing at two genome loci, namely, hemoglobin subunit beta (19.1%) and rhomboid 5 homolog 1 (RHBDF1) (7.0%). Sanger sequencing further confirms the successful genome editing at these loci. This study defines a new strategy for the delivery of the large plasmid encoding Cas9/sgRNA for efficient genome editing.


Asunto(s)
Proteína 9 Asociada a CRISPR/metabolismo , Sistemas CRISPR-Cas/genética , Edición Génica , Técnicas de Transferencia de Gen , Plásmidos/genética , Polímeros/química , Secuencia de Bases , Cationes , Células HeLa , Humanos , Nanopartículas/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA