Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
PLoS Biol ; 11(11): e1001717, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24302884

RESUMEN

Histone deacetylase (HDAC) 4 is a transcriptional repressor that contains a glutamine-rich domain. We hypothesised that it may be involved in the molecular pathogenesis of Huntington's disease (HD), a protein-folding neurodegenerative disorder caused by an aggregation-prone polyglutamine expansion in the huntingtin protein. We found that HDAC4 associates with huntingtin in a polyglutamine-length-dependent manner and co-localises with cytoplasmic inclusions. We show that HDAC4 reduction delayed cytoplasmic aggregate formation, restored Bdnf transcript levels, and rescued neuronal and cortico-striatal synaptic function in HD mouse models. This was accompanied by an improvement in motor coordination, neurological phenotypes, and increased lifespan. Surprisingly, HDAC4 reduction had no effect on global transcriptional dysfunction and did not modulate nuclear huntingtin aggregation. Our results define a crucial role for the cytoplasmic aggregation process in the molecular pathology of HD. HDAC4 reduction presents a novel strategy for targeting huntingtin aggregation, which may be amenable to small-molecule therapeutics.


Asunto(s)
Histona Desacetilasas/genética , Enfermedad de Huntington/enzimología , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Corteza Cerebral/enzimología , Corteza Cerebral/patología , Epigénesis Genética , Femenino , Técnicas de Silenciamiento del Gen , Histona Desacetilasas/metabolismo , Proteína Huntingtina , Enfermedad de Huntington/fisiopatología , Enfermedad de Huntington/terapia , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Noqueados , Neuronas/fisiología , Fenotipo , Prueba de Desempeño de Rotación con Aceleración Constante , Transmisión Sináptica , Transcripción Genética
3.
Proc Natl Acad Sci U S A ; 110(39): 15704-9, 2013 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-24023063

RESUMEN

Development of isoform-selective histone deacetylase (HDAC) inhibitors is important in elucidating the function of individual HDAC enzymes and their potential as therapeutic agents. Among the eleven zinc-dependent HDACs in humans, HDAC6 is structurally and functionally unique. Here, we show that a hydroxamic acid-based small-molecule N-hydroxy-4-(2-[(2-hydroxyethyl)(phenyl)amino]-2-oxoethyl)benzamide (HPOB) selectively inhibits HDAC6 catalytic activity in vivo and in vitro. HPOB causes growth inhibition of normal and transformed cells but does not induce cell death. HPOB enhances the effectiveness of DNA-damaging anticancer drugs in transformed cells but not normal cells. HPOB does not block the ubiquitin-binding activity of HDAC6. The HDAC6-selective inhibitor HPOB has therapeutic potential in combination therapy to enhance the potency of anticancer drugs.


Asunto(s)
Inhibidores de Histona Desacetilasas/síntesis química , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Acetilación/efectos de los fármacos , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Línea Celular Transformada/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Roturas del ADN de Doble Cadena/efectos de los fármacos , Doxorrubicina/farmacología , Etopósido/farmacología , Inhibidores de Histona Desacetilasas/química , Histonas/metabolismo , Humanos , Ácidos Hidroxámicos/farmacología , Ratones , Trehalosa/farmacología , Tubulina (Proteína)/metabolismo , Vorinostat , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Adv Cancer Res ; 116: 39-86, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23088868

RESUMEN

Histone deacetylase (HDAC) inhibitors are a new class of anticancer agents. HDAC inhibitors induce acetylation of histones and nonhistone proteins which are involved in regulation of gene expression and in various cellular pathways including cell growth arrest, differentiation, DNA damage and repair, redox signaling, and apoptosis (Marks, 2010). The U.S. Food and Drug Administration has approved two HDAC inhibitors, vorinostat and romidepsin, for the treatment of cutaneous T-cell lymphoma (Duvic & Vu, 2007; Grant et al., 2010; Marks & Breslow, 2007). Over 20 chemically different HDAC inhibitors are in clinical trials for hematological malignancies and solid tumors. This review considers the mechanisms of resistance to HDAC inhibitors that have been identified which account for the selective effects of these agents in inducing cancer but not normal cell death. These mechanisms, such as functioning Chk1, high levels of thioredoxin, or the prosurvival BCL-2, may also contribute to resistance of cancer cells to HDAC inhibitors.


Asunto(s)
Resistencia a Antineoplásicos , Inhibidores de Histona Desacetilasas/farmacología , Proteínas de Neoplasias/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Animales , Histona Desacetilasas/química , Humanos
5.
Autophagy ; 8(10): 1521-2, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22894919

RESUMEN

Cells respond to cytotoxicity by activating a variety of signal transduction pathways. One pathway frequently upregulated during cytotoxic response is macroautophagy (hereafter referred to as autophagy). Previously, we demonstrated that pan-histone deacetylase (HDAC) inhibitors, such as the anticancer agent suberoylanilide hydroxamic acid (SAHA, Vorinostat), can induce autophagy. In this study, we show that HDAC inhibition triggers autophagy by suppressing MTOR and activating the autophagic kinase ULK1. Furthermore, autophagy inhibition can sensitize cells to both apoptotic and nonapoptotic cell death induced by SAHA, suggesting the therapeutic potential of autophagy targeting in combination with SAHA therapy. This study also raised a series of questions: What is the role of HDACs in regulating autophagy? Do individual HDACs have distinct functions in autophagy? How do HDACs regulate the nutrient-sensing kinase MTOR? Since SAHA-induced nonapoptotic cell death is not driven by autophagy, what then is the mechanism underlying the apoptosis-independent death? Tackling these questions should lead to a better understanding of autophagy and HDAC biology and contribute to the development of novel therapeutic strategies.


Asunto(s)
Autofagia/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Neoplasias/patología , Animales , Línea Celular Tumoral , Humanos , Ácidos Hidroxámicos/farmacología , Modelos Biológicos , Interferencia de ARN/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Vorinostat
6.
Proc Natl Acad Sci U S A ; 109(17): 6561-5, 2012 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-22493260

RESUMEN

Autophagy is a cellular catabolic pathway by which long-lived proteins and damaged organelles are targeted for degradation. Activation of autophagy enhances cellular tolerance to various stresses. Recent studies indicate that a class of anticancer agents, histone deacetylase (HDAC) inhibitors, can induce autophagy. One of the HDAC inhibitors, suberoylanilide hydroxamic acid (SAHA), is currently being used for treating cutaneous T-cell lymphoma and under clinical trials for multiple other cancer types, including glioblastoma. Here, we show that SAHA increases the expression of the autophagic factor LC3, and inhibits the nutrient-sensing kinase mammalian target of rapamycin (mTOR). The inactivation of mTOR results in the dephosphorylation, and thus activation, of the autophagic protein kinase ULK1, which is essential for autophagy activation during SAHA treatment. Furthermore, we show that the inhibition of autophagy by RNAi in glioblastoma cells results in an increase in SAHA-induced apoptosis. Importantly, when apoptosis is pharmacologically blocked, SAHA-induced nonapoptotic cell death can also be potentiated by autophagy inhibition. Overall, our findings indicate that SAHA activates autophagy via inhibiting mTOR and up-regulating LC3 expression; autophagy functions as a prosurvival mechanism to mitigate SAHA-induced apoptotic and nonapoptotic cell death, suggesting that targeting autophagy might improve the therapeutic effects of SAHA.


Asunto(s)
Apoptosis/efectos de los fármacos , Autofagia , Muerte Celular/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Ácidos Hidroxámicos/farmacología , Animales , Homólogo de la Proteína 1 Relacionada con la Autofagia , Línea Celular , Ratones , Proteínas Serina-Treonina Quinasas , Interferencia de ARN , Reacción en Cadena en Tiempo Real de la Polimerasa , Células Tumorales Cultivadas , Vorinostat
7.
PLoS One ; 6(11): e27746, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22140466

RESUMEN

Huntington's disease (HD) is a progressive neurological disorder for which there are no disease-modifying treatments. Transcriptional dysregulation is a major molecular feature of HD, which significantly contributes to disease progression. Therefore, the development of histone deacetylase (HDAC) inhibitors as therapeutics for HD has been energetically pursued. Suberoylanilide hydroxamic acid (SAHA) - a class I HDAC as well an HDAC6 inhibitor, improved motor impairment in the R6/2 mouse model of HD. Recently it has been found that SAHA can also promote the degradation of HDAC4 and possibly other class IIa HDACs at the protein level in various cancer cell lines. To elucidate whether SAHA is a potent modifier of HDAC protein levels in vivo, we performed two independent mouse trials. Both WT and R6/2 mice were chronically treated with SAHA and vehicle. We found that prolonged SAHA treatment causes the degradation of HDAC4 in cortex and brain stem, but not hippocampus, without affecting its transcript levels in vivo. Similarly, SAHA also decreased HDAC2 levels without modifying the expression of its mRNA. Consistent with our previous data, SAHA treatment diminishes Hdac7 transcript levels in both wild type and R6/2 brains and unexpectedly was found to decrease Hdac11 in R6/2 but not wild type. We investigated the effects of SAHA administration on well-characterised molecular readouts of disease progression. We found that SAHA reduces SDS-insoluble aggregate load in the cortex and brain stem but not in the hippocampus of the R6/2 brains, and that this was accompanied by restoration of Bdnf cortical transcript levels.


Asunto(s)
Histona Desacetilasa 2/metabolismo , Histona Desacetilasas/metabolismo , Enfermedad de Huntington/tratamiento farmacológico , Enfermedad de Huntington/enzimología , Ácidos Hidroxámicos/uso terapéutico , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Exones/genética , Histona Desacetilasa 2/genética , Histona Desacetilasas/genética , Ácidos Hidroxámicos/administración & dosificación , Ácidos Hidroxámicos/farmacología , Ratones , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Vorinostat
8.
Proc Natl Acad Sci U S A ; 108(49): 19629-34, 2011 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-22106282

RESUMEN

Histone deacetylase inhibitors (HDACi) are a new group of anticancer drugs with tumor selective toxicity. Normal cells are relatively resistant to HDACi-induced cell death compared with cancer cells. Previously, we found that vorinostat induces DNA breaks in normal and transformed cells, which normal but not cancer cells can repair. In this study, we found that checkpoint kinase 1 (Chk1), a component of the G2 DNA damage checkpoint, is important in the resistance of normal cells to HDACi in vitro and in vivo. Inhibition of Chk1 activity with Chk1 inhibitor (UCN-01, AZD7762, or CHIR-124) in normal cells increases their sensitivity to HDACi (vorinostat, romidepsin, or entinostat) induced cell death, associated with extensive mitotic disruption. Mitotic abnormalities included loss of sister chromatid cohesion and chromosomal disruption. Inhibition of Chk1 did increase HDACi-induced cell death of transformed cells. Thus, Chk1 is an important factor in the resistance of normal cells, and some transformed cells, to HDACi-induced cell death. Use of Chk1 inhibitors in combination with anticancer agents to treat cancers may be associated with substantial toxicity.


Asunto(s)
Resistencia a Medicamentos , Inhibidores de Histona Desacetilasas/farmacología , Proteínas Quinasas/metabolismo , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Daño del ADN , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Prepucio/citología , Histonas/metabolismo , Humanos , Ácidos Hidroxámicos/farmacología , Immunoblotting , Masculino , Ratones , Proteínas Quinasas/genética , Quinolinas/farmacología , Quinuclidinas/farmacología , Interferencia de ARN/fisiología , Bazo/efectos de los fármacos , Bazo/metabolismo , Estaurosporina/análogos & derivados , Estaurosporina/farmacología , Tiofenos/farmacología , Urea/análogos & derivados , Urea/farmacología , Vorinostat
10.
Proc Natl Acad Sci U S A ; 107(46): 20003-8, 2010 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-21037108

RESUMEN

Histone deacetylase 6 (HDAC6) is structurally and functionally unique among the 11 human zinc-dependent histone deacetylases. Here we show that chemical inhibition with the HDAC6-selective inhibitor tubacin significantly enhances cell death induced by the topoisomerase II inhibitors etoposide and doxorubicin and the pan-HDAC inhibitor SAHA (vorinostat) in transformed cells (LNCaP, MCF-7), an effect not observed in normal cells (human foreskin fibroblast cells). The inactive analogue of tubacin, nil-tubacin, does not sensitize transformed cells to these anticancer agents. Further, we show that down-regulation of HDAC6 expression by shRNA in LNCaP cells enhances cell death induced by etoposide, doxorubicin, and SAHA. Tubacin in combination with SAHA or etoposide is more potent than either drug alone in activating the intrinsic apoptotic pathway in transformed cells, as evidenced by an increase in PARP cleavage and partial inhibition of this effect by the pan-caspase inhibitor Z-VAD-fmk. HDAC6 inhibition with tubacin induces the accumulation of γH2AX, an early marker of DNA double-strand breaks. Tubacin enhances DNA damage induced by etoposide or SAHA as indicated by increased accumulation of γH2AX and activation of the checkpoint kinase Chk2. Tubacin induces the expression of DDIT3 (CHOP/GADD153), a transcription factor up-regulated in response to cellular stress. DDIT3 induction is further increased when tubacin is combined with SAHA. These findings point to mechanisms by which HDAC6-selective inhibition can enhance the efficacy of certain anti-cancer agents in transformed cells.


Asunto(s)
Anilidas/farmacología , Antineoplásicos/farmacología , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/patología , Daño del ADN , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Ácidos Hidroxámicos/farmacología , Caspasas/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Quinasa de Punto de Control 2 , Replicación del ADN/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Doxorrubicina/farmacología , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Etopósido/farmacología , Fase G1/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Histona Desacetilasa 6 , Histonas/metabolismo , Humanos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Inhibidores de Topoisomerasa II/farmacología , Regulación hacia Arriba/efectos de los fármacos
11.
Expert Opin Investig Drugs ; 19(9): 1049-66, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20687783

RESUMEN

IMPORTANCE OF THE FIELD: Histone deacetylase (HDAC) inhibitors are being developed as a new, targeted class of anticancer drugs. AREA COVERED IN THIS REVIEW: This review focuses on the mechanisms of action of the HDAC inhibitors, which selectively induce cancer cell death. WHAT THE READER WILL GAIN: There are 11 zinc-dependent HDACs in humans and the biological roles of these lysine deacetylases are not completely understood. It is clear that these different HDACs are not redundant in their activity. This review focuses on the mechanisms by which HDAC inhibitors can induce transformed cell growth arrest and cell death, inhibit cell mobility and have antiangiogenesis activity. There are more than a dozen HDAC inhibitors, including hydroxamates, cyclic peptides, benzamides and fatty acids, in various stages of clinical trials and many more compounds in preclinical development. The chemically different HDAC inhibitors may target different HDACs. TAKE HOME MESSAGE: There are extensive preclinical studies with transformed cells in culture and tumor-bearing animal models, as well as limited clinical studies reported to date, which indicate that HDAC inhibitors will be most useful when used in combination with cytotoxic or other targeted anticancer agents.


Asunto(s)
Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Inhibidores de Histona Desacetilasas/farmacología , Inhibidores de Histona Desacetilasas/uso terapéutico , Neoplasias/tratamiento farmacológico , Inhibidores de la Angiogénesis/farmacología , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Ensayos Clínicos como Asunto , Resistencia a Antineoplásicos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Histona Desacetilasas/metabolismo , Humanos , Ratones , Terapia Molecular Dirigida , Neoplasias/enzimología , Neoplasias/genética , Zinc/metabolismo
12.
Proc Natl Acad Sci U S A ; 107(33): 14639-44, 2010 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-20679231

RESUMEN

Histone deacetylase inhibitors (HDACi) developed as anti-cancer agents have a high degree of selectivity for killing cancer cells. HDACi induce acetylation of histones and nonhistone proteins, which affect gene expression, cell cycle progression, cell migration, and cell death. The mechanism of the tumor selective action of HDACi is unclear. Here, we show that the HDACi, vorinostat (Suberoylanilide hydroxamic acid, SAHA), induces DNA double-strand breaks (DSBs) in normal (HFS) and cancer (LNCaP, A549) cells. Normal cells in contrast to cancer cells repair the DSBs despite continued culture with vorinostat. In transformed cells, phosphorylated H2AX (gammaH2AX), a marker of DNA DSBs, levels increased with continued culture with vorinostat, whereas in normal cells, this marker decreased with time. Vorinostat induced the accumulation of acetylated histones within 30 min, which could alter chromatin structure-exposing DNA to damage. After a 24-h culture of cells with vorinostat, and reculture without the HDACi, gammaH2AX was undetectable by 2 h in normal cells, while persisting in transformed cells for the duration of culture. Further, we found that vorinostat suppressed DNA DSB repair proteins, e.g., RAD50, MRE11, in cancer but not normal cells. Thus, the HDACi, vorinostat, induces DNA damage which normal but not cancer cells can repair. This DNA damage is associated with cancer cell death. These findings can explain, in part, the selectivity of vorinostat in causing cancer cell death at concentrations that cause little or no normal cell death.


Asunto(s)
Daño del ADN , Reparación del ADN , Inhibidores de Histona Desacetilasas/farmacología , Ácidos Hidroxámicos/farmacología , Acetilación/efectos de los fármacos , Antineoplásicos/farmacología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Células Cultivadas , Roturas del ADN de Doble Cadena/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Citometría de Flujo , Prepucio/citología , Histonas/metabolismo , Humanos , Immunoblotting , Masculino , Microscopía Fluorescente , Vorinostat
13.
Biochim Biophys Acta ; 1799(10-12): 717-25, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20594930

RESUMEN

There are eleven zinc dependent histone deacetylases (HDAC) in humans which have histones and many non-histone substrates. The substrates of these enzymes include proteins that have a role in regulation of gene expression, cell proliferation, cell migration, cell death, immune pathways and angiogenesis. Inhibitors of HDACs (HDACi) have been developed which alter the structure and function of these proteins, causing molecular and cellular changes that induce transformed cell death. The HDACi are being developed as anti-cancer drugs and have therapeutic potential for many non-oncologic diseases.


Asunto(s)
Inhibidores de Histona Desacetilasas/química , Inhibidores de Histona Desacetilasas/metabolismo , Histona Desacetilasas/química , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Animales , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/uso terapéutico , Muerte Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Proliferación Celular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Inhibidores de Histona Desacetilasas/uso terapéutico , Humanos , Neovascularización Fisiológica/efectos de los fármacos , Neovascularización Fisiológica/genética , Relación Estructura-Actividad
16.
Invest Ophthalmol Vis Sci ; 50(9): 4072-9, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19387079

RESUMEN

PURPOSE: To characterize the molecular sequelae induced in retinoblastoma (Rb) cells by histone deacetylase inhibitors (HDACIs). Hydroxamic acid-based HDACIs such as vorinostat (suberoylanilide hydroxamic acid) induce the differentiation and apoptosis of transformed cells. Vorinostat has demonstrated significant anticancer activity against hematologic and solid tumors at doses well tolerated by patients and has been approved for the treatment of patients with cutaneous T-cell lymphoma. METHODS: The authors evaluated the effects of the HDACIs vorinostat and m-carboxycinnamic acid bis-hydroxamide on the Rb cell lines Y79 and WERI-Rb1 with the use of the MTT assay, BrdU incorporation assay, flow cytometry, immunoblotting, gene-expression profiling, quantitative RT-PCR, and NF-kappaB DNA-binding assay. RESULTS: Both HDACIs were effective against both Rb cell lines, inducing growth arrest and apoptosis in vitro. Vorinostat increased p53 expression and activated caspases -8, -9 and -3, whereas caspase inhibition abrogated vorinostat-induced apoptosis. Vorinostat downregulated baseline NF-kappaB activity and potentiated the activity of the DNA-damaging chemotherapeutic doxorubicin. Gene expression profiling and qRT-PCR demonstrated that vorinostat modulated the mRNA levels of genes important for signal transduction, cell cycle, cellular metabolism, stress response, apoptosis, extracellular matrix synthesis, and cell differentiation. Notably, several transcripts involved in the ephrin and Notch signaling pathways were upregulated. CONCLUSIONS: HDACIs, such as vorinostat, induce caspase-dependent apoptosis in Rb cells, downregulate baseline NF-kappaB activity, and potentiate the effectiveness of conventional chemotherapy. The finding that vorinostat augments the effectiveness of doxorubicin provides a rationale for future clinical studies looking at the use of vorinostat in combination with conventional chemotherapy in Rb.


Asunto(s)
Apoptosis , Inhibidores Enzimáticos/farmacología , Inhibidores de Histona Desacetilasas , Neoplasias de la Retina/patología , Retinoblastoma/patología , Acetilación , Bromodesoxiuridina/metabolismo , Caspasas/metabolismo , Línea Celular Tumoral , Proliferación Celular , Cinamatos/farmacología , Doxorrubicina/farmacología , Sinergismo Farmacológico , Citometría de Flujo , Perfilación de la Expresión Génica , Histonas/metabolismo , Humanos , Ácidos Hidroxámicos/farmacología , Immunoblotting , FN-kappa B/metabolismo , Proteínas de Neoplasias/genética , Neoplasias de la Retina/enzimología , Retinoblastoma/enzimología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína p53 Supresora de Tumor/metabolismo , Vorinostat
17.
J Clin Invest ; 118(4): 1223-4, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18382727

RESUMEN

The annual Atlantic City meeting of the Young Turks was an exciting event--an opportunity to hear great science, to explore career opportunities, and to meet and make friends. Though the 1950s, '60s, and '70s, the meetings remained relatively intimate, broadly covering the best in medicine and constantly growing until the participants outgrew the Atlantic City venue to eventually spawn the numerous specialty annual meetings that we have today.


Asunto(s)
Congresos como Asunto/historia , Sociedades Médicas , Historia del Siglo XX , Historia del Siglo XXI , Sociedades Médicas/historia , Estados Unidos
18.
Mol Cancer Res ; 5(10): 981-9, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17951399

RESUMEN

Histone deacetylase inhibitors (HDACi) comprise structurally diverse compounds that are a group of targeted anticancer agents. The first of these new HDACi, vorinostat (suberoylanilide hydroxamic acid), has received Food and Drug Administration approval for treating patients with cutaneous T-cell lymphoma. This review focuses on the activities of the 11 zinc-containing HDACs, their histone and nonhistone protein substrates, and the different pathways by which HDACi induce transformed cell death. A hypothesis is presented to explain the relative resistance of normal cells to HDACi-induced cell death.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Inhibidores de Histona Desacetilasas , Metaloproteínas/antagonistas & inhibidores , Neoplasias/enzimología , Antineoplásicos/uso terapéutico , Ensayos Clínicos como Asunto , Inhibidores Enzimáticos/uso terapéutico , Histona Desacetilasas/clasificación , Histona Desacetilasas/metabolismo , Humanos , Metaloproteínas/clasificación , Metaloproteínas/metabolismo , Neoplasias/tratamiento farmacológico , Zinc/metabolismo
19.
Mol Cancer Ther ; 6(9): 2525-34, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17876049

RESUMEN

There are 18 histone deacetylases (HDAC) generally divided into four classes based on homology to yeast HDACs. HDACs have many protein substrates in addition to histones that are involved in regulation of gene expression, cell proliferation, and cell death. Inhibition of HDACs can cause accumulation of acetylated forms of these proteins, thus altering their function. HDAC inhibitors (HDACi), such as the hydroxamic acid-based vorinostat (suberoylanilide hydroxamic acid), inhibit the zinc-containing classes I, II, and IV, but not the NAD(+)-dependent class III, enzymes. HDACis are a group of novel anticancer agents. Vorinostat is the first HDACi approved for clinical use in the treatment of the cancer cutaneous T-cell lymphoma. Factors affecting expression of HDACs are not well understood. This study focuses on the effect of the HDACi vorinostat on the expression of class I and class II HDACs. We found that vorinostat selectively down-regulates HDAC7 with little or no effect on the expression of other class I or class II HDACs. Fourteen cell lines were examined, including normal, immortalized, genetically transformed, and human cancer-derived cell lines. Down-regulation of HDAC7 by vorinostat is more pronounced in transformed cells sensitive to inhibitor-induced cell death than in normal cells or cancer cells resistant to induced cell death. Modulation of HDAC7 levels by small interfering RNA-mediated knockdown or by HDAC7 overexpression is associated with growth arrest but without detectable changes in acetylation of histones or p21 gene expression. Selective down-regulation of HDAC7 protein may serve as a marker of response of tumors to HDACi.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Histona Desacetilasas/metabolismo , Ácidos Hidroxámicos/farmacología , Acetilación , Northern Blotting , Western Blotting , Transformación Celular Neoplásica/patología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/enzimología , Inhibidores de Histona Desacetilasas , Histona Desacetilasas/clasificación , Histona Desacetilasas/genética , Humanos , Pulmón/efectos de los fármacos , Pulmón/enzimología , Masculino , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Neoplasias/patología , Próstata/efectos de los fármacos , Próstata/enzimología , ARN Interferente Pequeño/farmacología , Piel/efectos de los fármacos , Piel/enzimología , Vorinostat
20.
Proc Natl Acad Sci U S A ; 104(19): 8071-6, 2007 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-17470784

RESUMEN

Histone deacetylase inhibitors (HDACi) can elicit a range of biological responses that affect tumor growth and survival, including inhibition of cell cycle progression, induction of tumor cell-selective apoptosis, suppression of angiogenesis, and modulation of immune responses, and show promising activity against hematological malignancies in clinical trials. Using the Emu-myc model of B cell lymphoma, we screened tumors with defined genetic alterations in apoptotic pathways for therapeutic responsiveness to the HDACi vorinostat. We demonstrated a direct correlation between induction of tumor cell apoptosis in vivo and therapeutic efficacy. Vorinostat did not require p53 activity or a functional death receptor pathway to kill Emu-myc lymphomas and mediate a therapeutic response but depended on activation of the intrinsic apoptotic pathway with the proapoptotic BH3-only proteins Bid and Bim playing an important role. Our studies provide important information regarding the mechanisms of action of HDACi that have broad implications regarding stratification of patients receiving HDACi therapy alone or in combination with other anticancer agents.


Asunto(s)
Apoptosis/efectos de los fármacos , Inhibidores Enzimáticos/uso terapéutico , Inhibidores de Histona Desacetilasas , Ácidos Hidroxámicos/uso terapéutico , Linfoma de Células B/tratamiento farmacológico , Animales , Proteínas Reguladoras de la Apoptosis/fisiología , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/fisiología , Proteína 11 Similar a Bcl2 , Genes myc , Proteínas de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas/fisiología , Proteínas Proto-Oncogénicas c-bcl-2/fisiología , Ligando Inductor de Apoptosis Relacionado con TNF/fisiología , Proteína p53 Supresora de Tumor/fisiología , Vorinostat , Proteína bcl-X/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA