Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Death Dis ; 14(1): 60, 2023 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-36702832

RESUMEN

The spindle assembly checkpoint (SAC) is an essential mechanism that ensures the accurate chromosome segregation during mitosis, thus preventing genomic instability. Deubiquitinases have emerged as key regulators of the SAC, mainly by determining the fate of proteins during cell cycle progression. Here, we identify USP49 deubiquitinase as a novel regulator of the spindle checkpoint. We show that loss of USP49 in different cancer cell lines impairs proliferation and increases aneuploidy. In addition, USP49-depleted cells overcome the arrest induced by the SAC in the presence of nocodazole. Finally, we report new binding partners of USP49, including ribophorin 1, USP44, and different centrins.


Asunto(s)
Puntos de Control de la Fase M del Ciclo Celular , Huso Acromático , Humanos , Huso Acromático/metabolismo , Aneuploidia , Mitosis , Enzimas Desubicuitinizantes/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Ubiquitina Tiolesterasa/genética , Ubiquitina Tiolesterasa/metabolismo
2.
Drug Des Devel Ther ; 15: 3997-4009, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34588764

RESUMEN

Non-alcoholic steatohepatitis (NASH) is a progressive form of non-alcoholic fatty liver disease (NAFLD), characterized by chronic inflammation and accumulation of fat in liver tissue. Affecting estimated 35 million people globally, NASH is the most common chronic liver condition in Western populations, and with patient numbers growing rapidly, the market for NASH therapy is projected to rise to $27.2 B in 2029. Despite this clinical need and attractive commercial opportunity, there are no Food and Drug Administration (FDA)-approved therapies specifically for this disease. Many have tried and unfortunately failed to find a drug, or drug combination, capable of unravelling the complexities of this metabolic condition. At the time of writing this review, only Zydus Cadila's new drug application for Saroglitazar had been approved (2020) for NASH therapy in India. However, it is hoped that this dearth of therapy options will improve as several drug candidates progress through late-stage clinical development. Obeticholic acid (Intercept Pharmaceuticals), Cenicriviroc (Allergan), Aramchol (Galmed Pharmaceuticals), Resmetirom (Madrigal Pharmaceuticals), Dapagliflozin and Semaglutide (Novo Nordisk) are in advanced Phase 3 clinical trials, while Belapectin (Galectin Therapeutics), MSDC-0602K (Cirius Therapeutics), Lanifibranor (Inventiva), Efruxifermin (Akero) and Tesamorelin (Theratechnologies) are expected to start Phase 3 trials soon. Here, we have performed an exhaustive review of the current therapeutic landscape for this disease and compared, in some detail, the fortunes of different drug classes (biologics vs small molecules) and target molecules. Given the complex pathophysiology of NASH, the use of drug combination, different mechanisms of actions and the targeting of each stage of the disease will likely be required. Hence, the development of a single therapy for NASH seems challenging and unlikely, despite the plethora of later stage trials due to report. We therefore predict that clinical, patient and company interest in pipeline and next-generation therapies will remain high for some time to come.


Asunto(s)
Desarrollo de Medicamentos , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Quimioterapia Combinada , Humanos , Inflamación/tratamiento farmacológico , Inflamación/fisiopatología , Enfermedad del Hígado Graso no Alcohólico/fisiopatología
3.
Methods Mol Biol ; 1731: 261-269, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29318560

RESUMEN

Proteases play key roles in the execution and regulation of most if not all biological functions, and alterations in their activity, expression, or location are associated with multiple pathological conditions, including cancer and aging. In this regard, the use of RNA interference-based approaches to specifically target the expression of individual proteases constitutes an invaluable tool to identify enzymes involved in central aspects of these processes and to explore their potential as targets of therapeutic interventions. Here we describe simple protocols to optimize and monitor the specific silencing of cancer- and aging-related proteases.


Asunto(s)
Envejecimiento/metabolismo , Proteínas de la Membrana/genética , Metaloendopeptidasas/genética , Neoplasias/patología , Interferencia de ARN , Proteasas Ubiquitina-Específicas/genética , Envejecimiento/genética , Apoptosis/genética , Línea Celular Tumoral , Técnicas de Silenciamiento del Gen/instrumentación , Técnicas de Silenciamiento del Gen/métodos , Vectores Genéticos/genética , Humanos , Lentivirus/genética , Proteínas de la Membrana/metabolismo , Metaloendopeptidasas/metabolismo , Neoplasias/genética , ARN Interferente Pequeño/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteasas Ubiquitina-Específicas/metabolismo
4.
J Biol Chem ; 293(6): 2183-2194, 2018 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-29273634

RESUMEN

Deubiquitinases are proteases with a wide functional diversity that profoundly impact multiple biological processes. Among them, the ubiquitin-specific protease 36 (USP36) has been implicated in the regulation of nucleolar activity. However, its functional relevance in vivo has not yet been fully described. Here, we report the generation of an Usp36-deficient mouse model to examine the function of this enzyme. We show that Usp36 depletion is lethal in preimplantation mouse embryos, where it blocks the transition from morula to blastocyst during embryonic development. USP36 reduces the ubiquitination levels and increases the stability of the DEAH-box RNA helicase DHX33, which is critically involved in ribosomal RNA synthesis and mRNA translation. In agreement with this finding, O-propargyl-puromycin incorporation experiments, Northern blot, and electron microscopy analyses demonstrated the role of USP36 in ribosomal RNA and protein synthesis. Finally, we show that USP36 down-regulation alters cell proliferation in human cancer cells by inducing both apoptosis and cell cycle arrest, and that reducing DHX33 levels through short hairpin RNA interference has the same effect. Collectively, these results support that Usp36 is essential for cell and organism viability because of its role in ribosomal RNA processing and protein synthesis, which is mediated, at least in part, by regulating DHX33 stability.


Asunto(s)
Blastocisto , ARN Helicasas DEAD-box/química , Enzimas Desubicuitinizantes/fisiología , ARN Helicasas/química , Ubiquitina Tiolesterasa/fisiología , Animales , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Enzimas Desubicuitinizantes/genética , Pérdida del Embrión , Humanos , Ratones , Ratones Noqueados , Biosíntesis de Proteínas , Estabilidad Proteica , ARN Ribosómico , Ubiquitina Tiolesterasa/genética
5.
J Biol Chem ; 292(10): 4164-4175, 2017 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-28154181

RESUMEN

KRAS is the most frequently mutated oncogene in human cancer, but its therapeutic targeting remains challenging. Here, we report a synthetic lethal screen with a library of deubiquitinases and identify USP39, which encodes an essential splicing factor, as a critical gene for the viability of KRAS-dependent cells. We show that splicing fidelity inhibitors decrease preferentially the proliferation rate of KRAS-active cells. Moreover, depletion of DHX38, encoding an USP39-interacting splicing factor, also reduces the viability of these cells. In agreement with these results, USP39 depletion caused a significant reduction in pre-mRNA splicing efficiency, as demonstrated through RNA-seq experiments. Furthermore, we show that USP39 is up-regulated in lung and colon carcinomas and its expression correlates with KRAS levels and poor clinical outcome. Accordingly, our work provides critical information for the development of splicing-directed antitumor treatments and supports the potential of USP39-targeting strategies as the basis of new anticancer therapies.


Asunto(s)
Neoplasias del Colon/patología , Neoplasias Pulmonares/patología , Mutación/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteasas Ubiquitina-Específicas/metabolismo , Animales , Apoptosis , Western Blotting , Proliferación Celular , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Ratones , Ratones Desnudos , Pronóstico , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tasa de Supervivencia , Células Tumorales Cultivadas , Proteasas Ubiquitina-Específicas/genética , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Oncotarget ; 7(46): 74427-74434, 2016 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-27769071

RESUMEN

Ubiquitin-Specific Proteases (USPs) are deubiquitinating enzymes frequently deregulated in human malignancies. Here, we show that USP54 is overexpressed in intestinal stem cells and demonstrate that its downregulation in colorectal carcinoma cells impedes tumorigenesis. We have generated mutant mice deficient for this deubiquitinase, which are viable and fertile, and protected against chemically-induced colorectal carcinoma. Furthermore, we show that USP54 is upregulated in human colon cancer and associates with poor prognosis. In agreement with these results, Usp54 downregulation in mouse melanoma cells inhibits lung metastasis formation. Collectively, this work has uncovered the pro-tumorigenic properties of USP54, highlighting the importance of deubiquitinating enzymes as promising targets for the development of specific anti-cancer therapies.


Asunto(s)
Neoplasias Colorrectales/genética , Expresión Génica , Células Madre Neoplásicas/metabolismo , Proteasas Ubiquitina-Específicas/genética , Animales , Biomarcadores de Tumor , Línea Celular Tumoral , Proliferación Celular , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Progresión de la Enfermedad , Desarrollo Embrionario/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones Noqueados , Invasividad Neoplásica , Metástasis de la Neoplasia , Proteasas Ubiquitina-Específicas/metabolismo
8.
Oncotarget ; 4(11): 1919-32, 2013 11.
Artículo en Inglés | MEDLINE | ID: mdl-24243807

RESUMEN

Proteolytic enzymes play important roles during tumor development and progression through their ability to promote cell growth or by facilitating the invasion of surrounding tissues. The human genome contains more than 570 protease-coding genes, many of them forming functional networks, which has forced the use of global strategies for the analysis of this group of enzymes. In this study, we have designed a new quantitative PCR-based device for profiling the entire degradome in human malignancies. We have used this method to evaluate protease expression levels in colorectal carcinomas with the finding that most proteases with altered expression in these tumors exert their function in the extracellular compartment. In addition, we have found that among genes encoding repressed proteases there was a higher proportion with somatic mutations in colorectal cancer when compared to genes coding for upregulated proteases (14% vs. 4%, p<0.05). One of these genes, MASP3, is consistently repressed in colorectal carcinomas as well as in colorectal cancer cell lines when compared to normal colonic mucosa. Functional analysis of this gene revealed that ectopic expression of MASP3 reduces cell proliferation in vitro and restrains subcutaneous tumor growth, whereas its downregulation induces an increase in the tumorigenic potential of colorectal cancer cells. These results provide new insights into the diversity of proteases associated with cancer and support the utility of degradome profiling to identify novel proteases with tumor-defying functions.


Asunto(s)
Neoplasias Colorrectales/enzimología , Péptido Hidrolasas/metabolismo , Animales , Células CACO-2 , Proteínas de Caenorhabditis elegans , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , Células HCT116 , Células HEK293 , Humanos , Ratones , Ratones Desnudos , Péptido Hidrolasas/genética , Transfección
9.
Am J Hum Genet ; 88(5): 650-6, 2011 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-21549337

RESUMEN

Accelerated aging syndromes represent a valuable source of information about the molecular mechanisms involved in normal aging. Here, we describe a progeroid syndrome that partially phenocopies Hutchinson-Gilford progeria syndrome (HGPS) but also exhibits distinctive features, including the absence of cardiovascular deficiencies characteristic of HGPS, the lack of mutations in LMNA and ZMPSTE24, and a relatively long lifespan of affected individuals. Exome sequencing and molecular analysis in two unrelated families allowed us to identify a homozygous mutation in BANF1 (c.34G>A [p.Ala12Thr]), encoding barrier-to-autointegration factor 1 (BAF), as the molecular abnormality responsible for this Mendelian disorder. Functional analysis showed that fibroblasts from both patients have a dramatic reduction in BAF protein levels, indicating that the p.Ala12Thr mutation impairs protein stability. Furthermore, progeroid fibroblasts display profound abnormalities in the nuclear lamina, including blebs and abnormal distribution of emerin, an interaction partner of BAF. These nuclear abnormalities are rescued by ectopic expression of wild-type BANF1, providing evidence for the causal role of this mutation. These data demonstrate the utility of exome sequencing for identifying the cause of rare Mendelian disorders and underscore the importance of nuclear envelope alterations in human aging.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas Nucleares/genética , Adulto , Núcleo Celular , Células Cultivadas , Preescolar , Análisis Mutacional de ADN , Femenino , Fibroblastos/metabolismo , Ligamiento Genético , Homocigoto , Humanos , Lamina Tipo A/genética , Lamina Tipo A/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Mutación , Proteínas Nucleares/metabolismo , Linaje , Fenotipo , Progeria/genética , Progeria/metabolismo , Progeria/patología , Conformación Proteica , Alineación de Secuencia
10.
Front Oncol ; 1: 25, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22655236

RESUMEN

The "oncogene addiction" concept refers to the dependence of cancer cells on the function of the oncogenes responsible for their transformed phenotype, while the term "non-oncogene addiction" has been introduced to define the exacerbated necessity of the normal function of non-mutated genes. In this Perspective, we focus on the importance of proteolytic enzymes to maintain the viability of cancer cells and hypothesize that most, if not all, tumors present "addiction" to a number of proteolytic activities, which in turn may represent valuable targets of anti-cancer therapies, even without being mutated or over-expressed by the malignant cells.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA