Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Invest New Drugs ; 31(5): 1251-6, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23543109

RESUMEN

PURPOSE: To determine the maximum tolerated dose (MTD) and characterize the dose-limiting toxicities (DLT) of tanespimycin when given in combination with bortezomib. EXPERIMENTAL DESIGN: Phase I dose-escalating trial using a standard cohort "3+3" design performed in patients with advanced solid tumors. Patients were given tanespimycin and bortezomib twice weekly for 2 weeks in a 3 week cycle (days 1, 4, 8, 11 every 21 days). RESULTS: Seventeen patients were enrolled in this study, fifteen were evaluable for toxicity, and nine patients were evaluable for tumor response. The MTD was 250 mg/m(2) of tanespimycin and 1.0 mg/m(2) of bortezomib when used in combination. DLTs of abdominal pain (13 %), complete atrioventricular block (7 %), fatigue (7 %), encephalopathy (7 %), anorexia (7 %), hyponatremia (7 %), hypoxia (7 %), and acidosis (7 %) were observed. There were no objective responses. One patient had stable disease. CONCLUSIONS: The recommended phase II dose for twice weekly 17-AAG and PS341 are 250 mg/m(2) and 1.0 mg/m(2), respectively, on days 1, 4, 8 and 11 of a 21 day cycle.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Neoplasias/tratamiento farmacológico , Anciano , Anciano de 80 o más Años , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Benzoquinonas/administración & dosificación , Benzoquinonas/efectos adversos , Ácidos Borónicos/administración & dosificación , Ácidos Borónicos/efectos adversos , Bortezomib , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Lactamas Macrocíclicas/administración & dosificación , Lactamas Macrocíclicas/efectos adversos , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/efectos adversos , Pirazinas/administración & dosificación , Pirazinas/efectos adversos , Resultado del Tratamiento
2.
Invest New Drugs ; 29(3): 473-80, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20082116

RESUMEN

PURPOSE: To determine the maximum tolerated dose (MTD) and characterize the dose-limiting toxicities (DLT) of 17-AAG, gemcitabine and/or cisplatin. Levels of the proteins Hsp90, Hsp70 and ILK were measured in peripheral blood mononuclear cell (PMBC) lysates to assess the effects of 17-AAG. EXPERIMENTAL DESIGN: Phase I dose-escalating trial using a "3 + 3" design performed in patients with advanced solid tumors. Once the MTD of gemcitabine + 17-AAG + cisplatin was determined, dose escalation of 17-AAG with constant doses of gemcitabine and cisplatin was attempted. After significant hematologic toxicity occurred, the protocol was amended to evaluate three cohorts: gemcitabine and 17-AAG; 17-AAG and cisplatin; and gemcitabine, 17-AAG and cisplatin with modified dosing. RESULTS: The 39 patients enrolled were evaluable for toxicity and response. The MTD for cohort A was 154 mg/m(2) of 17-AAG, 750 mg/m(2) of gemcitabine, and 40 mg/m(2) of cisplatin. In cohort A, DLTs were observed at the higher dose level and included neutropenia, hyperbilirubinemia, dehydration, GGT elevation, hyponatremia, nausea, vomiting, and thrombocytopenia. The MTD for cohort C was 154 mg/m(2) of 17-AAG and 750 mg/m(2) of gemcitabine, with one DLT observed (alkaline phosphatase elevation) observed. In cohort C, DLTs of thrombocytopenia, fever and dyspnea were seen at the higher dose level. The remaining cohorts were closed to accrual due to toxicity. Six patients experienced partial responses. Mean Hsp90 levels were decreased and levels of Hsp70 were increased compared to baseline. CONCLUSIONS: 17-AAG in combination with gemcitabine and cisplatin demonstrated antitumor activity, but significant hematologic toxicities were encountered. 17-AAG combined with gemcitabine is tolerable and has demonstrated evidence of activity at the MTD. The recommended phase II dose is defined as 154 mg/m(2) of 17-AAG and 750 mg/m(2) of gemcitabine, and is currently being investigated in phase II studies in ovarian and pancreatic cancers. There is no recommended phase II dose for the cisplatin-containing combinations.


Asunto(s)
Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Benzoquinonas/uso terapéutico , Cisplatino/uso terapéutico , Desoxicitidina/análogos & derivados , Lactamas Macrocíclicas/uso terapéutico , Neoplasias/tratamiento farmacológico , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Benzoquinonas/efectos adversos , Biomarcadores de Tumor/metabolismo , Estudios de Cohortes , Desoxicitidina/uso terapéutico , Relación Dosis-Respuesta a Droga , Femenino , Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Lactamas Macrocíclicas/efectos adversos , Masculino , Persona de Mediana Edad , Neoplasias/enzimología , Proteínas Serina-Treonina Quinasas/metabolismo , Resultado del Tratamiento , Gemcitabina
3.
Mol Cell Biol ; 26(5): 1722-30, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16478993

RESUMEN

The hsp90 chaperoning pathway is a multiprotein system that is required for the production or activation of many cell regulatory proteins, including the progesterone receptor (PR). We report here the identity of GCUNC-45 as a novel modulator of PR chaperoning by hsp90. GCUNC-45, previously implicated in the activities of myosins, can interact in vivo and in vitro with both PR-A and PR-B and with hsp90. Overexpression and knockdown experiments show GCUNC-45 to be a positive factor in promoting PR function in the cell. GCUNC-45 binds to the ATP-binding domain of hsp90 to prevent the activation of its ATPase activity by the cochaperone Aha1. This effect limits PR chaperoning by hsp90, but this can be reversed by FKBP52, a cochaperone that is thought to act later in the pathway. These findings reveal a new cochaperone binding site near the N terminus of hsp90, add insight on the role of FKBP52, and identify GCUNC-45 as a novel regulator of the PR signaling pathway.


Asunto(s)
Proteínas HSP90 de Choque Térmico/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Receptores de Progesterona/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Células Cultivadas , Clonación Molecular , Citoplasma/metabolismo , Proteínas HSP90 de Choque Térmico/genética , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Humanos , Chaperonas Moleculares/metabolismo , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Receptores de Progesterona/genética , Transducción de Señal , Proteínas de Unión a Tacrolimus/metabolismo
4.
Mol Endocrinol ; 22(10): 2229-40, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18451092

RESUMEN

An involvement of molecular chaperones in the action and well-being of steroid receptors was recognized early in the molecular era of hormone research. However, this has continued to be a topic of much enquiry and some confusion. All steroid receptors associate with heat shock protein 90, the main character of a series of multiprotein chaperone complexes generally referred to as the "heat shock protein 90 chaperoning machine." Receptor association with chaperones occurs in an ordered, step-wise fashion and is necessary for the maintenance of unliganded receptor in a state ready to bind and respond to hormone. Chaperones additionally modulate how receptors respond to hormone and activate target genes. Although much is known about the participants in this chaperoning process and the consequences of chaperoning, many key questions remain unanswered, particularly those concerning molecular mechanisms, cellular dynamics, and the functions of an array of cochaperone proteins. Here, we point out several areas in need of investigation to encourage new ideas and participants in this burgeoning field.


Asunto(s)
Chaperonas Moleculares/metabolismo , Receptores de Esteroides/metabolismo , Animales , Humanos , Chaperonas Moleculares/genética , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Unión Proteica , Conformación Proteica , Pliegue de Proteína , Receptores de Esteroides/química , Receptores de Esteroides/genética
5.
Mol Cancer Ther ; 7(10): 3256-64, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18852129

RESUMEN

Benzoquinone ansamycin antibiotics such as geldanamycin (GA) bind to the NH(2)-terminal ATP-binding domain of heat shock protein (Hsp) 90 and inhibit its chaperone functions. Despite in vitro and in vivo studies indicating promising antitumor activity, derivatives of GA, including 17-allylaminogeldanamycin (17-AAG), have shown little clinical efficacy as single agents. Thus, combination studies of 17-AAG and several cancer chemotherapeutics, including cisplatin (CDDP), have begun. In colony-forming assays, the combination of CDDP and GA or 17-AAG was synergistic and caused increased apoptosis compared with each agent alone. One measurable response that results from treatment with Hsp90-targeted agents is the induction of a heat shock factor-1 (HSF-1) heat shock response. Treatment with GA + CDDP revealed that CDDP suppresses up-regulation of HSF-1 transcription, causing decreased levels of stress-inducible proteins such as Hsp27 and Hsp70. However, CDDP treatment did not prevent trimerization and nuclear localization of HSF-1 but inhibited DNA binding of HSF-1 as shown by chromatin immunoprecipitation. Melphalan, but not camptothecin, caused similar inhibition of GA-induced HSF-1-mediated Hsp70 up-regulation. 3-(4,5-Dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium salt cell survival assays revealed that deletion of Hsp70 caused increased sensitivity to GA (Hsp70(+/+) IC(50) = 63.7 +/- 14.9 nmol/L and Hsp70(-/-) IC(50) = 4.3 +/- 2.9 nmol/L), which confirmed that a stress response plays a critical role in decreasing GA sensitivity. Our results suggest that the synergy of GA + CDDP is due, in part, to CDDP-mediated abrogation of the heat shock response through inhibition of HSF-1 activity. Clinical modulation of the HSF-1-mediated heat shock response may enhance the efficacy of Hsp90-directed therapy.


Asunto(s)
Antineoplásicos/farmacología , Benzoquinonas/farmacología , Cisplatino/farmacología , Respuesta al Choque Térmico/efectos de los fármacos , Lactamas Macrocíclicas/farmacología , Camptotecina/farmacología , Línea Celular Tumoral , Cromatina/metabolismo , Proteínas de Unión al ADN/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Sinergismo Farmacológico , Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Factores de Transcripción del Choque Térmico , Humanos , Melfalán/farmacología , Unión Proteica/efectos de los fármacos , Factores de Transcripción/metabolismo , Regulación hacia Arriba/efectos de los fármacos
6.
Biochemistry ; 47(31): 8203-13, 2008 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-18620420

RESUMEN

The molecular chaperones Hsp90 and Hsp70 are highly regulated by various cochaperones that participate in the activation of steroid receptors. Here we study Tpr2 (also called DjC7), a TPR domain-containing type III J protein implicated in steroid receptor chaperoning. We propose that Tpr2 plays a role in the Hsp90-dependent chaperoning of the progesterone receptor (PR). Tpr2 overexpression or knockdown resulted in slight reductions in PR transcriptional activity in HeLa cells. Immunoprecipitation and pulldown experiments indicated that Tpr2 associates with Hsp90 and Hsp70 complexes, some of which also contain the PR. Tpr2 can bind Hsp90 and Hsp70 simultaneously, which is also a property of the cochaperone Hop. However, unlike Hop, Tpr2 binding to Hsp70 in the presence of Hsp90 is ATP-dependent, and Tpr2 cannot replace Hop in Hsp90 chaperoning in vitro or in vivo. While Tpr2 was not detected as a component of PR heterocomplexes in cell lysates, purified Tpr2 bound the PR readily. Surprisingly, Tpr2 replaced type I and II J proteins in the Hsp90-dependent chaperoning of the PR and the protein kinase, Chk1. Unlike other J proteins, Tpr2 promoted the accumulation of Hsp70 in PR heterocomplexes in the presence of Hsp90. Thus, Tpr2 has the potential to regulate PR chaperoning.


Asunto(s)
Proteínas del Choque Térmico HSP40/fisiología , Proteínas HSP90 de Choque Térmico/metabolismo , Adenosina Trifosfato/metabolismo , Proteínas del Choque Térmico HSP40/genética , Proteínas del Choque Térmico HSP40/metabolismo , Células HeLa , Proteínas de Choque Térmico , Humanos , Inmunoprecipitación , Modelos Biológicos , Chaperonas Moleculares , Unión Proteica , Interferencia de ARN , ARN Interferente Pequeño/genética , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Proteínas Recombinantes/metabolismo , Transcripción Genética
7.
Cancer Res ; 66(22): 10967-75, 2006 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-17108135

RESUMEN

17-Allylamino-demethoxygeldanamycin (17-AAG), currently in phase I and II clinical trials as an anticancer agent, binds to the ATP pocket of heat shock protein (Hsp90). This binding induces a cellular stress response that up-regulates many proteins including Hsp27, a member of the small heat shock protein family that has cytoprotective roles, including chaperoning of cellular proteins, regulation of apoptotic signaling, and modulation of oxidative stress. Therefore, we hypothesized that Hsp27 expression may affect cancer cell sensitivity to 17-AAG. In colony-forming assays, overexpression of Hsp27 increased cell resistance to 17-AAG whereas down-regulation of Hsp27 by siRNA increased sensitivity. Because Hsp27 is known to modulate levels of glutathione (GSH), we examined cellular levels of GSH and found that it was decreased in cells transfected with Hsp27 siRNA when compared with control siRNA. Treatment with buthionine sulfoximine, an inhibitor of GSH synthesis, also sensitized cells to 17-AAG. Conversely, treatment of Hsp27 siRNA-transfected cells with N-acetylcysteine, an antioxidant and GSH precursor, reversed their sensitivity to 17-AAG. A cell line selected for stable resistance to geldanamycin relative to parent cells showed increased Hsp27 expression. When these geldanamycin- and 17-AAG-resistant cells were transfected with Hsp27 siRNA, 17-AAG resistance was dramatically diminished. Our results suggest that Hsp27 up-regulation has a significant role in 17-AAG resistance, which may be mediated in part through GSH regulation. Clinical modulation of GSH may therefore enhance the efficacy of Hsp90-directed therapy.


Asunto(s)
Benzoquinonas/farmacología , Glutatión/metabolismo , Proteínas de Choque Térmico/biosíntesis , Lactamas Macrocíclicas/farmacología , Resistencia a Antineoplásicos , Ensayos de Selección de Medicamentos Antitumorales , Células HeLa , Proteínas de Choque Térmico/genética , Humanos , ARN Interferente Pequeño/genética , Regulación hacia Arriba
8.
Cell Stress Chaperones ; 12(4): 353-63, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-18229454

RESUMEN

Hsp90 is an abundant and highly conserved chaperone that functions at later stages of protein folding to maintain and regulate the activity of client proteins. Using a recently described in vitro system to fold a functional model kinase Chk1, we performed a side-by-side comparison of the Hsp90-dependent chaperoning of Chk1 to that of the progesterone receptor (PR) and show that these distinct types of clients have different chaperoning requirements. The less stable PR required more total chaperone protein(s) and p23, whereas Chk1 folding was critically dependent on Cdc37. When the 2 clients were reconstituted under identical conditions, each client folding was dose dependent for Hsp90 protein levels and was inhibited by geldanamycin. Using this tractable system, we found that Chk1 kinase folding was more effective if we used a type II Hsp40 cochaperone, whereas PR is chaperoned equally well with a type I or type II Hsp40. Additional dissection of Chk1-chaperone complexes and the resulting kinase activity suggests that kinase folding, like that previously shown for PR, is a dynamic, multistep process. Importantly, the cochaperones Hop and Cdc37 cooperate as the kinase transitions from immature Hsp70- to mature Hsp90-predominant complexes.


Asunto(s)
Proteínas HSP90 de Choque Térmico/metabolismo , Proteínas Quinasas/metabolismo , Receptores de Progesterona/metabolismo , Animales , Benzoquinonas/farmacología , Proteínas de Ciclo Celular/metabolismo , Chaperoninas/metabolismo , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Pollos , Proteínas del Choque Térmico HSP40/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas HSP90 de Choque Térmico/química , Proteínas de Homeodominio/metabolismo , Humanos , Lactamas Macrocíclicas/farmacología , Modelos Biológicos , Unión Proteica/efectos de los fármacos , Pliegue de Proteína , Transporte de Proteínas/efectos de los fármacos , Proteínas Supresoras de Tumor/metabolismo
9.
Clin Cancer Res ; 12(20 Pt 1): 6087-93, 2006 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-17062684

RESUMEN

PURPOSE: To determine the maximum tolerated dose (MTD), dose-limiting toxicity, and pharmacokinetics of 17-allylamino-demethoxy-geldanamycin (17-AAG) administered on days 1, 4, 8, and 11 every 21 days and to examine the effect of 17-AAG on the levels of chaperone and client proteins. EXPERIMENTAL DESIGN: A phase I dose escalating trial in patients with advanced solid tumors was done. Toxicity and tumor responses were evaluated by standard criteria. Pharmacokinetics were done and level of target proteins was measured at various points during cycle one. RESULTS: Thirteen patients were enrolled in the study. MTD was defined as 220 mg/m2. Dose-limiting toxicities were as follows: dehydration, diarrhea, hyperglycemia, and liver toxicity. At the MTD, the mean clearance of 17-AAG was 18.7 L/h/m2. There was a significant decrease in integrin-linked kinase at 6 hours after infusion on day 1 but not at 25 hours in peripheral blood mononuclear cells. Treatment with 17-AAG on day 1 significantly increased pretreatment levels of heat shock protein (HSP) 70 on day 4, which is consistent with the induction of a stress response. In vitro induction of a stress response and up-regulation of HSP70 resulted in an increased resistance to HSP90-targeted therapy in A549 cells. CONCLUSIONS: The MTD of 17-AAG on a twice-weekly schedule was 220 mg/m2. Treatment at this dose level resulted in significant changes of target proteins and also resulted in a prolonged increase in HSP70. This raises the possibility that HSP70 induction as part of the stress response may contribute to resistance to 17-AAG.


Asunto(s)
Antineoplásicos/toxicidad , Benzoquinonas/toxicidad , Lactamas Macrocíclicas/toxicidad , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Adulto , Antineoplásicos/administración & dosificación , Benzoquinonas/administración & dosificación , Proteínas HSP70 de Choque Térmico/efectos de los fármacos , Proteínas HSP70 de Choque Térmico/metabolismo , Humanos , Infusiones Intravenosas , Lactamas Macrocíclicas/administración & dosificación , Esperanza de Vida , Estadificación de Neoplasias , Selección de Paciente
10.
Structure ; 13(4): 579-90, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15837196

RESUMEN

Hsp90 is an abundant molecular chaperone involved in many biological systems. We report here the crystal structures of the unliganded and ADP bound fragments containing the N-terminal and middle domains of HtpG, an E. coli Hsp90. These domains are not connected through a flexible linker, as often portrayed in models, but are intimately associated with one another. The individual HtpG domains have similar folding to those of DNA gyrase B but assemble differently, suggesting somewhat different mechanisms for the ATPase superfamily. ADP binds to a subpocket of a large site that is jointly formed by the N-terminal and middle domains and induces conformational changes of the N-terminal domain. We speculate that this large pocket serves as a putative site for binding of client proteins/cochaperones. Modeling shows that ATP is not exposed to the molecular surface, thus implying that ATP activation of hsp90 chaperone activities is accomplished via conformational changes.


Asunto(s)
Adenosina Difosfato/metabolismo , Proteínas Bacterianas/química , Escherichia coli/química , Proteínas HSP90 de Choque Térmico/química , Secuencia de Aminoácidos , Proteínas Bacterianas/metabolismo , Secuencia de Bases , Cartilla de ADN , Proteínas HSP90 de Choque Térmico/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Conformación Proteica , Homología de Secuencia de Aminoácido
11.
J Clin Oncol ; 23(6): 1078-87, 2005 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-15718306

RESUMEN

PURPOSE: We determined the maximum-tolerated dose (MTD) and the dose-limiting toxicities (DLT) of 17-allylamino-17-demethoxygeldanamycin (17-AAG) when infused on days 1, 8, and 15 of a 28-day cycle in advanced solid tumor patients. We also characterized the pharmacokinetics of 17-AAG, its effect on chaperone and client proteins, and whether cytochrome P450 (CYP) 3A5 and NAD(P)H:quinone oxidoreductase 1 (NQO1) polymorphisms affected 17-AAG disposition or toxicity. PATIENTS AND METHODS: An accelerated titration design was used. Biomarkers were measured in peripheral-blood mononuclear cells (PBMCs) at baseline and on days 1 and 15, and pharmacokinetic analysis was performed on day 1 of cycle 1. CYP3A5*3 and NQO1*2 genotypes were determined and correlated with pharmacokinetics and toxicity. RESULTS: Twenty-one patients received 52 courses at 11 dose levels. DLTs at 431 mg/m(2) were grade 3 bilirubin (n = 1), AST (n = 1), anemia (n = 1), nausea (n = 1), vomiting (n = 1), and myalgias (n = 1). No tumor responses were seen. 17-AAG consistently increased heat shock protein (Hsp) 70 levels in PBMCs. At the MTD, the clearance and half-life (t(1/2)) of 17-AAG were 11.6 L/h/m(2) and 4.15 hours, respectively; whereas the active metabolite 17-aminogeldanamycin had a t(1/2) of 7.63 hours. The CYP3A5*3 and NQO1*2 polymorphisms were not associated with 17-AAG toxicity. The CYP3A5*3 polymorphism was associated with higher 17-AAG clearance. CONCLUSION: The MTD of weekly 17-AAG is 308 mg/m(2). 17-AAG induced Hsp70 in PBMCs, indicating that Hsp90 has been affected. Further evaluation of 17-AAG is ongoing using a twice-weekly regimen, and this schedule of 17-AAG is being tested in combination with chemotherapy.


Asunto(s)
Neoplasias/tratamiento farmacológico , Rifabutina/análogos & derivados , Rifabutina/administración & dosificación , Rifabutina/efectos adversos , Adulto , Anciano , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Benzoquinonas , Biomarcadores/análisis , Citocromo P-450 CYP3A , Sistema Enzimático del Citocromo P-450/genética , Esquema de Medicación , Femenino , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Lactamas Macrocíclicas , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , NAD(P)H Deshidrogenasa (Quinona)/genética , Polimorfismo Genético
12.
Sci STKE ; 2004(238): pl10, 2004 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-15213337

RESUMEN

The development of green fluorescent protein (GFP) technology combined with live cell microscopy techniques have revealed the dynamic properties of GFP-tagged proteins in the nucleus. The mobility of a GFP-tagged protein can be assessed using a quantitative photobleaching technique, fluorescence recovery after photobleaching (FRAP) analysis. FRAP experiments demonstrate that many nuclear proteins are highly mobile within the nucleus. However, the factors within the nucleus that regulate this mobility are not known. This is partly due to an absence of protocols that can be used to identify such nuclear mobility factors. We developed a novel in situ assay that combines a biochemical permeabilization and extraction procedure with a quantitative FRAP technique, a method we used to uncover a new functional role for molecular chaperones in the nuclear mobility of steroid receptors. This assay can readily be adapted to identify and characterize other nuclear mobility factors.


Asunto(s)
Núcleo Celular/química , Núcleo Celular/metabolismo , Transporte Activo de Núcleo Celular/fisiología , Adenocarcinoma/química , Adenocarcinoma/patología , Animales , Línea Celular , Línea Celular Tumoral , Fibroblastos/química , Fibroblastos/citología , Recuperación de Fluorescencia tras Fotoblanqueo/métodos , Proteínas Fluorescentes Verdes , Humanos , Proteínas Luminiscentes/biosíntesis , Proteínas Luminiscentes/metabolismo , Neoplasias Mamarias Animales/química , Neoplasias Mamarias Animales/patología , Ratones , Chaperonas Moleculares/química , Chaperonas Moleculares/metabolismo , Receptores de Glucocorticoides/biosíntesis , Proteínas Recombinantes de Fusión/biosíntesis
13.
Mol Endocrinol ; 17(12): 2543-53, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14551264

RESUMEN

To elucidate the earliest molecular steps in the activation of transcription by the progesterone receptor (PR), we investigated its activity in a cell-free transcription system utilizing chromatin templates. PR prepared as a ligand-free, recombinant protein failed to induce transcription on chromatin templates. However, transcriptional competence could be restored by coincubation with rabbit reticulocyte lysate (RRL). The interaction of PR with chaperones results in a receptor conformation competent to bind ligand and RRL contains abundant chaperone-mediated protein folding activity. Blocking this activity with the specific inhibitor geldanamycin inhibited receptor-dependent transcriptional activity. However, recombinant chaperones could not replace RRL in the restoration of transcriptional activity on chromatin templates, suggesting the presence of an additional activity in the lysate. Under chromatin assembly conditions, PR could bind naked DNA and RRL did not increase that binding. In contrast, PR bound to a chromatin template only poorly. Interestingly, RRL stimulated sequence-specific binding by PR to target sites in chromatin and the concomitant recruitment of the steroid receptor coactivator 1 to the promoter. Thus, our results indicate that a novel protein-mediated activity in RRL is involved in an additional, heretofore unrecognized, activation step required for PR to become transcriptionally competent on chromatin templates.


Asunto(s)
Cromatina/genética , Receptores de Progesterona/genética , Transcripción Genética/genética , Animales , Secuencia de Bases , ADN/genética , ADN/metabolismo , Drosophila/embriología , Embrión no Mamífero/fisiología , Humanos , Progesterona/metabolismo , Conejos , Proteínas Recombinantes/metabolismo , Reticulocitos/metabolismo , Moldes Genéticos
14.
Pharmacogenetics ; 13(9): 555-64, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12972954

RESUMEN

Thiopurine S-methyltransferase (TPMT) catalyses the S-methylation of thiopurine drugs such as 6-mercaptopurine. A common genetic polymorphism for TPMT is associated with large individual variations in thiopurine drug toxicity and therapeutic efficacy. TPMT*3A, the most common variant allele in Caucasians, has two alterations in amino acid sequence, resulting in striking decreases in TPMT protein levels. This phenomenon results, in part, from rapid degradation through a ubiquitin-proteasome-mediated process. We set out to test the hypothesis that chaperone proteins might be involved in targeting TPMT for degradation. As a first step, hsp90, hsp70 and the cochaperone hop were immunoprecipitated from a rabbit reticulocyte lysate (RRL) that included radioactively labelled *3A and wild-type TPMT. TPMT*3A was much more highly associated with all three chaperones than was the wild-type enzyme. The RRL was also used to confirm the accelerated degradation of *3A compared to wild-type TPMT. Treatment of RRL with the hsp90 inhibitor geldanamycin resulted in enhanced association of hsp90 with wild-type TPMT, an observation that correlated with accelerated ubiquitin-dependent degradation of wild-type TPMT. Geldanamycin treatment of COS-1 cells transfected with FLAG-tagged wild-type also resulted in a time and geldanamycin concentration-dependent decrease in TPMT activity and protein, which was compatible with results obtained in the RRL. These observations indicate that TPMT is a client protein for hsp90 and suggest that chaperone proteins, especially hsp90, are involved in targeting both TPMT*3A and, in the presence of geldanamycin, the wild-type allozyme for degradation. Therefore, chaperone proteins play an important mechanistic role in this clinically significant example of pharmacogenetic variation in drug metabolism.


Asunto(s)
Proteínas de Choque Térmico/metabolismo , Isoenzimas/metabolismo , Metiltransferasas/genética , Farmacogenética , Alelos , Animales , Benzoquinonas , Células COS , Chlorocebus aethiops , Inhibidores Enzimáticos/farmacología , Variación Genética , Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas HSP90 de Choque Térmico/efectos de los fármacos , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Lactamas Macrocíclicas , Metiltransferasas/metabolismo , Polimorfismo Genético , Quinonas/farmacología , Conejos , Reticulocitos/química , Población Blanca
15.
Cell Stress Chaperones ; 8(2): 108-13, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14627195

RESUMEN

p23 is a small but important cochaperone for the Hsp90 chaperoning pathway. It appears to facilitate the adenosine triphosphate-driven cycle of Hsp90 binding to client proteins. It enters at a late stage of the cycle and enhances the maturation of client proteins. Although this role of p23 is fairly well established, recent studies suggest that it may have additional functions in the cell that merit further exploration.


Asunto(s)
Chaperonas Moleculares/metabolismo , Fosfoproteínas/metabolismo , Animales , Regulación Fúngica de la Expresión Génica , Proteínas HSP90 de Choque Térmico/metabolismo , Oxidorreductasas Intramoleculares , Mamíferos/genética , Mamíferos/metabolismo , Chaperonas Moleculares/genética , Fosfoproteínas/genética , Prostaglandina-E Sintasas , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo
16.
Cell Stress Chaperones ; 7(1): 55-64, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11894840

RESUMEN

In the ligand-binding inactive state, the steroid receptor heterocomplex contains Hsp90, Hsp70, high-molecular weight immunophilins, and other proteins. Hsp90 acts in association with co-chaperones to maintain the native state of the receptor within the cells. It was reported earlier that Hsp90 might not be as important for the androgen receptor (AR) activity as for the glucocorticoid receptor (GR) and the progesterone receptor (PR) activities. We used the Hsp90 inhibitor geldanamycin (GA) to explore the role of Hsp90 in the function of the AR heterocomplex. GA selectively binds to Hsp90 and inhibits its activity, leading to the loss of steroid receptor activity, and frequently, its degradation. In our study, LNCaP prostate cancer cells were treated with GA for 30 minutes or 24 hours, in the presence of mibolerone, a synthetic androgen. GA reduced the androgen-induced AR protein levels to 15% after 24 hours of treatment. Several androgen up-regulated genes, including immunophilin FKBP51 and prostate specific antigen (PSA), were reduced by GA treatment. In cells treated with GA after transfection with a PSA promoter or an androgen response element-driven reporter gene, AR-mediated transactivation of reporter gene expression was reversibly inhibited by GA. Loss of androgen-binding ability and AR levels was attributed to reduced transcription of AR-regulated gene expression. Degradation rate of 35S-labeled AR was significantly increased by GA in the presence or absence of mibolerone. GA induced the degradation of AR through the proteasomal pathway. AR in cells treated with proteasomal inhibitor lactacystin, was insoluble in Nonidet P-40 (NP40)-based buffer and could not restore the androgen-binding ability. We report here that GA treatment disrupted both hormone-binding activity and receptor protein stability, resulting in a dramatic loss of androgen-induced gene activation. These results show that Hsp90 activity is important for both the chaperone-mediated folding of the AR into a high-affinity ligand-binding conformation and the functional activity of the AR.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Neoplasias de la Próstata , Quinonas/farmacología , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Benzoquinonas , División Celular/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Humanos , Lactamas Macrocíclicas , Ligandos , Masculino , Antígeno Prostático Específico/genética , ARN Mensajero/análisis , Proteínas de Unión a Tacrolimus/metabolismo , Células Tumorales Cultivadas/efectos de los fármacos
17.
Cell Stress Chaperones ; 9(2): 167-81, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15497503

RESUMEN

The high-affinity ligand-binding form of unactivated steroid receptors exists as a multicomponent complex that includes heat shock protein (Hsp)90; one of the immunophilins cyclophilin 40 (CyP40), FKBP51, or FKBP52; and an additional p23 protein component. Assembly of this heterocomplex is mediated by Hsp70 in association with accessory chaperones Hsp40, Hip, and Hop. A conserved structural element incorporating a tetratricopeptide repeat (TPR) domain mediates the interaction of the immunophilins with Hsp90 by accommodating the C-terminal EEVD peptide of the chaperone through a network of electrostatic and hydrophobic interactions. TPR cochaperones recognize the EEVD structural motif common to both Hsp90 and Hsp70 through a highly conserved clamp domain. In the present study, we investigated in vitro the molecular interactions between CyP40 and FKBP52 and other stress-related components involved in steroid receptor assembly, namely Hsp70 and Hop. Using a binding protein-retention assay with CyP40 fused to glutathione S-transferase immobilized on glutathione-agarose, we have identified the constitutively expressed form of Hsp70, heat shock cognate (Hsc)70, as an additional target for CyP40. Deletion mapping studies showed the binding determinants to be similar to those for CyP40-Hsp90 interaction. Furthermore, a mutational analysis of CyP40 clamp domain residues confirmed the importance of this motif in CyP40-Hsc70 interaction. Additional residues thought to mediate binding specificity through hydrophobic interactions were also important for Hsc70 recognition. CyP40 was shown to have a preference for Hsp90 over Hsc70. Surprisingly, FKBP52 was unable to compete with CyP40 for Hsc70 binding, suggesting that FKBP52 discriminates between the TPR cochaperone-binding sites in Hsp90 and Hsp70. Hop, which contains multiple units of the TPR motif, was shown to be a direct competitor with CyP40 for Hsc70 binding. Similar to Hop, CyP40 was shown not to influence the adenosine triphosphatase activity of Hsc70. Our results suggest that CyP40 may have a modulating role in Hsc70 as well as Hsp90 cellular function.


Asunto(s)
Ciclofilinas/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Chaperonas Moleculares/metabolismo , Adenosina Trifosfatasas/análisis , Adenosina Trifosfatasas/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Sitios de Unión/genética , Unión Competitiva , Proteínas Portadoras/metabolismo , Mapeo Cromosómico , Peptidil-Prolil Isomerasa F , Ciclofilinas/química , Análisis Mutacional de ADN , Proteínas de Drosophila , Eliminación de Gen , Glutatión Transferasa/metabolismo , Proteínas del Choque Térmico HSP40 , Proteínas HSP70 de Choque Térmico/química , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP90 de Choque Térmico/química , Proteínas de Choque Térmico/metabolismo , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Quinasas Janus , Datos de Secuencia Molecular , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Tirosina Quinasas/química , Proteínas Tirosina Quinasas/metabolismo , Ratas , Proteínas Recombinantes de Fusión/metabolismo , Secuencias Repetitivas de Ácidos Nucleicos , Homología de Secuencia de Aminoácido , Electricidad Estática , Proteínas de Unión a Tacrolimus/metabolismo , Factores de Transcripción , Proteínas Supresoras de Tumor/metabolismo
18.
Exp Biol Med (Maywood) ; 228(2): 111-33, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12563018

RESUMEN

Nearly 100 proteins are known to be regulated by hsp90. Most of these substrates or "client proteins" are involved in signal transduction, and they are brought into complex with hsp90 by a multiprotein hsp90/hsp70-based chaperone machinery. In addition to binding substrate proteins at the chaperone site(s), hsp90 binds cofactors at other sites that are part of the heterocomplex assembly machinery as well as immunophilins that connect assembled substrate*hsp90 complexes to protein-trafficking systems. In the 5 years since we last reviewed this subject, much has been learned about hsp90 structure, nucleotide-binding, and cochaperone interactions; the most important concept is that ATP hydrolysis by an intrinsic ATPase activity results in a conformational change in hsp90 that is required to induce conformational change in a substrate protein. The conformational change induced in steroid receptors is an opening of the steroid-binding cleft so that it can be accessed by steroid. We have now developed a minimal system of five purified proteins-hsp90, hsp70, Hop, hsp40, and p23- that assembles stable receptor*hsp90 heterocomplexes. An hsp90*Hop*hsp70*hsp40 complex opens the cleft in an ATP-dependent process to produce a receptor*hsp90 heterocomplex with hsp90 in its ATP-bound conformation, and p23 then interacts with the hsp90 to stabilize the complex. Stepwise assembly experiments have shown that hsp70 and hsp40 first interact with the receptor in an ATP-dependent reaction to produce a receptor*hsp70*hsp40 complex that is "primed" to be activated to the steroid-binding state in a second ATP-dependent step with hsp90, Hop, and p23. Successful use of the five-protein system with other substrates indicates that it can assemble signal protein*hsp90 heterocomplexes whether the substrate is a receptor, a protein kinase, or a transcription factor. This purified system should facilitate understanding of how eukaryotic hsp70 and hsp90 work together as essential components of a process that alters the conformations of substrate proteins to states that respond in signal transduction.


Asunto(s)
Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Chaperonas Moleculares/metabolismo , Transporte de Proteínas/fisiología , Transducción de Señal/fisiología , Animales , Proteínas de Drosophila , Humanos , Inmunofilinas/metabolismo , Quinasas Janus , Sustancias Macromoleculares , Modelos Biológicos , Complejos Multiproteicos , Nucleótidos/metabolismo , Unión Proteica , Proteínas Tirosina Quinasas/metabolismo , Receptores de Superficie Celular/metabolismo , Relación Estructura-Actividad , Factores de Transcripción/metabolismo
20.
Mol Cell Biol ; 30(5): 1243-53, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20048054

RESUMEN

Prostate cancer progression to the androgen-independent (AI) state involves acquisition of pathways that allow tumor growth under low-androgen conditions. We hypothesized that expression of molecular chaperones that modulate androgen binding to AR might be altered in prostate cancer and contribute to progression to the AI state. Here, we report that the Hsp90 cochaperone FKBP51 is upregulated in LAPC-4 AI tumors grown in castrated mice and describe a molecular mechanism by which FKBP51 regulates AR activity. Using recombinant proteins, we show that FKBP51 stimulates recruitment of the cochaperone p23 to the ATP-bound form of Hsp90, forming an FKBP51-Hsp90-p23 superchaperone complex. In cells, FKBP51 expression promotes superchaperone complex association with AR and increases the number of AR molecules that undergo androgen binding. FKBP51 stimulates androgen-dependent transcription and cell growth, and FKBP51 is part of a positive feedback loop that is regulated by AR and androgen. Finally, depleting FKBP51 levels by short hairpin RNA reduces the transcript levels of genes regulated by AR and androgen. Because the superchaperone complex plays a critical role in determining the ligand-binding competence and transcription function of AR, it provides an attractive target for inhibiting AR activity in prostate cancer cells.


Asunto(s)
Proteínas HSP90 de Choque Térmico/metabolismo , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/metabolismo , Proteínas de Unión a Tacrolimus/metabolismo , Animales , Secuencia de Bases , Línea Celular Tumoral , Cartilla de ADN/genética , Proteínas HSP90 de Choque Térmico/genética , Humanos , Oxidorreductasas Intramoleculares/genética , Oxidorreductasas Intramoleculares/metabolismo , Masculino , Ratones , Ratones SCID , Modelos Biológicos , Complejos Multiproteicos , Trasplante de Neoplasias , Neoplasias Hormono-Dependientes/genética , Neoplasias Hormono-Dependientes/metabolismo , Prostaglandina-E Sintasas , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Receptores Androgénicos/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal , Proteínas de Unión a Tacrolimus/genética , Trasplante Heterólogo , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA