Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
FASEB J ; 36(6): e22335, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35506565

RESUMEN

Dysregulated transforming growth factor-beta (TGF-ß) signaling contributes to fibrotic liver disease and hepatocellular cancer (HCC), both of which are associated with fatty liver disease. SIRT6 limits fibrosis by inhibiting TGF-ß signaling through deacetylating SMAD2 and SMAD3 and limits lipogenesis by inhibiting SREBP1 and SREBP2 activity. Here, we showed that, compared to wild-type mice, high-fat diet-induced fatty liver is worse in TGF-ß signaling-deficient mice (SPTBN1+/- ) and the mutant mice had reduced SIRT6 abundance in the liver. Therefore, we hypothesized that altered reciprocal regulation between TGF-ß signaling and SIRT6 contributes to these liver pathologies. We found that deficiency in SMAD3 or SPTBN1 reduced SIRT6 mRNA and protein abundance and impaired TGF-ß induction of SIRT6 transcripts, and that SMAD3 bound to the SIRT6 promoter, suggesting that an SMAD3-SPTBN1 pathway mediated the induction of SIRT6 in response to TGF-ß. Overexpression of SIRT6 in HCC cells reduced the expression of TGF-ß-induced genes, consistent with the suppressive role of SIRT6 on TGF-ß signaling. Manipulation of SIRT6 abundance in HCC cells altered sterol regulatory element-binding protein (SREBP) activity and overexpression of SIRT6 reduced the amount of acetylated SPTBN1 and the abundance of both SMAD3 and SPTBN1. Furthermore, induction of SREBP target genes in response to SIRT6 overexpression was impaired in SPTBN1 heterozygous cells. Thus, we identified a regulatory loop between SIRT6 and SPTBN1 that represents a potential mechanism for susceptibility to fatty liver in the presence of dysfunctional TGF-ß signaling.


Asunto(s)
Carcinoma Hepatocelular , Hígado Graso , Sirtuinas , Factor de Crecimiento Transformador beta , Animales , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Hígado Graso/genética , Hígado Graso/metabolismo , Fibrosis , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Ratones , Sirtuinas/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles , Factor de Crecimiento Transformador beta/metabolismo
2.
Biochem Biophys Res Commun ; 464(4): 1016-1021, 2015 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-26168722

RESUMEN

Although Metastatic-tumor antigen 1 (MTA1) is differentially expressed in metastatic cancer and coregulates the status and activity of nuclear receptors, its role upon estrogen receptor ß (ERß) - a potent tumor suppressor, remains poorly understood. Here we investigated whether MTA1 regulates the expression and functions of ERß, an ER isoform predominantly expressed in salivary gland cancer cells. We found that the depletion of the endogenous MTA1 in the HSG and HSY salivary duct carcinoma cell lines enhances the expression of ERß while MTA1 overexpression augmented the expression of ERß in salivary duct carcinoma cells. Furthermore, MTA1 knockdown inhibited the proliferations and invasion of HSG and HSY cells. The noted ERß downregulation by MTA1 overexpression involves the process of proteasomal degradation, as a proteasome inhibitor could block it. In addition, both MTA1 knockdown and ERß overexpression attenuated the cell migration and inhibited the ERK1/2 signaling in the both cell lines. These findings imply that MTA1 dysregulation in a subset of salivary gland cancer might promote aggressive phenotypes by compromising the tumor suppressor activity of ERß, and hence, MTA1-ERß axis might serve a new therapeutic target for the salivary gland cancer.


Asunto(s)
Receptor beta de Estrógeno/metabolismo , Histona Desacetilasas/metabolismo , Proteínas Represoras/metabolismo , Neoplasias de las Glándulas Salivales/metabolismo , Carcinoma Ductal/genética , Carcinoma Ductal/metabolismo , Carcinoma Ductal/patología , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Técnicas de Silenciamiento del Gen , Histona Desacetilasas/genética , Humanos , Invasividad Neoplásica , Fenotipo , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/genética , Neoplasias de las Glándulas Salivales/genética , Neoplasias de las Glándulas Salivales/patología , Transducción de Señal , Transactivadores , Regulación hacia Arriba
3.
Proc Natl Acad Sci U S A ; 108(10): 4200-5, 2011 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-21368136

RESUMEN

Despite ubiquitous expression and a high level of metastasis-associated protein 1 (MTA1) coregulator, the physiological role of the MTA1 coactivator remains unknown. We found that MTA1 is a bona fide coactivator and stimulator of tyrosine hydroxylase (TH) transcription in neuronal cells and that MTA1-null mice had lower TH expression in the striatum and substantial nigra. MTA1 physically achieves these functions by interacting directly with DJ1 (Parkinson disease 7) and in turn recruits the DJ1/MTA1/RNA polymerase II complex to the bicoid binding element (BBE) in the TH promoter. Furthermore, we found that the MTA1/DJ1 complex is required for optimum stimulation of the TH expression by paired like homeodomain transcription factor (Pitx3) homeodomain transcription factor and that the MTA1/DJ1 complex is recruited to the TH gene chromatin via the direct interaction of MTA1 with Pitx3. These findings reveal a role for MTA1 as an upstream coactivator of TH and advance the notion of polygenic regulation of a disease-causing gene by coordinated interactions of three regulatory proteins.


Asunto(s)
Transcripción Genética/genética , Tirosina 3-Monooxigenasa/genética , Animales , Cuerpo Estriado/enzimología , Proteínas de Homeodominio/metabolismo , Proteínas de Homeodominio/fisiología , Ratones , Ratones Noqueados , Proteínas Represoras , Sustancia Negra/enzimología , Transactivadores , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/fisiología
4.
Proc Natl Acad Sci U S A ; 108(21): 8791-6, 2011 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-21555589

RESUMEN

Although metastasis-associated protein 1 (MTA1), a component of the nucleosome remodeling and histone deacetylation complex, is widely up-regulated in human cancers and correlates with tumor metastasis, its regulatory mechanism and related signaling pathways remain unknown. Here, we report a previously unrecognized bidirectional autoregulatory loop between MTA1 and tumor suppressor alternative reading frame (ARF). MTA1 transactivates ARF transcription by recruiting the transcription factor c-Jun onto the ARF promoter in a p53-independent manner. ARF, in turn, negatively regulates MTA1 expression independently of p53 and c-Myc. In this context, ARF interacts with transcription factor specificity protein 1 (SP1) and promotes its proteasomal degradation by enhancing its interaction with proteasome subunit regulatory particle ATPase 6, thereby abrogating the ability of SP1 to stimulate MTA1 transcription. ARF also physically associates with MTA1 and affects its protein stability. Thus, MTA1-mediated activation of ARF and ARF-mediated functional inhibition of MTA1 represent a p53-independent bidirectional autoregulatory mechanism in which these two opposites act in concert to regulate cell homeostasis and oncogenesis, depending on the cellular context and the environment.


Asunto(s)
Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Histona Desacetilasas/genética , Homeostasis/genética , Neoplasias/etiología , Proteínas Represoras/genética , Línea Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina/antagonistas & inhibidores , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Regulación de la Expresión Génica , Histona Desacetilasas/metabolismo , Humanos , Sistemas de Lectura , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/metabolismo , Transactivadores , Activación Transcripcional , Proteína p53 Supresora de Tumor
5.
Cell Rep ; 43(9): 114676, 2024 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-39217614

RESUMEN

Obesity and fatty liver diseases-metabolic dysfunction-associated steatotic liver disease (MASLD) and metabolic dysfunction-associated steatohepatitis (MASH)-affect over one-third of the global population and are exacerbated in individuals with reduced functional aldehyde dehydrogenase 2 (ALDH2), observed in approximately 560 million people. Current treatment to prevent disease progression to cancer remains inadequate, requiring innovative approaches. We observe that Aldh2-/- and Aldh2-/-Sptbn1+/- mice develop phenotypes of human metabolic syndrome (MetS) and MASH with accumulation of endogenous aldehydes such as 4-hydroxynonenal (4-HNE). Mechanistic studies demonstrate aberrant transforming growth factor ß (TGF-ß) signaling through 4-HNE modification of the SMAD3 adaptor SPTBN1 (ß2-spectrin) to pro-fibrotic and pro-oncogenic phenotypes, which is restored to normal SMAD3 signaling by targeting SPTBN1 with small interfering RNA (siRNA). Significantly, therapeutic inhibition of SPTBN1 blocks MASH and fibrosis in a human model and, additionally, improves glucose handling in Aldh2-/- and Aldh2-/-Sptbn1+/- mice. This study identifies SPTBN1 as a critical regulator of the functional phenotype of toxic aldehyde-induced MASH and a potential therapeutic target.

6.
Genes Cancer ; 15: 1-14, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38323119

RESUMEN

Hepatocellular carcinoma (HCC) is the third leading cause of death from cancer worldwide but is often diagnosed at an advanced incurable stage. Yet, despite the urgent need for blood-based biomarkers for early detection, few studies capture ongoing biology to identify risk-stratifying biomarkers. We address this gap using the TGF-ß pathway because of its biological role in liver disease and cancer, established through rigorous animal models and human studies. Using machine learning methods with blood levels of 108 proteomic markers in the TGF-ß family, we found a pattern that differentiates HCC from non-HCC in a cohort of 216 patients with cirrhosis, which we refer to as TGF-ß based Protein Markers for Early Detection of HCC (TPEARLE) comprising 31 markers. Notably, 20 of the patients with cirrhosis alone presented an HCC-like pattern, suggesting that they may be a group with as yet undetected HCC or at high risk for developing HCC. In addition, we found two other biologically relevant markers, Myostatin and Pyruvate Kinase M2 (PKM2), which were significantly associated with HCC. We tested these for risk stratification of HCC in multivariable models adjusted for demographic and clinical variables, as well as batch and site. These markers reflect ongoing biology in the liver. They potentially indicate the presence of HCC early in its evolution and before it is manifest as a detectable lesion, thereby providing a set of markers that may be able to stratify risk for HCC.

7.
J Biol Chem ; 286(51): 43793-43808, 2011 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-21965678

RESUMEN

Metastasis tumor antigen 1 (MTA1), a component of the Mi-2·nucleosome remodeling and deacetylase complex, plays a crucial role in gene transcription, but the mechanism involved remains largely unknown. Here, we report that MTA1 is a substrate for small ubiquitin-related modifier 2/3 (SUMO2/3) in vivo. Protein inhibitor of activated STAT (PIAS) proteins enhance SUMOylation of MTA1 and may participate in paralog-selective SUMOylation, whereas sentrin/SUMO-specific protease 1 (SENP1) and 2 may act as deSUMOylation enzymes for MTA1. Moreover, MTA1 contains a functional SUMO-interacting motif (SIM) at its C terminus, and SIM is required for the efficient SUMOylation of MTA1. SUMO conjugation on Lys-509, which is located within the SUMO consensus site, together with SIM synergistically regulates the co-repressor activity of MTA1 on PS2 transcription, probably by recruiting HDAC2 onto the PS2 promoter. Interestingly, MTA1 may up-regulate the expression of SUMO2 via interaction with RNA polymerase II and SP1 at the SUMO2 promoter. These findings not only provide novel mechanistic insights into the regulation of the transcriptional repressor function of MTA1 by SUMOylation and SIM but also uncover a potential function of MTA1 in modulating the SUMOylation pathway.


Asunto(s)
Regulación de la Expresión Génica , Histona Desacetilasa 2/química , Histona Desacetilasas/química , Proteínas Represoras/química , Proteína SUMO-1/metabolismo , Secuencias de Aminoácidos , Animales , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Glutatión Transferasa/metabolismo , Histona Desacetilasas/metabolismo , Humanos , Técnicas In Vitro , Lisina/química , Modelos Biológicos , Unión Proteica , Procesamiento Proteico-Postraduccional , Estructura Terciaria de Proteína , ARN Interferente Pequeño/metabolismo , Proteínas Represoras/metabolismo , Sumoilación , Transactivadores
8.
Hepatology ; 54(1): 285-95, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21488078

RESUMEN

UNLABELLED: Schistosoma haematobium is responsible for two-thirds of the world's 200 million to 400 million cases of human schistosomiasis. It is a group 1 carcinogen and a leading cause of bladder cancer that occurs after years of chronic inflammation, fibrosis, and hyperproliferation in the host liver. The coevolution of blood flukes of the genus Schistosoma and their human hosts is paradigmatic of long-term parasite development, survival, and maintenance in mammals. However, the contribution of host genes, especially those discrete from the immune system, necessary for parasite establishment and development remains poorly understood. This study investigated the role of metastasis-associated protein-1 gene (Mta1) product in the survival of S. haematobium and productive infection in the host. Using a Mta-1 null mouse model, here we provide genetic evidence to suggest that MTA1 expression positively influences survival and/or maturation of schistosomes in the host to patency, as we reproducibly recovered significantly fewer S. haematobium worms and eggs from Mta1-/- mice than wild-type mice. In addition, we found a distinct loss of cytokine interdependence and aberrant Th1 and Th2 cytokine responses in the Mta1-/- mice compared to age-matched wild-type mice. Thus, utilizing this Mta1-null mouse model, we identified a distinct contribution of the mammalian MTA1 in establishing a productive host-parasite interaction and thus revealed a host factor critical for the optimal survival of schistosomes and successful parasitism. Moreover, MTA1 appears to play a significant role in driving inflammatory responses to schistosome egg-induced hepatic granulomata reactions, and thus offers a survival cue for parasitism as well as an obligatory contribution of liver in schistosomiasis. CONCLUSION: These findings raise the possibility to develop intervention strategies targeting MTA1 to reduce the global burden of schistosomiasis, inflammation, and neoplasia.


Asunto(s)
Predisposición Genética a la Enfermedad , Inflamación/genética , Neoplasias Hepáticas/genética , Esquistosomiasis/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Interacciones Huésped-Parásitos/fisiología , Humanos , Inflamación/metabolismo , Inflamación/patología , Hígado/inmunología , Hígado/parasitología , Hígado/patología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Ratones , Ratones Noqueados , Proteínas Represoras , Schistosoma haematobium/inmunología , Schistosoma haematobium/fisiología , Esquistosomiasis/metabolismo , Esquistosomiasis/patología , Transducción de Señal/fisiología , Células TH1/metabolismo , Células TH1/patología , Células Th2/metabolismo , Células Th2/patología , Transactivadores
9.
Hepatology ; 54(4): 1388-97, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21725997

RESUMEN

UNLABELLED: Based on the recently established role for the master coregulator MTA1 and MTA1-containing nuclear remodeling complexes in oncogenesis and inflammation, we explored the links between parasitism by the carcinogenic liver fluke Opisthorchis viverrini and this coregulator using both an Mta1(-/-) mouse model of infection and a tissue microarray of liver fluke-induced human cholangiocarcinomas (CCAs). Intense foci of inflammation and periductal fibrosis in the liver and kidneys of wild-type Mta1(+/+) mice were evident at 23 days postinfection with O. viverrini. In contrast, little inflammatory response was observed in the same organs of infected Mta1(-/-) mice. Livers of infected Mta1(+/+) mice revealed strong up-regulation of fibrosis-associated markers such as cytokeratins 18 and 19 and annexin 2, as determined both by immunostaining and by reverse-transcription polymerase chain reaction compared with infected Mta1(-/-) mice. CD4 expression was up-regulated by infection in the livers of both experimental groups; however, its levels were several-fold higher in the Mta1(+/+) mice than in infected Mta1(-/-) mice. Mta1(-/-) infected mice also exhibited significantly higher systemic and hepatic levels of host cytokines such as interleukin (IL)-12p70, IL-10, and interferon-γ compared with the levels of these cytokines in the Mta1(+/+) mice, suggesting an essential role of MTA1 in the cross-regulation of the Th1 and Th2 responses, presumably due to chromatin remodeling of the target chromatin genes. Immunohistochemical analysis of ≈ 300 liver tissue cores from confirmed cases of O. viverrini-induced CCA showed that MTA1 expression was elevated in >80% of the specimens. CONCLUSION: These findings suggest that MTA1 status plays an important role in conferring an optimal cytokine response in mice following infection with O. viverrini and is a major player in parasite-induced CCA in humans.


Asunto(s)
Neoplasias de los Conductos Biliares/parasitología , Colangiocarcinoma/parasitología , Histona Desacetilasas/metabolismo , Opistorquiasis/genética , Proteínas Represoras/metabolismo , Animales , Antígenos Helmínticos/análisis , Neoplasias de los Conductos Biliares/genética , Neoplasias de los Conductos Biliares/patología , Conductos Biliares Intrahepáticos , Biomarcadores/análisis , Colangiocarcinoma/genética , Colangiocarcinoma/patología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Fasciola hepatica/genética , Fasciola hepatica/metabolismo , Histona Desacetilasas/genética , Interacciones Huésped-Parásitos/genética , Humanos , Inmunohistoquímica , Neoplasias Hepáticas Experimentales , Ratones , Opistorquiasis/fisiopatología , Opisthorchis/genética , Opisthorchis/inmunología , Distribución Aleatoria , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Represoras/genética , Medición de Riesgo , Sensibilidad y Especificidad , Transactivadores , Regulación hacia Arriba
10.
EMBO Rep ; 11(9): 691-7, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20651739

RESUMEN

High expression of metastasis-associated protein 1 co-regulator (MTA1), a component of the nuclear remodelling and histone deacetylase complex, has been associated with human tumours. However, the precise role of MTA1 in tumorigenesis remains unknown. In this study, we show that induced levels of MTA1 are sufficient to transform Rat1 fibroblasts and that the transforming potential of MTA1 is dependent on its acetylation at Lys626. Underlying mechanisms of MTA1-mediated transformation include activation of the Ras-Raf pathway by MTA1 but not by acetylation-inactive MTA1; this was due to the repression of Galphai2 transcription, which negatively influences Ras activation. We observed that acetylated MTA1-histone deacetylase (HDAC) interaction was required for the recruitment of the MTA1-HDAC complex to the Galphai2 regulatory element and consequently for the repression of Galphai2 transcription and expression leading to activation of the Ras-Raf pathway. The findings presented in this study provide for the first time--to the best of our knowledge--evidence of acetylation-dependent oncogenic activity of a cancer-relevant gene product.


Asunto(s)
Transformación Celular Neoplásica , Histona Desacetilasas/metabolismo , Oncogenes , Proteínas Represoras/metabolismo , Acetilación , Animales , Línea Celular , Movimiento Celular , Femenino , Fibroblastos/citología , Fibroblastos/fisiología , Subunidad alfa de la Proteína de Unión al GTP Gi2/genética , Subunidad alfa de la Proteína de Unión al GTP Gi2/metabolismo , Regulación Neoplásica de la Expresión Génica , Histona Desacetilasas/genética , Humanos , Lisina/metabolismo , Ratones , Ratones Desnudos , Neoplasias Experimentales , Proteínas Represoras/genética , Transactivadores , Transcripción Genética , Trasplante Heterólogo , Proteínas ras/genética , Proteínas ras/metabolismo
11.
Proc Natl Acad Sci U S A ; 106(41): 17493-8, 2009 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-19805145

RESUMEN

Metastasis-associated protein 1 (MTA1), a component of the nucleosome remodeling and histone deacetylation (NuRD) complex, is widely upregulated in human cancers. However, the mechanism for regulating its protein stability remains unknown. Here we report that MTA1 is an ubiquitinated protein and targeted by the RING-finger E3 ubiquitin-protein ligase constitutive photomorphogenesis protein 1 (COP1) for degradation via the ubiquitin-proteasome pathway. Induced expression of wild-type COP1 but not its RING motif mutants promotes the ubiquitination and degradation of MTA1, indicating that the ligase activity is required for the COP1-mediated proteolysis of MTA1. Conversely, depletion of endogenous COP1 resulted in a marked decrease in MTA1 ubiquitination, accompanied by a pronounced accumulation of MTA1 protein. MTA1, in turn, destabilizes COP1 by promoting its autoubiquitination, thus creating a tight feedback loop that regulates both MTA1 and COP1 protein stability. Accordingly, disruption of the COP1-mediated proteolysis by ionizing radiation leads to MTA1 stabilization, accompanied by an increased coregulatory function of MTA1 on its target. Furthermore, we discovered that MTA1 is required for optimum DNA double-strand break repair after ionizing radiation. These findings provide novel insights into the regulation of MTA1 protein and reveal a novel function of MTA1 in DNA damage response.


Asunto(s)
Histona Desacetilasas/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Línea Celular , Daño del ADN , Reparación del ADN , Estabilidad de Enzimas , Fibroblastos/citología , Fibroblastos/fisiología , Histona Desacetilasas/química , Histona Desacetilasas/efectos de la radiación , Humanos , Ratones , Proteínas Nucleares/genética , Radiación Ionizante , Proteínas Represoras/química , Proteínas Represoras/efectos de la radiación , Transactivadores , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/genética
12.
J Biol Chem ; 285(26): 19802-12, 2010 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-20427275

RESUMEN

MTA1 (metastasis-associated protein 1), an integral component of the nucleosome remodeling and deacetylase complex, has recently been implicated in the ionizing radiation-induced DNA damage response. However, whether MTA1 also participates in the UV-induced DNA damage checkpoint pathway remains unknown. In response to UV radiation, ATR (ataxia teleangiectasia- and Rad3-related) is the major kinase activated that orchestrates cell cycle progression with DNA repair machinery by phosphorylating and activating a number of downstream substrates, such as Chk1 (checkpoint kinase 1) and H2AX (histone 2A variant X). Here, we report that UV radiation stabilizes MTA1 in an ATR-dependent manner and increases MTA1 binding to ATR. On the other hand, depletion of MTA1 compromises the ATR-mediated Chk1 activation following UV treatment, accompanied by a marked down-regulation of Chk1 and its interacting partner Claspin, an adaptor protein that is required for the phosphorylation and activation of Chk1 by ATR. Furthermore, MTA1 deficiency decreases the induction of phosphorylated H2AX (referred to as gamma-H2AX) and gamma-H2AX focus formation after UV treatment. Consequently, depletion of MTA1 results in a defect in the G(2)-M checkpoint and increases cellular sensitivity to UV-induced DNA damage. Thus, MTA1 is required for the activation of the ATR-Claspin-Chk1 and ATR-H2AX pathways following UV treatment, and the noted abrogation of the DNA damage checkpoint in the MTA1-depleted cells may be, at least in part, a consequence of dysregulation of the expression of these two pathways. These findings suggest that, in addition to its role in the repair of double strand breaks caused by ionizing radiation, MTA1 also participates in the UV-induced ATR-mediated DNA damage checkpoint pathway.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Daño del ADN , Histona Desacetilasas/metabolismo , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Represoras/metabolismo , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Western Blotting , Proteínas de Ciclo Celular/genética , Línea Celular , Línea Celular Tumoral , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Reparación del ADN , Relación Dosis-Respuesta en la Radiación , Histona Desacetilasas/genética , Histonas/genética , Histonas/metabolismo , Humanos , Cinética , Fosforilación/efectos de la radiación , Biosíntesis de Proteínas/efectos de la radiación , Proteínas Quinasas/genética , Proteínas Serina-Treonina Quinasas/genética , Interferencia de ARN , Proteínas Represoras/genética , Transactivadores , Ubiquitinación/efectos de la radiación , Rayos Ultravioleta
13.
J Biol Chem ; 285(13): 10044-10052, 2010 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-20071335

RESUMEN

Although metastasis-associated protein 1 (MTA1), a component of the nucleosome remodeling and deacetylase (NuRD) complex, is a DNA-damage response protein and regulates p53-dependent DNA repair, it remains unknown whether MTA1 also participates in p53-independent DNA damage response. Here, we provide evidence that MTA1 is a p53-independent transcriptional corepressor of p21(WAF1), and the underlying mechanism involves recruitment of MTA1-histone deacetylase 2 (HDAC2) complexes onto two selective regions of the p21(WAF1) promoter. Accordingly, MTA1 depletion, despite its effect on p53 down-regulation, superinduces p21(WAF1), increases p21(WAF1) binding to proliferating cell nuclear antigen (PCNA), and decreases the nuclear accumulation of PCNA in response to ionizing radiation. In support of a p53-independent role of MTA1 in DNA damage response, we further demonstrate that induced expression of MTA1 in p53-null cells inhibits p21(WAF1) promoter activity and p21(WAF1) binding to PCNA. Consequently, MTA1 expression in p53-null cells results in increased induction of gamma H2AX foci and DNA double strand break repair, and decreased DNA damage sensitivity following ionizing radiation treatment. These findings uncover a new target of MTA1 and the existence of an additional p53-independent role of MTA1 in DNA damage response, at least in part, by modulating the p21(WAF1)-PCNA pathway, and thus, linking two previously unconnected NuRD complex and DNA-damage response pathways.


Asunto(s)
Antígenos Nucleares/metabolismo , Núcleo Celular/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Histona Desacetilasas/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Humanos , Ratones , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas , Transactivadores
14.
Sci Transl Med ; 13(624): eabk2267, 2021 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-34910547

RESUMEN

The prevalence of nonalcoholic steatohepatitis (NASH) and liver cancer is increasing. De novo lipogenesis and fibrosis contribute to disease progression and cancerous transformation. Here, we found that ß2-spectrin (SPTBN1) promotes sterol regulatory element (SRE)­binding protein (SREBP)­stimulated lipogenesis and development of liver cancer in mice fed a high-fat diet (HFD) or a western diet (WD). Either hepatocyte-specific knockout of SPTBN1 or siRNA-mediated therapy protected mice from HFD/WD-induced obesity and fibrosis, lipid accumulation, and tissue damage in the liver. Biochemical analysis suggested that HFD/WD induces SPTBN1 and SREBP1 cleavage by CASPASE-3 and that the cleaved products interact to promote expression of genes with sterol response elements. Analysis of human NASH tissue revealed increased SPTBN1 and CASPASE-3 expression. Thus, our data indicate that SPTBN1 represents a potential target for therapeutic intervention in NASH and liver cancer.


Asunto(s)
Neoplasias , Enfermedad del Hígado Graso no Alcohólico , Animales , Dieta Alta en Grasa/efectos adversos , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Neoplasias/metabolismo , Enfermedad del Hígado Graso no Alcohólico/genética , Espectrina/metabolismo
15.
Cancer Metastasis Rev ; 28(1-2): 51-63, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19165420

RESUMEN

Some of the characteristics of cancer cells are high rates of cell proliferation, cell survival, and the ability to invade surrounding tissue. The cytoskeleton has an essential role in these processes. Dynamic changes in the cytoskeleton are necessary for cell motility and cancer cells are dependent on motility for invasion and metastasis. The signaling pathways behind the reshaping and migrating properties of the cytoskeleton in cancer cells involve a group of Ras-related small GTPases and their effectors, including the p21-activated kinases (Paks). Paks are a family of serine/threonine protein kinases comprised of six isoforms (Pak 1-6), all of which are direct targets of the small GTPases Rac and Cdc42. Besides their role in cytoskeletal dynamics, Paks have recently been shown to regulate various other cellular activities, including cell survival, mitosis, and transcription. Paks are overexpressed and/or hyperactivated in several human tumors and their role in cell transformation makes them attractive therapeutic targets. Pak-targeted therapeutics may efficiently inhibit certain types of tumors and efforts to identify selective Pak-inhibitors are underway.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Neoplasias/metabolismo , Quinasas p21 Activadas/metabolismo , Animales , Apoptosis , Transformación Celular Neoplásica , Citoesqueleto/metabolismo , Humanos , Estructura Terciaria de Proteína , Transducción de Señal , Especificidad por Sustrato , Proteína de Unión al GTP cdc42/metabolismo , Proteínas de Unión al GTP rac/metabolismo
16.
Genes Cancer ; 11(1-2): 43-52, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32577156

RESUMEN

Recently, we observed that the TGF-ß pathway is altered in 39% of HCCs. The alterations are correlated with a raised HMGA2 level. Therefore, we compared genetic alterations of HMGA2 and 43 TGF-ß pathway core genes in HCC patients from TCGA database. Genetic alterations of 15 genes, including INHBE, INHBC, GDF11, ACVRL and TGFB2 out of 43 core genes, highly-moderately matched that of HMGA2. Co-occurrences of mutation amplification, gains, deletions and high/low mRNA of HMGA2 with those of the core genes were highly significant in INHBE, INHBC, ACVR1B, ACVRL and GDF11. Mass spectrometry studies revealed that HMGA2 interacted with an E3 ligase, PJA1, and that this interaction is enhanced by TGF-ß treatment in the nuclear of HCC cells. Co-localization of nuclear PJA1 and HMGA2 in HCC cells increased upon TGF-ß treatment. Raised HMGA2 levels that occur with alterations in the TGF-ß signaling pathway may reflect an altered activity of E3 ligases, such as PJA1, and potentially contribute to the tumor-promoting roles of TGF-ß signaling. Here, we report that the co-occurrence of genetic alterations in HMGA2 and TGF-ß pathway core genes is implicated in HCC progression, and propose that HMGA2 and PJA1 may be potential novel targets in dysfunctional TGF-ß signaling in HCC.

17.
Cancer Res ; 67(17): 8164-71, 2007 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-17804729

RESUMEN

Resveratrol, a well-established phytoestrogen and chemopreventive agent, has gained much attention among oncologists because it can act as both estrogen receptor agonist and antagonist, depending on dosage and cell context. It is increasingly accepted that steroidal receptor coregulators may also function in the cytoplasmic compartment. Deregulation and altered localization of these coregulators could influence target gene expression and participate in the development of hormone-responsive cancers. Proline-, glutamic acid-, and leucine-rich protein-1 (PELP1), a novel estrogen receptor (ER) coactivator, plays an important role in the genomic and nongenomic actions of ER. Furthermore, recent studies have shown that differential compartmentalization of PELP1 could be crucial in modulating sensitivity to tamoxifen. In this study, we investigated the role of PELP1 in resveratrol-induced autophagy in lung cancer and salivary gland adenocarcinoma cell lines. Resveratrol reversibly inhibited the growth of these cancer cell lines and induced autophagy, as evidenced by microtubule-associated protein 1 light chain 3 (LC3) up-regulation in a time-dependent and 3-methyladenine-sensitive manner. Confocal microscopic analysis showed that resveratrol induced PELP1 accumulation in autophagosomes with green fluorescent protein-LC3. The intermediary molecule involved in PELP1 accumulation in resveratrol-induced autophagosomes is hepatocyte growth factor-regulated tyrosine kinase substrate (HRS), a trafficking molecule that binds to PELP1. These results identify PELP1 for the first time in autophagosomes, implying that both PELP1 and HRS reallocate to autophagosomes in response to resveratrol treatment, which might be important in the process of autophagy in the cancer cells.


Asunto(s)
Autofagia/efectos de los fármacos , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Receptores de Estrógenos/metabolismo , Estilbenos/farmacología , Transactivadores/metabolismo , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Antineoplásicos Fitogénicos/farmacología , Autofagia/fisiología , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Proteínas Co-Represoras , Complejos de Clasificación Endosomal Requeridos para el Transporte , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Proteínas Asociadas a Microtúbulos/metabolismo , Modelos Biológicos , Fosfoproteínas/metabolismo , Transporte de Proteínas/efectos de los fármacos , Resveratrol , Neoplasias de las Glándulas Salivales/genética , Neoplasias de las Glándulas Salivales/metabolismo , Neoplasias de las Glándulas Salivales/patología , Transactivadores/aislamiento & purificación , Factores de Transcripción , Células Tumorales Cultivadas
18.
Clin Cancer Res ; 13(14): 4291-9, 2007 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-17634559

RESUMEN

PURPOSE: Insulin-like growth factor type I receptor (IGF-IR) plays critical roles in epithelial cancer cell development, proliferation, motility, and survival, and new therapeutic agents targeting IGF-IR are in development. Another receptor tyrosine kinase, the epidermal growth factor receptor (EGFR), is an established therapeutic target in head and neck cancer and IGF-IR/EGFR heterodimerization has been reported in other epithelial cancers. The present study was undertaken to determine the effects of anti-IGF-IR therapeutic targeting on cell signaling and cancer cell phenotypes in squamous cell carcinomas of the head and neck (SCCHN). EXPERIMENTAL DESIGN: The therapeutic efficacy of the human anti-IGF-IR antibody IMC-A12 alone and in combination with the EGFR blocking antibody cetuximab (C225) was tested in SCCHN cell lines and in tumor xenografts. RESULTS: IGF-IR was overexpressed in human head and neck cancer cell lines and tumors. Pretreatment of serum-starved 183A or TU159 SCCHN cell lines with A12 (10 microg/mL) blocked IGF-stimulated activation of IGF-IR, insulin receptor substrate (IRS)-1 and IRS-2, mitogen-activated protein kinase, and phosphatidylinositol 3-kinase. A12 induced G(0)-G(1) cell cycle arrest and blocked cell growth, motility, and anchorage-independent growth. Stimulation of head and neck cancer cells with either IGF or EGF resulted in IGF-IR and EGFR heterodimerization, but only IGF caused activating phosphorylation of both receptors. Combined treatment with A12 and the EGFR blocking antibody C225 was more effective at reducing cell proliferation and migration than either agent alone. Finally, TU159 tongue cancer cell xenografts grown in athymic nude mice were treated thrice weekly for 4 weeks with vehicle, A12 (40 mg/kg i.p.), C225 (40 mg/kg i.p.), or both agents (n=8 mice per group; 2 tumors per mouse). Linear regression slope analysis showed significant differences in median tumor volume over time between all three treatment groups and the control group. Complete regression was seen in 31% (A12), 31% (C225), and 44% (A12 + C225) of tumors. CONCLUSION: Here we found the overexpression of IGF-IR, the functional heterodimerization of IGF-IR and EGFR, and effective therapeutic targeting of these receptors in human head and neck cancer xenografts.


Asunto(s)
Neoplasias de Cabeza y Cuello/genética , Receptor IGF Tipo 1/genética , Somatomedinas/farmacología , Animales , Antineoplásicos/toxicidad , Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Línea Celular Tumoral , Dimerización , Receptores ErbB/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias de Cabeza y Cuello/patología , Humanos , Ratones , Ratones Desnudos , Microscopía Confocal , Receptor IGF Tipo 1/efectos de los fármacos , Receptor IGF Tipo 1/metabolismo , Trasplante Heterólogo
19.
Cell Syst ; 7(4): 422-437.e7, 2018 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-30268436

RESUMEN

We present an integromic analysis of gene alterations that modulate transforming growth factor ß (TGF-ß)-Smad-mediated signaling in 9,125 tumor samples across 33 cancer types in The Cancer Genome Atlas (TCGA). Focusing on genes that encode mediators and regulators of TGF-ß signaling, we found at least one genomic alteration (mutation, homozygous deletion, or amplification) in 39% of samples, with highest frequencies in gastrointestinal cancers. We identified mutation hotspots in genes that encode TGF-ß ligands (BMP5), receptors (TGFBR2, AVCR2A, and BMPR2), and Smads (SMAD2 and SMAD4). Alterations in the TGF-ß superfamily correlated positively with expression of metastasis-associated genes and with decreased survival. Correlation analyses showed the contributions of mutation, amplification, deletion, DNA methylation, and miRNA expression to transcriptional activity of TGF-ß signaling in each cancer type. This study provides a broad molecular perspective relevant for future functional and therapeutic studies of the diverse cancer pathways mediated by the TGF-ß superfamily.


Asunto(s)
Tasa de Mutación , Neoplasias/genética , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Proteína Morfogenética Ósea 5/genética , Proteína Morfogenética Ósea 5/metabolismo , Metilación de ADN , Humanos , MicroARNs/genética , Receptor Tipo I de Factor de Crecimiento Transformador beta/genética , Receptor Tipo I de Factor de Crecimiento Transformador beta/metabolismo , Proteínas Smad/genética , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta/genética
20.
Int J Oncol ; 30(3): 743-9, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17273777

RESUMEN

The objective of this study was to assess the effect of pre-analytical processing on proteomic analysis of saliva and to identify salivary biomarkers for potential clinical applications. Saliva samples from five healthy individuals and three head and neck squamous carcinoma (HNSC) patients were initially depleted of major protein constituents. Saliva from healthy subjects was divided and processed by three different methods prior to liquid chromatography and tandem mass spectrometry technique (LC-MS/MS) analysis. The results showed marked differences amongst the methods. The SDS-PAGE separation and in-gel digestion method yielded the highest number of proteins that included the majority of those identified by the other two methods. The in gel-digestion method was used in the LC-MS/MS analysis of saliva from three HNSC patients and the results were compared with those from healthy subjects. Our analysis identified two proteins, alpha-1-B-glycoprotein and complement factor B proteins, to be present in patients but not in normal specimens. Paradoxically, cystatin S, parotid secretory factor, and poly-4-hydrolase beta-subunit proteins were detected in most normal salivas but not in patient specimens. Subsequent analysis of complement factor B by Western blotting showed strong immunoreactive bands of complement factor B in HNSC patients' and negative or weakly positive in normal saliva samples. We conclude that: 1) initial saliva processing affects protein analysis, 2) in-gel digestion followed by LC-MS/MS detects the most saliva proteins, 3) certain proteins are differentially found in patient and normal salivas and 4) a small set of proteins can be targeted for future validation for clinical investigation.


Asunto(s)
Biomarcadores de Tumor/biosíntesis , Carcinoma de Células Escamosas/diagnóstico , Carcinoma de Células Escamosas/metabolismo , Neoplasias de Cabeza y Cuello/diagnóstico , Neoplasias de Cabeza y Cuello/metabolismo , Proteómica/métodos , Saliva/metabolismo , Biomarcadores/metabolismo , Cromatografía Liquida/métodos , Electroforesis en Gel de Poliacrilamida , Femenino , Humanos , Masculino , Tamizaje Masivo/métodos , Espectrometría de Masas/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA