Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nat Cell Biol ; 25(5): 765-777, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37095322

RESUMEN

PBRM1 encodes an accessory subunit of the PBAF SWI/SNF chromatin remodeller, and the inactivation of PBRM1 is a frequent event in kidney cancer. However, the impact of PBRM1 loss on chromatin remodelling is not well examined. Here we show that, in VHL-deficient renal tumours, PBRM1 deficiency results in ectopic PBAF complexes that localize to de novo genomic loci, activating the pro-tumourigenic NF-κB pathway. PBRM1-deficient PBAF complexes retain the association between SMARCA4 and ARID2, but have loosely tethered BRD7. The PBAF complexes redistribute from promoter proximal regions to distal enhancers containing NF-κB motifs, heightening NF-κB activity in PBRM1-deficient models and clinical samples. The ATPase function of SMARCA4 maintains chromatin occupancy of pre-existing and newly acquired RELA specific to PBRM1 loss, activating downstream target gene expression. Proteasome inhibitor bortezomib abrogates RELA occupancy, suppresses NF-κB activation and delays growth of PBRM1-deficient tumours. In conclusion, PBRM1 safeguards the chromatin by repressing aberrant liberation of pro-tumourigenic NF-κB target genes by residual PBRM1-deficient PBAF complexes.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Humanos , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/metabolismo , Cromatina/genética , Proteínas Cromosómicas no Histona/genética , ADN Helicasas/genética , Proteínas de Unión al ADN/genética , Genómica , Neoplasias Renales/metabolismo , FN-kappa B/genética , Proteínas Nucleares/genética , Factores de Transcripción/genética
2.
Theranostics ; 10(6): 2727-2743, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32194831

RESUMEN

Oncogene-addicted cancers are predominantly driven by specific oncogenic pathways and display initial exquisite sensitivity to designer therapies, but eventually become refractory to treatments. Clear understanding of lung tumorigenic mechanisms is essential for improved therapies. Methods: Lysosomes were analyzed in EGFR-WT and mutant cells and corresponding patient samples using immunofluorescence or immunohistochemistry and immunoblotting. Microtubule organization and dynamics were studied using immunofluorescence analyses. Also, we have validated our findings in a transgenic mouse model that contain EGFR-TKI resistant mutations. Results: We herein describe a novel mechanism that a mutated kinase disrupts the microtubule organization and results in a defective endosomal/lysosomal pathway. This prevents the efficient degradation of phosphorylated proteins that become trapped within the endosomes and continue to signal, therefore amplifying downstream proliferative and survival pathways. Phenotypically, a distinctive subcellular appearance of LAMP1 secondary to microtubule dysfunction in cells expressing EGFR kinase mutants is seen, and this may have potential diagnostic applications for the detection of such mutants. We demonstrate that lysosomal-inhibitors re-sensitize resistant cells to EGFR tyrosine-kinase inhibitors (TKIs). Identifying the endosome-lysosome pathway and microtubule dysfunction as a mechanism of resistance allows to pharmacologically intervene on this pathway. Conclusions: We find that the combination of microtubule stabilizing agent and lysosome inhibitor could reduce the tumor progression in EGFR TKI resistant mouse models of lung cancer.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Resistencia a Antineoplásicos/efectos de los fármacos , Receptores ErbB/antagonistas & inhibidores , Neoplasias Pulmonares/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Animales , Células COS , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Línea Celular Tumoral , Chlorocebus aethiops , Receptores ErbB/metabolismo , Humanos , Neoplasias Pulmonares/metabolismo , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Ratones , Ratones Transgénicos , Microtúbulos/efectos de los fármacos , Microtúbulos/metabolismo
3.
Cancer Res ; 77(18): 4921-4933, 2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-28720576

RESUMEN

The core LATS kinases of the Hippo tumor suppressor pathway phosphorylate and inhibit the downstream transcriptional co-activators YAP and TAZ, which are implicated in various cancers. Recent studies have identified various E3 ubiquitin ligases that negatively regulate the Hippo pathway via ubiquitination, yet few deubiquitinating enzymes (DUB) have been implicated. In this study, we report the DUB USP9X is an important regulator of the core kinases of this pathway. USP9X interacted strongly with LATS kinase and to a lesser extent with WW45, KIBRA, and Angiomotin, and LATS co-migrated exclusively with USP9X during gel filtration chromatography analysis. Knockdown of USP9X significantly downregulated and destabilized LATS and resulted in enhanced nuclear translocation of YAP and TAZ, accompanied with activation of their target genes. In the absence of USP9X, cells exhibited an epithelial-to-mesenchymal transition phenotype, acquired anchorage-independent growth in soft agar, and led to enlarged, disorganized, three-dimensional acini. YAP/TAZ target gene activation in response to USP9X knockdown was suppressed by knockdown of YAP, TAZ, and TEAD2. Deletion of USP9X in mouse embryonic fibroblasts resulted in significant downregulation of LATS. Furthermore, USP9X protein expression correlated positively with LATS but negatively with YAP/TAZ in pancreatic cancer tissues as well as pancreatic and breast cancer cell lines. Overall, these results strongly indicate that USP9X potentiates LATS kinase to suppress tumor growth. Cancer Res; 77(18); 4921-33. ©2017 AACR.


Asunto(s)
Neoplasias de la Mama/patología , Endopeptidasas/fisiología , Neoplasias Pancreáticas/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Angiomotinas , Animales , Apoptosis , Biomarcadores de Tumor , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Proliferación Celular , Células Cultivadas , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Estudios de Seguimiento , Vía de Señalización Hippo , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Proteínas de Microfilamentos , Estadificación de Neoplasias , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosforilación , Pronóstico , Proteínas Serina-Treonina Quinasas/genética , Transducción de Señal , Tasa de Supervivencia , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Ubiquitina Tiolesterasa/genética , Ubiquitinación , Proteínas Señalizadoras YAP
4.
Nat Commun ; 6: 6184, 2015 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-25630468

RESUMEN

Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related deaths globally. The identity and role of cell surface molecules driving complex biological events leading to HCC progression are poorly understood, hence representing major lacunae in HCC therapies. Here, combining SILAC quantitative proteomics and biochemical approaches, we uncover a critical oncogenic role of Agrin, which is overexpressed and secreted in HCC. Agrin enhances cellular proliferation, migration and oncogenic signalling. Mechanistically, Agrin's extracellular matrix sensor activity provides oncogenic cues to regulate Arp2/3-dependent ruffling, invadopodia formation and epithelial-mesenchymal transition through sustained focal adhesion integrity that drives liver tumorigenesis. Furthermore, Agrin signalling through Lrp4-muscle-specific tyrosine kinase (MuSK) forms a critical oncogenic axis. Importantly, antibodies targeting Agrin reduced oncogenic signalling and tumour growth in vivo. Together, we demonstrate that Agrin is frequently upregulated and important for oncogenic property of HCC, and is an attractive target for antibody therapy.


Asunto(s)
Agrina/metabolismo , Carcinoma Hepatocelular/metabolismo , Adhesiones Focales/metabolismo , Neoplasias Hepáticas/metabolismo , Oncogenes , Animales , Anticuerpos Bloqueadores/farmacología , Apoptosis/efectos de los fármacos , Carcinogénesis/efectos de los fármacos , Carcinogénesis/metabolismo , Carcinogénesis/patología , Carcinoma Hepatocelular/patología , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Endocitosis/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Adhesiones Focales/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Integrinas/metabolismo , Marcaje Isotópico , Proteínas Relacionadas con Receptor de LDL/metabolismo , Neoplasias Hepáticas/patología , Microdominios de Membrana/efectos de los fármacos , Microdominios de Membrana/metabolismo , Ratones Desnudos , Invasividad Neoplásica , Proteínas de Neoplasias/metabolismo , Seudópodos/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores Colinérgicos/metabolismo , Ensayo de Tumor de Célula Madre , Ensayos Antitumor por Modelo de Xenoinjerto
5.
J Cell Sci ; 127(Pt 13): 2825-39, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24806965

RESUMEN

Searching and evaluating the Human Protein Atlas for transmembrane proteins enabled us to identify an integral membrane protein, TMEM115, that is enriched in the Golgi complex. Biochemical and cell biological analysis suggested that TMEM115 has four candidate transmembrane domains located in the N-terminal region. Both the N- and C-terminal domains are oriented towards the cytoplasm. Immunofluorescence analysis supports that TMEM115 is enriched in the Golgi cisternae. Functionally, TMEM115 knockdown or overexpression delays Brefeldin-A-induced Golgi-to-ER retrograde transport, phenocopying cells with mutations or silencing of the conserved oligomeric Golgi (COG) complex. Co-immunoprecipitation and in vitro binding experiments reveals that TMEM115 interacts with the COG complex, and might self-interact to form dimers or oligomers. A short region (residues 206-229) immediately to the C-terminal side of the fourth transmembrane domain is both necessary and sufficient for Golgi targeting. Knockdown of TMEM115 also reduces the binding of the lectins peanut agglutinin (PNA) and Helix pomatia agglutinin (HPA), suggesting an altered O-linked glycosylation profile. These results establish that TMEM115 is an integral membrane protein of the Golgi stack regulating Golgi-to-ER retrograde transport and is likely to be part of the machinery of the COG complex.


Asunto(s)
Aparato de Golgi/metabolismo , Proteínas de la Membrana/metabolismo , Animales , Células HeLa , Humanos , Ratones , Unión Proteica , Transporte de Proteínas , Proteínas de Transporte Vesicular/metabolismo
6.
J Cell Biol ; 192(1): 121-35, 2011 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-21220512

RESUMEN

Oxysterol-binding protein (OSBP) and its related proteins (ORPs) constitute a large and evolutionarily conserved family of lipid-binding proteins that target organelle membranes to mediate sterol signaling and/or transport. Here we characterize ORP5, a tail-anchored ORP protein that localizes to the endoplasmic reticulum. Knocking down ORP5 causes cholesterol accumulation in late endosomes and lysosomes, which is reminiscent of the cholesterol trafficking defect in Niemann Pick C (NPC) fibroblasts. Cholesterol appears to accumulate in the limiting membranes of endosomal compartments in ORP5-depleted cells, whereas depletion of NPC1 or both ORP5 and NPC1 results in luminal accumulation of cholesterol. Moreover, trans-Golgi resident proteins mislocalize to endosomal compartments upon ORP5 depletion, which depends on a functional NPC1. Our results establish the first link between NPC1 and a cytoplasmic sterol carrier, and suggest that ORP5 may cooperate with NPC1 to mediate the exit of cholesterol from endosomes/lysosomes.


Asunto(s)
Colesterol/metabolismo , Endosomas/metabolismo , Receptores de Esteroides/metabolismo , Transporte Biológico , Proteínas Portadoras/metabolismo , Retículo Endoplásmico/metabolismo , Esterificación , Técnicas de Silenciamiento del Gen , Aparato de Golgi/metabolismo , Células HeLa , Humanos , Inmunoprecipitación , Membranas Intracelulares/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Lipoproteínas LDL/metabolismo , Lisosomas/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteína Niemann-Pick C1 , Unión Proteica , Estructura Terciaria de Proteína , ARN Interferente Pequeño/metabolismo , Receptores de Esteroides/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Toxina Shiga/metabolismo
7.
J Cell Biol ; 190(3): 331-45, 2010 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-20679433

RESUMEN

Coat protein II (COPII)-mediated export from the endoplasmic reticulum (ER) involves sequential recruitment of COPII complex components, including the Sar1 GTPase, the Sec23/Sec24 subcomplex, and the Sec13/Sec31 subcomplex. p125A was originally identified as a Sec23A-interacting protein. Here we demonstrate that p125A also interacts with the C-terminal region of Sec31A. The Sec31A-interacting domain of p125A is between residues 260-600, and is therefore a distinct domain from that required for interaction with Sec23A. Gel filtration and immunodepletion studies suggest that the majority of cytosolic p125A exists as a ternary complex with the Sec13/Sec31A subcomplex, suggesting that Sec 13, Sec31A, and p125A exist in the cytosol primarily as preassembled Sec13/Sec31A/p125A heterohexamers. Golgi morphology and protein export from the ER were affected in p125A-silenced cells. Our results suggest that p125A is part of the Sec13/Sec31A subcomplex and facilitates ER export in mammalian cells.


Asunto(s)
Vesículas Cubiertas por Proteínas de Revestimiento/química , Proteínas Portadoras/metabolismo , Retículo Endoplásmico/metabolismo , Aparato de Golgi/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Vesículas Cubiertas por Proteínas de Revestimiento/metabolismo , Línea Celular , Citosol/metabolismo , Células HeLa , Humanos , Proteínas de Unión al ARN
8.
Mol Cell ; 37(3): 383-95, 2010 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-20159557

RESUMEN

While most SNAREs are permanently anchored to membranes by their transmembrane domains, the dually lipidated SNARE Ykt6 is found both on intracellular membranes and in the cytosol. The cytosolic Ykt6 is inactive due to the autoinhibition of the SNARE core by its longin domain, although the molecular basis of this inhibition is unknown. Here, we demonstrate that unlipidated Ykt6 adopts multiple conformations, with a small population in the closed state. The structure of Ykt6 in complex with a fatty acid suggests that, upon farnesylation, the Ykt6 SNARE core forms four alpha helices that wrap around the longin domain, forming a dominantly closed conformation. The fatty acid, buried in a hydrophobic groove formed between the longin domain and its SNARE core, is essential for maintaining the autoinhibited conformation of Ykt6. Our study reveals that the posttranslationally attached farnesyl group can actively regulate Ykt6 fusion activity in addition to its anticipated membrane-anchoring role.


Asunto(s)
Proteínas R-SNARE/química , Secuencia de Aminoácidos , Animales , Cristalografía por Rayos X , Citosol/metabolismo , Células HeLa , Humanos , Datos de Secuencia Molecular , Resonancia Magnética Nuclear Biomolecular , Fosforilcolina/análogos & derivados , Fosforilcolina/metabolismo , Prenilación , Estructura Terciaria de Proteína , Proteínas R-SNARE/metabolismo , Proteínas R-SNARE/fisiología , Ratas , Alineación de Secuencia
9.
Biochim Biophys Acta ; 1744(3): 293-303, 2005 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-15979503

RESUMEN

First discovered by genetic analysis of yeast secretion mutants, the evolutionarily conserved vesicular coat protein II (COPII) complex is responsible for membrane transport from the endoplasmic reticulum (ER) to the Golgi apparatus. In recent years, extensive efforts in structural, morphological, genetic and molecular analysis have greatly enhanced our understanding of the structural and molecular basis of COPII subunit assembly and selective cargo packaging during ER export. Very recent data have also indicated that a more "classical" picture of vesicle formation from ER exit sites (ERES) followed by their transport to the Golgi is far from accurate. Proteins modulating the function of COPII have also emerged in recent analysis. They either affect COPII-based cargo selection, the formation of vesicle/transport carrier, or subsequent targeting of the transport carrier. Together, elucidation of COPII-mediated ER export has painted a fascinating picture of molecular complexity for an essential process in all eukaryotic cells.


Asunto(s)
Vesículas Cubiertas por Proteínas de Revestimiento/metabolismo , Retículo Endoplásmico/fisiología , Aparato de Golgi/fisiología , Animales , Vesículas Cubiertas por Proteínas de Revestimiento/química , Vesículas Cubiertas por Proteínas de Revestimiento/ultraestructura , Aparato de Golgi/ultraestructura , Transporte de Proteínas , Ratas , Levaduras/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...