Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cancer Med ; 8(5): 2449-2461, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30972950

RESUMEN

BACKGROUND: Vitamin D3 (VitD) deficiency is linked to increased incidence and worse survival in bladder cancer (BCa). In addition to cystectomy, patients are treated with cisplatin-based chemotherapy, however 30%-50% of patients do not benefit from this treatment. The effects of VitD deficiency on response to chemotherapy remain unknown. METHODS: To test effects of VitD supplementation on the response to cisplatin we analyzed patient serum VitD levels and correlated that with survival. In vivo, VitD deficient mice were treated with cisplatin, with or without pretreatment with the active VitD metabolite, 1,25 dihydroxyvitamin D3 (1,25D3 ). Lastly, using BCa cell lines, T24 and RT-112, the mechanism of action of 1,25D3 and cisplatin combination treatment was determined by apoptosis assays, as well as western blot and RT-PCR. RESULTS: In this study, we determined that low serum 25 hydroxyvitamin D3 (25D3 ) levels was significantly associated with worse response to cisplatin. Pretreating deficient mice with 1,25D3 , reduced tumor volume compared to cisplatin monotherapy. In vitro, 1,25D3 pretreatment increased the apoptotic response to cisplatin. 1,25D3 pretreatment increased expression of TAp73 and its pro-apoptotic targets, in a VDR dependent manner. VDR and its transcriptional targets were induced after 1,25D3 treatment and further increased after the combination of 1,25D3 and cisplatin in a TAp73 dependent manner. CONCLUSIONS: Our data suggest that VitD deficiency could be a biomarker for poor response to cisplatin, and pretreating with VitD can increase the apoptotic response to cisplatin through VDR and TAp73 signaling crosstalk.


Asunto(s)
Colecalciferol/farmacología , Cisplatino/farmacología , Receptores de Calcitriol/metabolismo , Transducción de Señal/efectos de los fármacos , Proteína Tumoral p73/metabolismo , Neoplasias de la Vejiga Urinaria/metabolismo , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Femenino , Expresión Génica , Humanos , Inmunohistoquímica , Ratones , Modelos Biológicos , Pronóstico , Receptores de Calcitriol/genética , Proteína Tumoral p73/genética , Neoplasias de la Vejiga Urinaria/etiología , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/mortalidad , Deficiencia de Vitamina D/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
2.
J Clin Invest ; 128(1): 323-340, 2018 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-29202474

RESUMEN

Metabolic reprogramming in breast tumors is linked to increases in putative oncogenic metabolites that may contribute to malignant transformation. We previously showed that accumulation of the oncometabolite, 2-hydroxyglutarate (2HG), in breast tumors was associated with MYC signaling, but not with isocitrate dehydrogenase (IDH) mutations, suggesting a distinct mechanism for increased 2HG in breast cancer. Here, we determined that D-2HG is the predominant enantiomer in human breast tumors and show that the D-2HG-producing mitochondrial enzyme, alcohol dehydrogenase, iron-containing protein 1 (ADHFE1), is a breast cancer oncogene that decreases patient survival. We found that MYC upregulates ADHFE1 through changes in iron metabolism while coexpression of both ADHFE1 and MYC strongly enhanced orthotopic tumor growth in MCF7 cells. Moreover, ADHFE1 promoted metabolic reprogramming with increased formation of D-2HG and reactive oxygen, a reductive glutamine metabolism, and modifications of the epigenetic landscape, leading to cellular dedifferentiation, enhanced mesenchymal transition, and phenocopying alterations that occur with high D-2HG levels in cancer cells with IDH mutations. Together, our data support the hypothesis that ADHFE1 and MYC signaling contribute to D-2HG accumulation in breast tumors and show that D-2HG is an oncogenic metabolite and potential driver of disease progression.


Asunto(s)
Oxidorreductasas de Alcohol/metabolismo , Neoplasias de la Mama/metabolismo , Desdiferenciación Celular , Reprogramación Celular , Proteínas Mitocondriales/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Transducción de Señal , Oxidorreductasas de Alcohol/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Femenino , Glutaratos/metabolismo , Humanos , Células MCF-7 , Proteínas Mitocondriales/genética , Proteínas Proto-Oncogénicas c-myc/genética , Especies Reactivas de Oxígeno/metabolismo
3.
Nat Commun ; 8(1): 1654, 2017 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-29162831

RESUMEN

Cisplatin chemotherapy causes permanent hearing loss in 40-80% of treated patients. It is unclear whether the cochlea has unique sensitivity to cisplatin or is exposed to higher levels of the drug. Here we use inductively coupled plasma mass spectrometry (ICP-MS) to examine cisplatin pharmacokinetics in the cochleae of mice and humans. In most organs cisplatin is detected within one hour after injection, and is eliminated over the following days to weeks. In contrast, the cochlea retains cisplatin for months to years after treatment in both mice and humans. Using laser ablation coupled to ICP-MS, we map cisplatin distribution within the human cochlea. Cisplatin accumulation is consistently high in the stria vascularis, the region of the cochlea that maintains the ionic composition of endolymph. Our results demonstrate long-term retention of cisplatin in the human cochlea, and they point to the stria vascularis as an important therapeutic target for preventing cisplatin ototoxicity.


Asunto(s)
Antineoplásicos/efectos adversos , Cisplatino/efectos adversos , Cóclea/química , Neoplasias/tratamiento farmacológico , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/análisis , Antineoplásicos/metabolismo , Cisplatino/administración & dosificación , Cisplatino/análisis , Cisplatino/metabolismo , Cóclea/metabolismo , Cóclea/fisiopatología , Femenino , Pérdida Auditiva/etiología , Pérdida Auditiva/metabolismo , Pérdida Auditiva/fisiopatología , Humanos , Masculino , Espectrometría de Masas , Ratones Endogámicos CBA , Estría Vascular/química , Estría Vascular/metabolismo
4.
Clin Cancer Res ; 23(6): 1397-1406, 2017 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-27663600

RESUMEN

Purpose: Our preclinical studies showed that the PARP inhibitor, olaparib, prior to carboplatin attenuated carboplatin cytotoxicity. We evaluated sequence-specific pharmacokinetic and pharmacodynamic effects, safety, and activity of the combination.Experimental Design: Eligible patients had metastatic or recurrent women's cancer. Olaparib tablets were introduced (100 or 200 mg twice daily, days 1-7) in a 3 + 3 dose escalation with carboplatin AUC4 or 5 every 21 days, up to eight cycles, followed by olaparib 300 mg twice daily maintenance. Patients were randomly assigned to starting schedule: cohort A (olaparib days 1-7, carboplatin on day 8) or B (carboplatin on day 1, olaparib days 2-8) during cycle 1. Patients received the reversed scheme in cycle 2. Blood was collected for olaparib pharmacokinetics, platinum-DNA adducts, comet assay, and PAR concentrations. The primary objectives were to examine schedule-dependent effects on olaparib pharmacokinetics and platinum-DNA adducts.Results: A total of 77 (60 ovarian, 14 breast, and 3 uterine cancer) patients were treated. Dose-limiting toxicity was thrombocytopenia and neutropenia, defining olaparib 200 mg twice daily + carboplatin AUC4 as the MTD. Olaparib clearance was increased approximately 50% when carboplatin was given 24 hours before olaparib. In vitro experiments demonstrated carboplatin preexposure increased olaparib clearance due to intracellular olaparib uptake. Quantities of platinum-DNA adducts were not different as a function of the order of drug administration. Responses included 2 CRs and 31 PRs (46%) with a higher RR in BRCA mutation carriers compared with nonmutation carriers (68% vs. 19%).Conclusions: Tablet olaparib with carboplatin is a safe and active combination. Carboplatin preexposure causes intracellular olaparib accumulation reducing bioavailable olaparib, suggesting carboplatin should be administered prior to olaparib. Clin Cancer Res; 23(6); 1397-406. ©2016 AACR.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Carboplatino/administración & dosificación , Neoplasias Ováricas/tratamiento farmacológico , Ftalazinas/administración & dosificación , Piperazinas/administración & dosificación , Neoplasias Uterinas/tratamiento farmacológico , Adulto , Anciano , Neoplasias de la Mama/sangre , Neoplasias de la Mama/patología , Carboplatino/efectos adversos , Carboplatino/farmacocinética , Aductos de ADN/sangre , Esquema de Medicación , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/sangre , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/patología , Femenino , Humanos , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/patología , Dosis Máxima Tolerada , Persona de Mediana Edad , Recurrencia Local de Neoplasia/sangre , Recurrencia Local de Neoplasia/patología , Neoplasias Ováricas/sangre , Neoplasias Ováricas/patología , Ftalazinas/efectos adversos , Ftalazinas/farmacocinética , Piperazinas/efectos adversos , Piperazinas/farmacocinética , Neoplasias Uterinas/sangre , Neoplasias Uterinas/patología
5.
Oncotarget ; 7(11): 12447-63, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26799670

RESUMEN

The protein phosphatase 2A (PP2A) inhibitor, LB100, has been shown in pre-clinical studies to be an effective chemo- and radio-sensitizer for treatment of various cancers. We investigated effects associated with LB100 treatment alone and in combination with cisplatin for medulloblastoma (MB) in vitro and in vivo in an intracranial xenograft model. We demonstrated that LB100 had a potent effect on MB cells. By itself, LB100 inhibited proliferation and induced significant apoptosis in a range of pediatric MB cell lines. It also attenuated MB cell migration, a pre-requirement for invasion. When used in combination, LB100 enhanced cisplatin-mediated cytotoxic effects. Cell viability in the presence of 1 uM cisplatin alone was 61% (DAOY), 100% (D341), and 58% (D283), but decreased with the addition of 2 µM of LB100 to 26% (DAOY), 67% (D341), and 27% (D283), (p < 0.005). LB100 suppressed phosphorylation of the STAT3 protein and several STAT3 downstream targets. Also, LB100 directly increased cisplatin uptake and overcame cisplatin-resistance in vitro. Finally, LB100 exhibited potent in vivo anti-neoplastic activity in combination with cisplatin in an intracranial xenograft model.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Neoplasias Cerebelosas/tratamiento farmacológico , Cisplatino/farmacología , Meduloblastoma/tratamiento farmacológico , Piperazinas/farmacología , Proteína Fosfatasa 2/antagonistas & inhibidores , Animales , Compuestos Bicíclicos Heterocíclicos con Puentes/administración & dosificación , Línea Celular Tumoral , Neoplasias Cerebelosas/enzimología , Neoplasias Cerebelosas/patología , Cisplatino/administración & dosificación , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Humanos , Meduloblastoma/enzimología , Meduloblastoma/patología , Ratones , Ratones SCID , Piperazinas/administración & dosificación , Proteína Fosfatasa 2/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Pharmacol Res ; 106: 27-36, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26804248

RESUMEN

Cisplatin is one of the most commonly used chemotherapy drugs, treating a wide range of cancer types. Unfortunately, many cancers initially respond to platinum treatment but when the tumor returns, drug resistance frequently occurs. Resistance to cisplatin is attributed to three molecular mechanisms: increased DNA repair, altered cellular accumulation, and increased drug inactivation. The use of precision medicine to make informed decisions on a patient's cisplatin resistance status and predicting the tumor response would allow the clinician to tailor the chemotherapy program based on the biology of the disease. In this review, key biomarkers of each molecular mechanism will be discussed along with the current clinical research. Additionally, known polymorphisms for each biomarker will be discussed in relation to their influence on cisplatin resistance.


Asunto(s)
Antineoplásicos/efectos adversos , Antineoplásicos/uso terapéutico , Cisplatino/efectos adversos , Cisplatino/uso terapéutico , Resistencia a Antineoplásicos/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Medicina de Precisión/métodos , Humanos
7.
Oncol Rep ; 32(2): 469-74, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24926795

RESUMEN

Cellular resistance to platinum anticancer compounds is governed by no less than two molecular processes; DNA repair and cellular accumulation of drug. Gli1 is an upstream regulator of nucleotide excision repair, effecting this process through c-jun. We, therefore, investigated whether Gli1 plays a role in cellular accumulation of cisplatin. Using a Gli1-specific shRNA, we explored the role of Gli1 in the cellular accumulation and efflux of cisplatin, in cisplatin-resistant A2780-CP70 human ovarian cancer cells. When Gli1 is inhibited, cellular uptake of cisplatin was approximately 33% of the level of uptake under control conditions. When Gli1 is inhibited, cellular efflux of cisplatin was completely abrogated, over a 12-h period of observation. We assayed nuclear lysates from these cells, for the ability to bind the DNA sequence that is the Gli-binding site (GBS) in the 5'UTR for each of five known cisplatin transmembrane transporters. Four of these transporters are active in cisplatin uptake; and, one is active in cisplatin efflux. In each case, nuclear lysate from A2780-CP70 cells binds the GBS of the respective cisplatin transport gene. We conclude that Gli1 plays a strong role in total cellular accumulation of cisplatin in these cells; and, that the combined effects on cellular accumulation of drug and on DNA repair may indicate a role for Gli1 in protecting cellular DNA from lethal types of DNA damage.


Asunto(s)
Antineoplásicos/farmacocinética , Cisplatino/farmacocinética , Resistencia a Antineoplásicos , Neoplasias Ováricas/metabolismo , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/metabolismo , Sitios de Unión , Proteínas de Transporte de Catión/metabolismo , Línea Celular Tumoral , Transportador de Cobre 1 , Daño del ADN , Femenino , Humanos , Proteínas de Transporte de Catión Orgánico/metabolismo , Proteína con Dedos de Zinc GLI1
8.
Int J Oncol ; 44(3): 655-61, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24366538

RESUMEN

The Hedgehog pathway is molecularly linked to increased resistance to cisplatin and increased repair of platinum-DNA damage, through C-JUN. GLI1, which has five known isoforms, is a positive transcriptional regulator in Hedgehog. Southwestern blot assay, EMSA and ChIP assays indicate that only one of five isoforms of GLI1 may be responsible for the Hedgehog link with C-JUN and thus, increased platinum-DNA adduct repair. Cancer tissues express this 130-kDa isoform at levels 6-fold higher than non-malignant tissues; and this isoform exists in abundance in six of seven ovarian cancer cell lines examined.


Asunto(s)
Resistencia a Antineoplásicos/genética , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Neoplasias Ováricas/genética , Factores de Transcripción/genética , Cisplatino/uso terapéutico , Daño del ADN/efectos de los fármacos , Femenino , Proteínas Hedgehog/metabolismo , Humanos , Neoplasias Ováricas/tratamiento farmacológico , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Factores de Transcripción/metabolismo , Células Tumorales Cultivadas , Proteína con Dedos de Zinc GLI1
9.
PLoS One ; 7(8): e41711, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22876290

RESUMEN

Receptor desensitization is a ubiquitous regulatory mechanism that defines the activatable pool of receptors, and thus, the ability of cells to respond to environmental stimuli. In recent years, the molecular mechanisms controlling the desensitization of a variety of receptors have been established. However, little is known about the molecular mechanisms that underlie desensitization of natriuretic peptide receptors, including natriuretic peptide receptor-A (NPR-A). Here we report that calcineurin (protein phosphatase 2B, PP2B, PPP3C) regulates homologous desensitization of NPR-A in murine Leydig tumor (MA-10) cells. We demonstrate that both pharmacological inhibition of calcineurin activity and siRNA-mediated suppression of calcineurin expression potentiate atrial natriuretic peptide (ANP)-induced cGMP synthesis. Treatment of MA-10 cells with inhibitors of other phosphoprotein phosphatases had little or no effect on ANP-induced cGMP accumulation. In addition, overexpression of calcineurin blunts ANP-induced cGMP synthesis. We also present data indicating that the inhibition of calcineurin potentiates ANP-induced testosterone production. To better understand the contribution of calcineurin in the regulation of NPR-A activity, we examined the kinetics of ANP-induced cGMP signals. We observed transient ANP-induced cGMP signals, even in the presence of phosphodiesterase inhibitors. Inhibition of both calcineurin and phosphodiesterase dramatically slowed the decay in the response. These observations are consistent with a model in which calcineurin mediated dephosphorylation and desensitization of NPR-A is associated with significant inhibition of cGMP synthesis. PDE activity hydrolyzes cGMP, thus lowering intracellular cGMP toward the basal level. Taken together, these data suggest that calcineurin plays a previously unrecognized role in the desensitization of NPR-A and, thereby, inhibits ANP-mediated increases in testosterone production.


Asunto(s)
Factor Natriurético Atrial/farmacología , Calcineurina/metabolismo , Receptores del Factor Natriurético Atrial/metabolismo , Testosterona/biosíntesis , Animales , Calcineurina/genética , Línea Celular Tumoral , Simulación por Computador , GMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/metabolismo , Genes Reporteros , Humanos , Cinética , Masculino , Ratones , Modelos Teóricos , Factores de Transcripción NFATC/genética , Fosfoproteínas Fosfatasas/metabolismo , Fosforilación , Interferencia de ARN , Transducción de Señal/efectos de los fármacos
10.
J Biol Chem ; 286(47): 40413-22, 2011 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-21921034

RESUMEN

PP5 is a ubiquitously expressed Ser/Thr protein phosphatase. High levels of PP5 have been observed in human cancers, and constitutive PP5 overexpression aids tumor progression in mouse models of tumor development. However, PP5 is highly conserved among species, and the roles of PP5 in normal tissues are not clear. Here, to help evaluate the biological actions of PP5, a Cre/loxP-conditional mouse line was generated. In marked contrast to the early embryonic lethality associated with the genetic disruption of other PPP family phosphatases (e.g. PP2A and PP4), intercrosses with mouse lines that ubiquitously express Cre recombinase starting early in development (e.g. MeuCre40 and ACTB-Cre) produced viable and fertile PP5-deficient mice. Phenotypic differences caused by the total disruption of PP5 were minor, suggesting that small molecule inhibitors of PP5 will not have widespread systemic toxicity. Examination of roles for PP5 in fibroblasts generated from PP5-deficient embryos (PP5(-/-) mouse embryonic fibroblasts) confirmed some known roles and identified new actions for PP5. PP5(-/-) mouse embryonic fibroblasts demonstrated increased sensitivity to UV light, hydroxyurea, and camptothecin, which are known activators of ATR (ataxia-telangiectasia and Rad3-related) kinase. Further study revealed a previously unrecognized role for PP5 downstream of ATR activation in a UV light-induced response. The genetic disruption of PP5 is associated with enhanced and prolonged phosphorylation of a single serine (Ser-345) on Chk1, increased phosphorylation of the p53 tumor suppressor protein (p53) at serine 18, and increased p53 protein levels. A comparable role for PP5 in the regulation of Chk1 phosphorylation was also observed in human cells.


Asunto(s)
Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Fosfoproteínas Fosfatasas/deficiencia , Fosfoproteínas Fosfatasas/genética , Proteínas Quinasas/metabolismo , Rayos Ultravioleta , Actinas/genética , Alelos , Animales , Blastocisto/metabolismo , Cruzamiento , Línea Celular , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/efectos de la radiación , Células HeLa , Humanos , Hidroxiurea/farmacología , Integrasas/genética , Masculino , Ratones , Proteínas Nucleares/metabolismo , Fenotipo , Fosfoproteínas Fosfatasas/metabolismo , Fosforilación/efectos de los fármacos , Fosforilación/genética , Fosforilación/efectos de la radiación , Regiones Promotoras Genéticas/genética , Proteínas Quinasas/química , Receptores de Neuropéptido Y/metabolismo , Serina/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Fosfatasas cdc25/metabolismo
11.
J Pharmacol Exp Ther ; 331(1): 45-53, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19592665

RESUMEN

Fostriecin and cytostatin are structurally related natural inhibitors of serine/threonine phosphatases, with promising antitumor activity. The total synthesis of these antitumor agents has enabled the production of structural analogs, which are useful to explore the biological significance of features contained in the parent compounds. Here, the inhibitory activity of fostriecin, cytostatin, and 10 key structural analogs were tested in side-by-side phosphatase assays to further characterize their inhibitory activity against PP1c (Ser/Thr protein phosphatase 1 catalytic subunit), PP2Ac (Ser/Thr protein phosphatase 2A catalytic subunit), PP5c (Ser/Thr protein phosphatase 5 catalytic subunit), and chimeras of PP1 (Ser/Thr protein phosphatase 1) and PP5 (Ser/Thr protein phosphatase 5), in which key residues predicted for inhibitor contact with PP2A (Ser/Thr protein phosphatase 2A) were introduced into PP1 and PP5 using site-directed mutagenesis. The data confirm the importance of the C9-phosphate and C11-alcohol for general inhibition and further demonstrate the importance of a predicted C3 interaction with a unique cysteine (Cys(269)) in the beta12-beta13 loop of PP2A. The data also indicate that additional features beyond the unsaturated lactone contribute to inhibitory potency and selectivity. Notably, a derivative of fostriecin lacking the entire lactone subunit demonstrated marked potency and selectivity for PP2A, while having substantially reduced and similar activity against PP1 and PP1/PP2A- PP5/PP2A-chimeras that have greatly increased sensitivity to both fostriecin and cytostatin. This suggests that other features [e.g., the (Z,Z,E)-triene] also contribute to inhibitory selectivity. When considered together with previous data, these studies suggest that, despite the high structural conservation of the catalytic site in PP1, PP2A and PP5, the development of highly selective catalytic inhibitors should be feasible.


Asunto(s)
Alquenos/farmacología , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Proteínas Mutantes Quiméricas/antagonistas & inhibidores , Proteínas Nucleares/antagonistas & inhibidores , Organofosfatos/farmacología , Fosfoproteínas Fosfatasas/antagonistas & inhibidores , Proteína Fosfatasa 1/antagonistas & inhibidores , Proteína Fosfatasa 2/antagonistas & inhibidores , Pironas/farmacología , Alquenos/química , Alquenos/metabolismo , Secuencia de Aminoácidos , Animales , Dominio Catalítico/efectos de los fármacos , Dominio Catalítico/genética , Bovinos , Inhibidores Enzimáticos/metabolismo , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Proteínas Mutantes Quiméricas/genética , Proteínas Mutantes Quiméricas/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Organofosfatos/química , Fosfoproteínas Fosfatasas/genética , Fosfoproteínas Fosfatasas/metabolismo , Polienos , Unión Proteica/efectos de los fármacos , Proteína Fosfatasa 1/genética , Proteína Fosfatasa 1/metabolismo , Proteína Fosfatasa 2/genética , Proteína Fosfatasa 2/metabolismo , Estructura Terciaria de Proteína/efectos de los fármacos , Pironas/química , Pironas/metabolismo , Conejos , Relación Estructura-Actividad
12.
Biochim Biophys Acta ; 1782(4): 259-70, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18280813

RESUMEN

Ser/Thr protein phosphatase 5 (PP5) regulates several signaling-cascades that suppress growth and/or facilitate apoptosis in response to genomic stress. The expression of PP5 is responsive to hypoxia inducible factor-1 (HIF-1) and estrogen, which have both been linked to the progression of human breast cancer. Still, it is not clear if PP5 plays a role in the development of human cancer. Here, immunostaining of breast cancer tissue-microarrays (TMAs) revealed a positive correlation between PP5 over-expression and ductal carcinoma in situ (DCIS; P value 0.0028), invasive ductal carcinoma (IDC; P value 0.012) and IDC with metastases at the time of diagnosis (P value 0.0001). In a mouse xenograft model, the constitutive over-expression of PP5 was associated with an increase in the rate of tumor growth. In a MCF-7 cell culture model over-expression correlated with both an increase in the rate of proliferation and protection from cell death induced by oxidative stress, UVC-irradiation, adriamycin, and vinblastine. PP5 over-expression had no apparent effect on the sensitivity of MCF-7 cells to taxol or rapamycin. Western analysis of extracts from cells over-expressing PP5 revealed a decrease in the phosphorylation of known substrates for PP5. Together, these studies indicate that elevated levels of PP5 protein occur in human breast cancer and suggest that PP5 over-expression may aid tumor progression.


Asunto(s)
Neoplasias de la Mama/enzimología , Carcinoma Ductal de Mama/enzimología , Carcinoma Intraductal no Infiltrante/enzimología , Animales , Muerte Celular , Línea Celular Tumoral , Doxorrubicina/farmacología , Humanos , Ratones , Ratones Desnudos , Proteínas Nucleares , Estrés Oxidativo , Fosfoproteínas Fosfatasas , Treonina , Factores de Tiempo , Rayos Ultravioleta , Regulación hacia Arriba , Vinblastina/farmacología
13.
J Am Chem Soc ; 128(51): 16720-32, 2006 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-17177422

RESUMEN

The total synthesis of cytostatin, an antitumor agent belonging to the fostriecin family of natural products, is described in full detail. The convergent approach relied on a key epoxide-opening reaction to join the two stereotriad units and a single-step late-stage stereoselective installation of the sensitive (Z,Z,E)-triene through a beta-chelation-controlled nucleophilic addition. The synthetic route provided rapid access to the C4-C6 stereoisomers of the cytostatin lactone, which were prepared and used to define the C4-C6 relative stereochemistry of the natural product. In addition to the natural product, each of the C10-C11 diastereomers of cytostatin was divergently prepared (11 steps from key convergence step) by this route and used to unequivocally confirm the relative and absolute stereochemistry of cytostatin. Each of the cytostatin diastereomers exhibited a reduced activity toward inhibition of PP2A (>100-fold), demonstrating the importance of the presence and stereochemistry of the C10-methyl and C11-hydroxy groups for potent PP2A inhibition. Extensions of the studies provided dephosphocytostatin, sulfocytostatin (a key analogue related to the natural product sultriecin), 11-deshydroxycytostatin, and an analogue lacking the entire C12-C18 (Z,Z,E)-triene segment, which were used to define the magnitude of the C9-phosphate (>4000-fold), C11-alcohol (250-fold), and triene (220-fold) contribution to PP2A inhibition. A model of cytostatin bound to the active site of PP2A is presented, compared to that of fostriecin, which is also presented in detail for the first time, and used to provide insights into the role of the key substituents. Notably, the alpha,beta unsaturated lactone of cytostatin, like that of fostriecin, is projected to serve as a key electrophile, providing a covalent adduct with Cys269 unique to PP2A, contributing to its potency (> or =200-fold for fostriecin) and accounting for its selectivity.


Asunto(s)
Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacología , Organofosfatos/síntesis química , Organofosfatos/farmacología , Fosfoproteínas Fosfatasas/antagonistas & inhibidores , Pironas/síntesis química , Pironas/farmacología , Alquenos/farmacología , Animales , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Inhibidores Enzimáticos/química , Humanos , Leucemia L1210 , Ratones , Modelos Moleculares , Conformación Molecular , Organofosfatos/química , Polienos , Pironas/química , Estereoisomerismo , Relación Estructura-Actividad
14.
Biochem J ; 376(Pt 1): 87-95, 2003 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-12932195

RESUMEN

The sizes and anomers of the products formed during the hydrolysis of chitin oligosaccharides by the Family 18 chitinase A (ChiA) from Serratia marcescens were analysed by hydrophilic interaction chromatography using a novel approach in which reactions were performed at 0 degrees C to stabilize the anomer conformations of the initial products. Crystallographic studies of the enzyme, having the structure of the complex of the ChiA E315L (Glu315-->Leu) mutant with a hexasaccharide, show that the oligosaccharide occupies subsites -4 to +2 in the substrate-binding cleft, consistent with the processing of beta-chitin by the release of disaccharide at the reducing end. Products of the hydrolysis of hexa- and penta-saccharides by wild-type ChiA, as well as by two mutants of the residues Trp275 and Phe396 important in binding the substrate at the +1 and +2 sites, show that the substrates only occupy sites -2 to +2 and that additional N -acetyl-D-glucosamines extend beyond the substrate-binding cleft at the reducing end. The subsites -3 and -4 are not used in this four-site binding mode. The explanation for these results is found in the high importance of individual binding sites for the processing of short oligosaccharides compared with the cumulative recognition and processive hydrolysis mechanism used to digest natural beta-chitin.


Asunto(s)
Quitina/metabolismo , Quitinasas/metabolismo , Serratia marcescens/enzimología , Sitios de Unión , Quitina/química , Quitinasas/química , Quitinasas/genética , Cristalografía por Rayos X , Hidrólisis , Isomerismo , Espectrometría de Masas , Modelos Moleculares , Mutación , Oligosacáridos/metabolismo , Especificidad por Sustrato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...