Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cell Rep Med ; 5(4): 101489, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38554705

RESUMEN

Lung adenocarcinoma is a type of cancer that exhibits a wide range of clinical radiological manifestations, from ground-glass opacity (GGO) to pure solid nodules, which vary greatly in terms of their biological characteristics. Our current understanding of this heterogeneity is limited. To address this gap, we analyze 58 lung adenocarcinoma patients via machine learning, single-cell RNA sequencing (scRNA-seq), and whole-exome sequencing, and we identify six lung multicellular ecotypes (LMEs) correlating with distinct radiological patterns and cancer cell states. Notably, GGO-associated neoantigens in early-stage cancers are recognized by CD8+ T cells, indicating an immune-active environment, while solid nodules feature an immune-suppressive LME with exhausted CD8+ T cells, driven by specific stromal cells such as CTHCR1+ fibroblasts. This study also highlights EGFR(L858R) neoantigens in GGO samples, suggesting potential CD8+ T cell activation. Our findings offer valuable insights into lung adenocarcinoma heterogeneity, suggesting avenues for targeted therapies in early-stage disease.


Asunto(s)
Adenocarcinoma del Pulmón , Adenocarcinoma , Neoplasias Pulmonares , Humanos , Neoplasias Pulmonares/genética , Adenocarcinoma/genética , Adenocarcinoma/patología , Linfocitos T CD8-positivos/patología , Ecotipo , Estudios Retrospectivos
2.
Signal Transduct Target Ther ; 9(1): 65, 2024 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-38461173

RESUMEN

Despite epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKI) have shown remarkable efficacy in patients with EGFR-mutant non-small cell lung cancer (NSCLC), acquired resistance inevitably develops, limiting clinical efficacy. We found that TET2 was poly-ubiquitinated by E3 ligase CUL7FBXW11 and degraded in EGFR-TKI resistant NSCLC cells. Genetic perturbation of TET2 rendered parental cells more tolerant to TKI treatment. TET2 was stabilized by MEK1 phosphorylation at Ser 1107, while MEK1 inactivation promoted its proteasome degradation by enhancing the recruitment of CUL7FBXW11. Loss of TET2 resulted in the upregulation of TNF/NF-κB signaling that confers the EGFR-TKI resistance. Genetic or pharmacological inhibition of NF-κB attenuate the TKI resistance both in vitro and in vivo. Our findings exemplified how a cell growth controlling kinase MEK1 leveraged the epigenetic homeostasis by regulating TET2, and demonstrated an alternative path of non-mutational acquired EGFR-TKI resistance modulated by TET2 deficiency. Therefore, combined strategy exploiting EGFR-TKI and inhibitors of TET2/NF-κB axis holds therapeutic potential for treating NSCLC patients who suffered from this resistance.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Dioxigenasas , Resistencia a Antineoplásicos , Neoplasias Pulmonares , Humanos , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Dioxigenasas/genética , Proteínas de Unión al ADN/genética , Receptores ErbB , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Mutación , FN-kappa B/genética , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , /uso terapéutico , Resistencia a Antineoplásicos/genética
4.
J Hepatol ; 74(4): 907-918, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33031845

RESUMEN

BACKGROUND & AIMS: There are currently limited therapeutic options for hepatocellular carcinoma (HCC), particularly when it is diagnosed at advanced stages. Herein, we examined the pathophysiological role of ROS1 and assessed the utility of ROS1-targeted therapy for the treatment of HCC. METHODS: Recombinant ribonucleases (RNases) were purified, and the ligand-receptor relationship between RNase7 and ROS1 was validated in HCC cell lines by Duolink, immunofluorescence, and immunoprecipitation assays. Potential interacting residues between ROS1 and RNase7 were predicted using a protein-protein docking approach. The oncogenic function of RNase7 was analyzed by cell proliferation, migration and invasion assays, and a xenograft mouse model. The efficacy of anti-ROS1 inhibitor treatment was evaluated in patient-derived xenograft (PDX) and orthotopic models. Two independent patient cohorts were analyzed to evaluate the pathological relevance of RNase7/ROS1. RESULTS: RNase7 associated with ROS1's N3-P2 domain and promoted ROS1-mediated oncogenic transformation. Patients with HCC exhibited elevated plasma RNase7 levels compared with healthy individuals. High ROS1 and RNase7 expression were strongly associated with poor prognosis in patients with HCC. In both HCC PDX and orthotopic mouse models, ROS1 inhibitor treatment markedly suppressed RNase7-induced tumorigenesis, leading to decreased plasma RNase7 levels and tumor shrinkage in mice. CONCLUSIONS: RNase7 serves as a high-affinity ligand for ROS1. Plasma RNase7 could be used as a biomarker to identify patients with HCC who may benefit from anti-ROS1 treatment. LAY SUMMARY: Receptor tyrosine kinases are known to be involved in tumorigenesis and have been targeted therapeutically for a number of cancers, including hepatocellular carcinoma. ROS1 is the only such receptor with kinase activity whose ligand has not been identified. Herein, we show that RNase7 acts as a ligand to activate ROS1 signaling. This has important pathophysiological and therapeutic implications. Anti-ROS1 inhibitors could be used to treatment patients with hepatocellular carcinoma and high RNase7 levels.


Asunto(s)
Carcinogénesis , Carcinoma Hepatocelular , Crizotinib/farmacología , Neoplasias Hepáticas , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Ribonucleasas/metabolismo , Animales , Biomarcadores de Tumor/metabolismo , Carcinogénesis/efectos de los fármacos , Carcinogénesis/metabolismo , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/metabolismo , Ensayos de Migración Celular/métodos , Proliferación Celular/efectos de los fármacos , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ligandos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/metabolismo , Ratones , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto
5.
J Clin Invest ; 129(8): 3324-3338, 2019 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-31305264

RESUMEN

Glycosylation of immune receptors and ligands, such as T cell receptor and coinhibitory molecules, regulates immune signaling activation and immune surveillance. However, how oncogenic signaling initiates glycosylation of coinhibitory molecules to induce immunosuppression remains unclear. Here we show that IL-6-activated JAK1 phosphorylates programmed death-ligand 1 (PD-L1) Tyr112, which recruits the endoplasmic reticulum-associated N-glycosyltransferase STT3A to catalyze PD-L1 glycosylation and maintain PD-L1 stability. Targeting of IL-6 by IL-6 antibody induced synergistic T cell killing effects when combined with anti-T cell immunoglobulin mucin-3 (anti-Tim-3) therapy in animal models. A positive correlation between IL-6 and PD-L1 expression was also observed in hepatocellular carcinoma patient tumor tissues. These results identify a mechanism regulating PD-L1 glycosylation initiation and suggest the combination of anti-IL-6 and anti-Tim-3 as an effective marker-guided therapeutic strategy.


Asunto(s)
Antígeno B7-H1/inmunología , Interleucina-6/inmunología , Janus Quinasa 1/inmunología , Proteínas de Neoplasias/inmunología , Neoplasias Experimentales/inmunología , Transducción de Señal/inmunología , Escape del Tumor , Animales , Línea Celular Tumoral , Humanos , Masculino , Ratones , Neoplasias Experimentales/patología , Neoplasias Experimentales/terapia , Estabilidad Proteica
8.
Cancer Cell ; 34(6): 954-969.e4, 2018 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-30537515

RESUMEN

Multiple mechanisms of resistance to epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) have been identified in EGFR-mutant non-small cell lung cancer (NSCLC); however, recurrent resistance to EGFR TKIs due to the heterogeneous mechanisms underlying resistance within a single patient remains a major challenge in the clinic. Here, we report a role of nuclear protein kinase Cδ (PKCδ) as a common axis across multiple known TKI-resistance mechanisms. Specifically, we demonstrate that TKI-inactivated EGFR dimerizes with other membrane receptors implicated in TKI resistance to promote PKCδ nuclear translocation. Moreover, the level of nuclear PKCδ is associated with TKI response in patients. The combined inhibition of PKCδ and EGFR induces marked regression of resistant NSCLC tumors with EGFR mutations.


Asunto(s)
Resistencia a Antineoplásicos/efectos de los fármacos , Proteína Quinasa C-delta/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Núcleo Celular/genética , Núcleo Celular/metabolismo , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Receptores ErbB/metabolismo , Femenino , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Masculino , Microscopía Confocal , Persona de Mediana Edad , Terapia Molecular Dirigida/métodos , Mutación , Proteína Quinasa C-delta/metabolismo , Interferencia de ARN
9.
Am J Cancer Res ; 8(9): 1837-1846, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30323975

RESUMEN

Triple-negative breast cancer (TNBC), the most difficult-to-treat breast cancer subtype, lacks well-defined molecular targets. TNBC has increased programmed death-ligand 1 (PD-L1) expression, and its immunosuppressive nature makes it suitable for immune checkpoint blockade therapy. However, the response rate of TNBC to anti-PD-L1 or anti-programmed cell death protein 1 (PD-1) therapy remains unsatisfactory, as only 10-20% of TNBC patients have a partial response. Glycosylated PD-L1, the functional form of PD-L1, is required for PD-L1-PD-1 interaction. TNBC cells have significantly higher levels of glycosylated PD-L1 than non-TNBC cells do. In a screening of glucose analogs to block PD-L1 glycosylation, we found that 2-deoxyglucose (2-DG) can act as a glucose analog to decrease PD-L1 glycosylation. Because PARP inhibition upregulates PD-L1, 2-DG reduced PARP inhibition-mediated expression of glycosylated PD-L1. The combination of PARP inhibition and 2-DG had potent anti-tumor activity. Together, our results provide a strong rationale for investigating the targeting of PD-L1 glycosylation in TNBC further.

10.
Cell Discov ; 2: 16005, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27462452

RESUMEN

Population-based and case-control studies in different ethnicities have linked a polymorphism, C825T, in exon 10 of GNB3 gene to hypertension and several additional diseases. The 825T allele is associated with alternative splicing and results in a shortened Gß3 protein, referred to as Gß3s, which loses 41 amino acids encompassing one WD40 repeat domain. The mechanism of how Gß3 C825T polymorphism is associated with hypertension has remained unclear, but an impairment of its canonical function in G-protein-coupled receptor signaling has been ruled out. Here, we report that Gß3, like other Gß proteins, binds to DDB1 and assembles a DDB1-CUL4A-ROC1 E3 ubiquitin ligase (CRL4A(Gß3)) to target GRK2 ubiquitination. The loss of the 41 amino-acid residues disrupts the Gß3-DDB1 binding and impairs the function of Gß3s to ubiquitinate GRK2. GRK2 ubiquitination levels were decreased and protein levels were accumulated in the blood samples of Gß3 825T allele carriers. Deletion of Cul4a in mice resulted in systolic pressure increased and weakened heart function in male mice that can be partially rescued by the deletion of one Grk2 allele. These results reveal a mechanism explaining the link between Gß3 C825T polymorphism and hypertension.

11.
Mol Cell ; 58(5): 794-803, 2015 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-25982117

RESUMEN

G protein-coupled receptors (GPCRs) comprise the largest family of cell surface receptors, regulate a wide range of physiological processes, and are the major targets of pharmaceutical drugs. Canonical signaling from GPCRs is relayed to intracellular effector proteins by trimeric G proteins, composed of α, ß, and γ subunits (Gαßγ). Here, we report that G protein ß subunits (Gß) bind to DDB1 and that Gß2 targets GRK2 for ubiquitylation by the DDB1-CUL4A-ROC1 ubiquitin ligase. Activation of GPCR results in PKA-mediated phosphorylation of DDB1 at Ser645 and its dissociation from Gß2, leading to increase of GRK2 protein. Deletion of Cul4a results in cardiac hypertrophy in male mice that can be partially rescued by the deletion of one Grk2 allele. These results reveal a non-canonical function of the Gß protein as a ubiquitin ligase component and a mechanism of feedback regulation of GPCR signaling.


Asunto(s)
Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Subunidades beta de la Proteína de Unión al GTP/fisiología , Proteínas Nucleares/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Animales , Proteínas de Unión al ADN/metabolismo , Femenino , Células HEK293 , Humanos , Masculino , Ratones Noqueados , Estabilidad Proteica , Proteolisis , Ratas , Ratas Wistar , Transducción de Señal
12.
J Biol Chem ; 287(31): 26245-53, 2012 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-22692215

RESUMEN

The Hippo tumor suppressor pathway plays a major role in development and organ size control, and its dysregulation contributes to tumorigenesis. TAZ (transcriptional co-activator with PDZ-binding motif; also known as WWTR1) is a transcription co-activator acting downstream of the Hippo pathway, and increased TAZ protein levels have been associated with human cancers, such as breast cancer. Previous studies have shown that TAZ is inhibited by large tumor suppressor (LATS)-dependent phosphorylation, leading to cytoplasmic retention and ubiquitin-dependent degradation. The LATS kinase, a core component of the Hippo pathway, phosphorylates the C-terminal phosphodegron in TAZ to promote its degradation. In this study, we have found that the N-terminal phosphodegron of TAZ also plays a role in TAZ protein level regulation, particularly in response to different status of cellular PI3K signaling. GSK3, which can be inhibited by high PI3K via AKT-dependent inhibitory phosphorylation, phosphorylates the N-terminal phosphodegron in TAZ, and the phosphorylated TAZ binds to ß-TrCP subunit of the SCF(ß-TrCP) E3 ubiquitin ligase, thereby leading to TAZ ubiquitylation and degradation. We observed that the TAZ protein level is elevated in tumor cells with high PI3K signaling, such as in PTEN mutant cancer cells. This study provides a novel mechanism of TAZ regulation and suggests a role of TAZ in modulating tissue growth and tumor development in response to PI3K signaling.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Procesamiento Proteico-Postraduccional , Proteolisis , Proteínas Ligasas SKP Cullina F-box/metabolismo , Animales , Proliferación Celular , Cromonas/farmacología , Glucógeno Sintasa Quinasa 3/metabolismo , Células HEK293 , Células HeLa , Humanos , Ratones , Morfolinas/farmacología , Células 3T3 NIH , Fosforilación , Estabilidad Proteica , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Transactivadores , Factores de Transcripción , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ
13.
Mol Cell ; 42(6): 719-30, 2011 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-21700219

RESUMEN

Most tumor cells take up more glucose than normal cells but metabolize glucose via glycolysis even in the presence of normal levels of oxygen, a phenomenon known as the Warburg effect. Tumor cells commonly express the embryonic M2 isoform of pyruvate kinase (PKM2) that may contribute to the metabolism shift from oxidative phosphorylation to aerobic glycolysis and tumorigenesis. Here we show that PKM2 is acetylated on lysine 305 and that this acetylation is stimulated by high glucose concentration. PKM2 K305 acetylation decreases PKM2 enzyme activity and promotes its lysosomal-dependent degradation via chaperone-mediated autophagy (CMA). Acetylation increases PKM2 interaction with HSC70, a chaperone for CMA, and association with lysosomes. Ectopic expression of an acetylation mimetic K305Q mutant accumulates glycolytic intermediates and promotes cell proliferation and tumor growth. These results reveal an acetylation regulation of pyruvate kinase and the link between lysine acetylation and CMA.


Asunto(s)
Autofagia , Chaperonas Moleculares/metabolismo , Neoplasias de la Próstata/metabolismo , Piruvato Quinasa/metabolismo , Factores de Transcripción p300-CBP/metabolismo , Acetilación , Animales , Proliferación Celular , Relación Dosis-Respuesta a Droga , Activación Enzimática , Glucosa/química , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Lisina/metabolismo , Lisosomas/metabolismo , Masculino , Ratones , Ratones Desnudos , Mutación , Neoplasias de la Próstata/patología , Piruvato Quinasa/genética , Relación Estructura-Actividad , Ensayos Antitumor por Modelo de Xenoinjerto
14.
J Biol Chem ; 285(48): 37159-69, 2010 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-20858893

RESUMEN

The TAZ transcription co-activator promotes cell proliferation and epithelial-mesenchymal transition. TAZ is inhibited by the Hippo tumor suppressor pathway, which promotes TAZ cytoplasmic localization by phosphorylation. We report here that TAZ protein stability is controlled by a phosphodegron recognized by the F-box protein ß-TrCP and ubiquitylated by the SCF/CRL1(ß-TrCP) E3 ligase. The interaction between TAZ and ß-TrCP is regulated by the Hippo pathway. Phosphorylation of a phosphodegron in TAZ by LATS primes it for further phosphorylation by CK1ε and subsequent binding by ß-TrCP. Therefore, the Hippo pathway negatively regulates TAZ function by both limiting its nuclear accumulation and promoting its degradation. The phosphodegron-mediated TAZ degradation plays an important role in negatively regulating TAZ biological functions.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Ligasas SKP Cullina F-box/metabolismo , Transducción de Señal , Animales , Quinasa de la Caseína I/genética , Quinasa de la Caseína I/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular/química , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Células 3T3 NIH , Fosforilación , Unión Proteica , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Estabilidad Proteica , Proteínas Ligasas SKP Cullina F-box/genética , Transactivadores , Factores de Transcripción , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ
15.
Science ; 324(5924): 261-5, 2009 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-19359588

RESUMEN

Heterozygous mutations in the gene encoding isocitrate dehydrogenase-1 (IDH1) occur in certain human brain tumors, but their mechanistic role in tumor development is unknown. We have shown that tumor-derived IDH1 mutations impair the enzyme's affinity for its substrate and dominantly inhibit wild-type IDH1 activity through the formation of catalytically inactive heterodimers. Forced expression of mutant IDH1 in cultured cells reduces formation of the enzyme product, alpha-ketoglutarate (alpha-KG), and increases the levels of hypoxia-inducible factor subunit HIF-1alpha, a transcription factor that facilitates tumor growth when oxygen is low and whose stability is regulated by alpha-KG. The rise in HIF-1alpha levels was reversible by an alpha-KG derivative. HIF-1alpha levels were higher in human gliomas harboring an IDH1 mutation than in tumors without a mutation. Thus, IDH1 appears to function as a tumor suppressor that, when mutationally inactivated, contributes to tumorigenesis in part through induction of the HIF-1 pathway.


Asunto(s)
Neoplasias Encefálicas/genética , Glioma/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Isocitrato Deshidrogenasa/genética , Isocitrato Deshidrogenasa/metabolismo , Adolescente , Adulto , Anciano , Astrocitoma/genética , Astrocitoma/metabolismo , Biocatálisis , Neoplasias Encefálicas/metabolismo , Línea Celular , Niño , Femenino , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , Glioblastoma/genética , Glioblastoma/metabolismo , Glioma/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Isocitrato Deshidrogenasa/química , Ácidos Cetoglutáricos/metabolismo , Masculino , Persona de Mediana Edad , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Oligodendroglioma/genética , Oligodendroglioma/metabolismo , Oxalatos/farmacología , Multimerización de Proteína
16.
J Biol Chem ; 284(20): 13355-13362, 2009 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-19324877

RESUMEN

The TAZ transcription co-activator has been shown to promote cell proliferation and to induce epithelial-mesenchymal transition. Recently we have demonstrated that TAZ is phosphorylated and inhibited by the Hippo tumor suppressor pathway, which is altered in human cancer. The mechanism of TAZ-mediated transcription is unclear. We demonstrate here that TEAD is a key downstream transcription factor mediating the function of TAZ. Disruption of TEAD-TAZ binding or silencing of TEAD expression blocked the function of TAZ to promote cell proliferation and to induce epithelial-mesenchymal transition, demonstrating TEAD as a key downstream effector of TAZ. We also identified CTGF, a gene that regulates cell adhesion, proliferation, and migration, as a direct target of TAZ and TEAD. Our study establishes a functional partnership between TAZ and TEAD under negative regulation by the Hippo signaling pathway.


Asunto(s)
Movimiento Celular/fisiología , Proliferación Celular , Epitelio/metabolismo , Mesodermo/metabolismo , Transducción de Señal/fisiología , Factores de Transcripción/metabolismo , Aciltransferasas , Adhesión Celular/fisiología , Línea Celular , Factor de Crecimiento del Tejido Conjuntivo/genética , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Silenciador del Gen , Humanos , Fosforilación/fisiología , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
17.
Mol Cell Biol ; 28(7): 2426-36, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18227151

RESUMEN

TAZ is a WW domain containing a transcription coactivator that modulates mesenchymal differentiation and development of multiple organs. In this study, we show that TAZ is phosphorylated by the Lats tumor suppressor kinase, a key component of the Hippo pathway, whose alterations result in organ and tissue hypertrophy in Drosophila and contribute to tumorigenesis in humans. Lats phosphorylates TAZ on several serine residues in the conserved HXRXXS motif and creates 14-3-3 binding sites, leading to cytoplasmic retention and functional inactivation of TAZ. Ectopic expression of TAZ stimulates cell proliferation, reduces cell contact inhibition, and promotes epithelial-mesenchymal transition (EMT). Elimination of the Lats phosphorylation sites results in a constitutively active TAZ, enhancing the activity of TAZ in promoting cell proliferation and EMT. Our results elucidate a molecular mechanism for TAZ regulation and indicate a potential function of TAZ as an important target of the Hippo pathway in regulating cell proliferation tumorigenesis.


Asunto(s)
Procesamiento Proteico-Postraduccional/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas/fisiología , Factores de Transcripción/fisiología , Proteínas 14-3-3/fisiología , Aciltransferasas , Secuencias de Aminoácidos , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/fisiología , División Celular , Línea Celular Tumoral , Movimiento Celular , Transdiferenciación Celular/fisiología , Transformación Celular Neoplásica , Secuencia Conservada , Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiología , Células Epiteliales/citología , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/fisiología , Mesodermo/citología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/fisiología , Proteínas Nucleares/fisiología , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Proteínas/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/fisiología , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/fisiología , Proteínas Recombinantes de Fusión/fisiología , Serina-Treonina Quinasa 3 , Factores de Transcripción/genética , Transcripción Genética , Transferasas (Grupos de Otros Fosfatos Sustitutos)/genética , Transferasas (Grupos de Otros Fosfatos Sustitutos)/fisiología , Tirosina Quinasa c-Mer
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...