Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Hypertens Pregnancy ; 42(1): 2285757, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38017693

RESUMEN

OBJECTIVE: The aim of this study was to investigate the possible causal relationship between COVID-19 and the risk of pre-eclampsia/eclampsia using a Mendelian randomized (MR) design. METHODS: We estimated their genetic correlations and then performed two-sample Mendelian randomization analyses using pooled statistics from the COVID-19 susceptibility/hospitalization genome-wide association study and the pre-eclampsia/eclampsia datasets. The main analyses were performed using the inverse variance weighting method, supplemented by the weighted median method and the MR-Egger method. RESULTS: We identified a significant and positive genetic correlation between COVID-19 susceptibility and pre-eclampsia/eclampsia [OR = 1.23 (1.01-1.51), p = 0.043]. Meanwhile, hospitalization of COVID-19 was significantly associated with a higher risk of pre-eclampsia/eclampsia [OR = 1.15 (1.02-1.30), p = 0.024]. Consistently, hospitalization of COVID-19 were nominally associated with higher risk of pre-eclampsia [OR = 1.14, (1.01-1.30), p = 0.040]. The results were robust under all sensitivity analyses. CONCLUSION: These results suggest that COVID-19 may increase the risk of pre-eclampsia/eclampsia. Future development of preventive or therapeutic interventions should emphasize this to mitigate the complications of COVID-19. [Figure: see text].


Asunto(s)
COVID-19 , Eclampsia , Preeclampsia , Femenino , Embarazo , Humanos , Estudio de Asociación del Genoma Completo , Análisis de la Aleatorización Mendeliana , Preeclampsia/genética
2.
Target Oncol ; 18(2): 269-285, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36826464

RESUMEN

BACKGROUND: Patients with triple-negative breast cancer (TNBC) expressing the androgen receptor (AR) respond poorly to neoadjuvant chemotherapy, although AR antagonists have shown promising clinical activity, suggesting these tumors are AR-dependent. cAMP responsive element binding protein (CREB)-binding protein (CBP) and p300 are transcriptional co-activators for the AR, a key driver of AR+ breast and prostate cancer, and may provide a novel therapeutic target in AR+ TNBC. OBJECTIVES: The aim of this study was to determine the therapeutic potential of FT-6876, a new CBP/p300 bromodomain inhibitor, in breast cancer models with a range of AR levels in vitro and in vivo. METHODS: Effects of FT-6876 on the CBP/p300 pathway were determined by combining chromatin immunoprecipitation (ChIP) with precision run-on sequencing (PRO-seq) complemented with H3K27 acetylation (Ac) and transcriptional profiling. The antiproliferative effect of FT-6876 was also measured in vitro and in vivo. RESULTS: We describe the discovery of FT-6876, a potent and selective CBP/p300 bromodomain inhibitor. The combination of ChIP and PRO-seq confirmed the reduction in H3K27Ac at specific promoter sites concurrent with a decrease in CBP/p300 on the chromatin and a reduction in nascent RNA and enhancer RNA. This was associated with a time- and concentration-dependent reduction in H3K37Ac associated with a decrease in AR and estrogen receptor (ER) target gene expression. This led to a time-dependent growth inhibition in AR+ models, correlated with AR expression. Tumor growth inhibition was also observed in AR+ tumor models of TNBC and ER+ breast cancer subtypes with consistent pharmacokinetics and pharmacodynamics. CONCLUSION: Our findings demonstrate FT-6876 as a promising new CBP/p300 bromodomain inhibitor, with efficacy in preclinical models of AR+ breast cancer.


Asunto(s)
Receptores Androgénicos , Neoplasias de la Mama Triple Negativas , Masculino , Humanos , Receptores Androgénicos/metabolismo , Proteína de Unión a CREB/genética , Proteína de Unión a CREB/metabolismo , Unión Proteica , ARN/metabolismo
3.
Nurs Open ; 8(5): 2686-2695, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33760375

RESUMEN

AIM: To analyse the application status of nursing assessment terminology for neurological conditions and determine whether the International Classification of Functioning, Disability, and Health (ICF) covers nursing assessment. DESIGN: A multi-centre cross-sectional study. METHODS: Four researchers extracted all nursing problems from the patients of three different hospitals and formed a pool of nursing terminology from the electronic nursing records, self-reports, family reports, medical examinations, and clinical records for all patients. The ICF Linking Rules were then used to map the nursing assessment terminology of neurological conditions with the ICF. RESULTS: Though 37.5% of nursing assessment terms were closely related to neurological diseases, this does not appear in the existing electronic nursing assessment records. The unrecorded rate of 9 (16.1%) terms ranged from 40%-50%, while the unrecorded rate of 8 (14.3%) terms was more than 80%. Overall, 96.4% of nursing assessment terms could be described by the corresponding categories of the ICF, with 37 (66.1%) of the "same" concepts, 9 (16.1%) "similar" concepts, 6 (10.7%) "narrower" concepts (the nursing assessment terms were more specific than the ICF categories), and 2 (3.6%) "broader" concepts (the nursing assessment were less specific than the ICF categories).


Asunto(s)
Clasificación Internacional del Funcionamiento, de la Discapacidad y de la Salud , Terminología Normalizada de Enfermería , Estudios Transversales , Evaluación de la Discapacidad , Humanos , Evaluación en Enfermería
4.
J Diabetes Investig ; 9(6): 1360-1369, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29575786

RESUMEN

AIMS/INTRODUCTION: To explore the association between lactation and type 2 diabetes incidence in women with prior gestational diabetes. MATERIALS AND METHODS: We searched PubMed, Embase and the Cochrane Library for cohort studies published through 12 June 2017 that evaluated the effect of lactation on the development of type 2 diabetes in women with prior gestational diabetes. A random effects model was used to estimate relative risks (RRs) with 95% confidence intervals (CIs). RESULTS: A total of 13 cohort studies were included in the meta-analysis. The pooled result suggested that compared with no lactation, lactation was significantly associated with a lower risk of type 2 diabetes (RR 0.66, 95% CI 0.48-0.90, I2  = 72.8%, P < 0.001). This relationship was prominent in a study carried out in the USA (RR 0.66, 95% CI 0.43-0.99), regardless of study design (prospective design RR 0.56, 95% CI 0.41-0.76; retrospective design RR 0.63, 95% CI 0.40-0.99), smaller sample size (RR 0.52, 95% CI 0.30-0.92, P = 0.024) and follow-up duration >1 years (RR 0.75, 95% CI 0.56-1.00), and the study used adjusted data (RR 0.69, 95% CI 0.50-0.94). Finally, by pooling data from three studies, we failed to show that compared with no lactation, long-term lactation (>1 to 3 months postpartum) was associated with the type 2 diabetes risk (RR 0.69, 95% CI 0.41-1.17). CONCLUSIONS: The present meta-analysis showed that lactation was associated with a lower risk of type 2 diabetes in women with prior gestational diabetes. Furthermore, no significant relationship between long-term lactation and type 2 diabetes risk was detected. The impact of long-term lactation and the risk of type 2 diabetes should be verified in further large-scale studies.


Asunto(s)
Diabetes Mellitus Tipo 2/epidemiología , Diabetes Gestacional/epidemiología , Progresión de la Enfermedad , Lactancia , Estudios de Cohortes , Diabetes Mellitus Tipo 2/fisiopatología , Diabetes Gestacional/fisiopatología , Femenino , Humanos , Embarazo , Factores de Riesgo
5.
Oncotarget ; 8(31): 51733-51747, 2017 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-28881682

RESUMEN

Because cancer stem cells (CSCs) have been implicated in chemo-resistance, metastasis and tumor recurrence, therapeutic targeting of CSCs holds promise to address these clinical challenges to cancer treatment. VS-4718 and VS-6063 are potent inhibitors of focal adhesion kinase (FAK), a non-receptor tyrosine kinase that mediates cell signals transmitted by integrins and growth factor receptors. We report here that inhibition of FAK kinase activity by VS-4718 or VS-6063 preferentially targets CSCs, as demonstrated by a panel of orthogonal CSC assays in cell line models and surgically resected primary breast tumor specimens cultured ex vivo. Oral administration of VS-4718 or VS-6063 to mice bearing xenograft models of triple-negative breast cancer (TNBC) significantly reduced the proportion of CSCs in the tumors, as evidenced by a reduced tumor-initiating capability upon re-implantation in limiting dilutions of cells prepared from these tumors. In contrast, the cytotoxic chemotherapeutic agents, paclitaxel and carboplatin, enriched for CSCs, consistent with previous reports that these cytotoxic agents preferentially target non-CSCs. Importantly, VS-4718 and VS-6063 attenuated the chemotherapy-induced enrichment of CSCs in vitro and delayed tumor regrowth following cessation of chemotherapy. An intriguing crosstalk between FAK and the Wnt/ß-catenin pathway was revealed wherein FAK inhibition blocks ß-catenin activation by reducing tyrosine 654 phosphorylation of ß-catenin. Furthermore, a constitutively active mutant form of ß-catenin reversed the preferential targeting of CSCs by FAK inhibition, suggesting that this targeting is mediated, at least in part, through attenuating ß-catenin activation. The preferential targeting of cancer stem cells by FAK inhibitors provides a rationale for the clinical development of FAK inhibitors aimed to increase durable responses for cancer patients.

6.
Int J Cancer ; 139(9): 2116-26, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27376928

RESUMEN

Pancreatic cancer, the fourth leading cause of cancer death in the United States, has a negative prognosis because metastasis occurs before symptoms manifest. Leiodermatolide, a polyketide macrolide with antimitotic activity isolated from a deep water sponge of the genus Leiodermatium, exhibits potent and selective cytotoxicity toward the pancreatic cancer cell lines AsPC-1, PANC-1, BxPC-3, and MIA PaCa-2, and potent cytotoxicity against skin, breast and colon cancer cell lines. Induction of apoptosis by leiodermatolide was confirmed in the AsPC-1, BxPC-3 and MIA PaCa-2 cells. Leiodermatolide induces cell cycle arrest but has no effects on in vitro polymerization or depolymerization of tubulin alone, while it enhances polymerization of tubulin containing microtubule associated proteins (MAPs). Observations through confocal microscopy show that leiodermatolide, at low concentrations, causes minimal effects on polymerization or depolymerization of the microtubule network in interphase cells, but disruption of spindle formation in mitotic cells. At higher concentrations, depolymerization of the microtubule network is observed. Visualization of the growing microtubule in HeLa cells expressing GFP-tagged plus end binding protein EB-1 showed that leiodermatolide stopped the polymerization of tubulin. These results suggest that leiodermatolide may affect tubulin dynamics without directly interacting with tubulin and hint at a unique mechanism of action. In a mouse model of metastatic pancreatic cancer, leiodermatolide exhibited significant tumor reduction when compared to gemcitabine and controls. The antitumor activities of leiodermatolide, as well as the proven utility of antimitotic compounds against cancer, make leiodermatolide an interesting compound with potential chemotherapeutic effects that may merit further research.


Asunto(s)
Antineoplásicos/administración & dosificación , Macrólidos/administración & dosificación , Microtúbulos/efectos de los fármacos , Neoplasias Pancreáticas/tratamiento farmacológico , Moduladores de Tubulina/administración & dosificación , Animales , Antineoplásicos/farmacología , Apoptosis , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HeLa , Humanos , Macrólidos/farmacología , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Metástasis de la Neoplasia , Neoplasias Pancreáticas/metabolismo , Moduladores de Tubulina/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Cancer Biol Ther ; 16(4): 589-601, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25729885

RESUMEN

Natural compound schweinfurthins are of considerable interest for novel therapy development because of their selective anti-proliferative activity against human cancer cells. We previously reported the isolation of highly active schweinfurthins E-H, and in the present study, mechanisms of the potent and selective anti-proliferation were investigated. We found that schweinfurthins preferentially inhibited the proliferation of PTEN deficient cancer cells by indirect inhibition of AKT phosphorylation. Mechanistically, schweinfurthins and their analogs arrested trans-Golgi-network trafficking, an intracellular vesicular trafficking system, resulting in the induction of endoplasmic reticulum stress and the suppression of both lipid raft-mediated PI3K activation and mTOR/RheB complex formation, which collectively led to an effective inhibition of mTOR/AKT signaling. The trans-Golgi-network traffic arresting effect of schweinfurthins was associated with their in vitro binding activity to oxysterol-binding proteins that are known to regulate intracellular vesicular trafficking. Moreover, schweinfurthins were found to be highly toxic toward PTEN-deficient B cell lymphoma cells, and displayed 2 orders of magnitude lower activity toward normal human peripheral blood mononuclear cells and primary fibroblasts in vitro. These results revealed a previously unrecognized role of schweinfurthins in regulating trans-Golgi-network trafficking, and linked mechanistically this cellular effect with mTOR/AKT signaling and with cancer cell survival and growth. Our findings suggest the schweinfurthin class of compounds as a novel approach to modulate oncogenic mTOR/AKT signaling for cancer treatment.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Estilbenos/farmacología , Serina-Treonina Quinasas TOR/metabolismo , Red trans-Golgi/efectos de los fármacos , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Linfoma de Células B/metabolismo , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo
8.
Cancer Res ; 75(2): 446-55, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25432176

RESUMEN

Cancer stem cells (CSC) have been implicated in disease recurrence, metastasis, and therapeutic resistance, but effective targeting strategies for these cells are still wanting. VS-5584 is a potent and selective dual inhibitor of mTORC1/2 and class I PI 3-kinases. Here, we report that VS-5584 is up to 30-fold more potent in inhibiting the proliferation and survival of CSC compared with non-CSC in solid tumor cell populations. VS-5584 preferentially diminished CSC levels in multiple mouse xenograft models of human cancer, as evidenced by marked reduction of tumor-initiating capacity in limiting dilution assays. Likewise, VS-5584 treatment ex vivo preferentially reduced CSC in surgically resected breast and ovarian patient tumors. In contrast, chemotherapeutics such as paclitaxel and cisplatin were less effective in targeting CSC than bulk tumor cells. Mechanistic investigations revealed that preferential targeting of CSC required inhibition of multiple components of the PI3K-mTOR pathway: coordinate RNAi-mediated silencing of PI3Kα, PI3Kß, and mTOR phenocopied the effect of VS-5584, exhibiting the strongest preferential targeting of CSC, while silencing of individual PI3K isoforms or mTOR failed to replicate the effect of VS-5584. Consistent with CSC ablation, VS-5584 delayed tumor regrowth following chemotherapy in xenograft models of small-cell lung cancer. Taken together, the preferential targeting of CSC prompts a new paradigm for clinical testing of VS-5584: clinical trials designed with CSC-directed endpoints may facilitate demonstration of the therapeutic benefit of VS-5584. We suggest that combining VS-5584 with classic chemotherapy that debulks tumors may engender a more effective strategy to achieve durable remissions in patients with cancer.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Morfolinas/farmacología , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/enzimología , Neoplasias Ováricas/tratamiento farmacológico , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/farmacología , Purinas/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Animales , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Femenino , Humanos , Células MCF-7 , Ratones , Ratones Endogámicos NOD , Ratones SCID , Neoplasias Ováricas/enzimología , Neoplasias Ováricas/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Distribución Aleatoria , Serina-Treonina Quinasas TOR/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Blood ; 124(17): 2675-86, 2014 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-25217697

RESUMEN

Proline-rich tyrosine kinase 2 (Pyk2) is a member of the focal adhesion kinase family that has been recently linked to tumor development. However, its role in modulating multiple myeloma (MM) biology and disease progression remains unexplored. We first demonstrated that patients with MM present with higher expression of Pyk2 compared with healthy individuals. By using loss-of-function approaches, we found that Pyk2 inhibition led to reduction of MM tumor growth in vivo as well as decreased cell proliferation, cell-cycle progression, and adhesion ability in vitro. In turn, overexpression of Pyk2 promoted the malignant phenotype, substantiated by enhanced tumor growth and reduced survival. Mechanistically, inhibition of Pyk2 reduced activation of Wnt/ß-catenin signaling by destabilizing ß-catenin, leading to downregulation of c-Myc and Cyclin D1. Furthermore, treatment of MM cells with the FAK/Pyk2 inhibitor VS-4718 effectively inhibited MM cell growth both in vitro and in vivo. Collectively, our findings describe the tumor-promoting role of Pyk2 in MM, thus providing molecular evidence for a novel tyrosine kinase inhibitor as a new therapeutic option in MM.


Asunto(s)
Aminopiridinas/farmacología , Quinasa 2 de Adhesión Focal/antagonistas & inhibidores , Mieloma Múltiple/prevención & control , Inhibidores de Proteínas Quinasas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Adhesión Celular/efectos de los fármacos , Adhesión Celular/genética , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Progresión de la Enfermedad , Femenino , Quinasa 2 de Adhesión Focal/genética , Quinasa 2 de Adhesión Focal/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Immunoblotting , Mediciones Luminiscentes , Ratones SCID , Mieloma Múltiple/genética , Mieloma Múltiple/metabolismo , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Supervivencia , Carga Tumoral/efectos de los fármacos , Carga Tumoral/genética , Vía de Señalización Wnt/efectos de los fármacos , beta Catenina/metabolismo
10.
Sci Transl Med ; 6(237): 237ra68, 2014 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-24848258

RESUMEN

The goal of targeted therapy is to match a selective drug with a genetic lesion that predicts for drug sensitivity. In a diverse panel of cancer cell lines, we found that the cells most sensitive to focal adhesion kinase (FAK) inhibition lack expression of the neurofibromatosis type 2 (NF2) tumor suppressor gene product, Merlin. Merlin expression is often lost in malignant pleural mesothelioma (MPM), an asbestos-induced aggressive cancer with limited treatment options. Our data demonstrate that low Merlin expression predicts for increased sensitivity of MPM cells to a FAK inhibitor, VS-4718, in vitro and in tumor xenograft models. Disruption of MPM cell-cell or cell-extracellular matrix (ECM) contacts with blocking antibodies suggests that weak cell-cell adhesions in Merlin-negative MPM cells underlie their greater dependence on cell-ECM-induced FAK signaling. This provides one explanation of why Merlin-negative cells are vulnerable to FAK inhibitor treatment. Furthermore, we validated aldehyde dehydrogenase as a marker of cancer stem cells (CSCs) in MPM, a cell population thought to mediate tumor relapse after chemotherapy. Whereas pemetrexed and cisplatin, standard-of-care agents for MPM, enrich for CSCs, FAK inhibitor treatment preferentially eliminates these cells. These preclinical results provide the rationale for a clinical trial in MPM patients using a FAK inhibitor as a single agent after first-line chemotherapy. With this design, the FAK inhibitor could potentially induce a more durable clinical response through reduction of CSCs along with a strong antitumor effect. Furthermore, our data suggest that patients with Merlin-negative tumors may especially benefit from FAK inhibitor treatment.


Asunto(s)
Antineoplásicos/farmacología , Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Neoplasias Pulmonares/tratamiento farmacológico , Mesotelioma/tratamiento farmacológico , Neurofibromina 2/deficiencia , Inhibidores de Proteínas Quinasas/farmacología , Aldehído Deshidrogenasa/metabolismo , Animales , Apoptosis/efectos de los fármacos , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Quinasa 1 de Adhesión Focal/genética , Quinasa 1 de Adhesión Focal/metabolismo , Humanos , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Mesotelioma/enzimología , Mesotelioma/genética , Mesotelioma/patología , Mesotelioma Maligno , Ratones , Terapia Molecular Dirigida , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/enzimología , Neurofibromina 2/genética , Interferencia de ARN , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Transfección , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Chem Commun (Camb) ; 48(96): 11766-8, 2012 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-23114612

RESUMEN

Rlyso, a highly selective and sensitive pH sensor, can stain lysosomes with a novel lysosome-locating group, methylcarbitol. Rlyso was successfully used to detect lysosomal pH changes during apoptosis or induced by chloroquine while avoiding the "alkalizing effect" on lysosomes of current lysosomal probes with nitrogen-containing sidechains.


Asunto(s)
Glicoles de Etileno/química , Colorantes Fluorescentes/química , Lisosomas/química , Animales , Antineoplásicos Hormonales/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Cloroquina/farmacología , Dexametasona/farmacología , Humanos , Concentración de Iones de Hidrógeno/efectos de los fármacos , Ratones , Microscopía Fluorescente , Espectrometría de Fluorescencia
12.
Anticancer Res ; 31(9): 2773-9, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21868519

RESUMEN

The marine natural product (+)-spongistatin 1 is an extremely potent growth inhibitory agent having activity against a wide variety of cancer cell lines, while exhibiting low cytotoxicity against quiescent human fibroblasts. Consistent with a microtubule-targeting mechanism of action, (+)-spongistatin 1 causes mitotic arrest in DU145 human prostate cancer cells. More importantly, (+)-spongistatin 1 exhibits significant in vivo antitumor activity in the LOX-IMVI human melanoma xenograft model. (+)-Spongistatin 1 is, thus, an important class of microtubule targeting anticancer agent that warrants further investigation.


Asunto(s)
Macrólidos/farmacología , Moduladores de Tubulina/farmacología , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Animales , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Técnicas In Vitro , Macrólidos/química , Macrólidos/metabolismo , Ratones , Microscopía Fluorescente , Estereoisomerismo , Moduladores de Tubulina/química , Moduladores de Tubulina/metabolismo
13.
J Am Chem Soc ; 133(35): 14042-53, 2011 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-21761891

RESUMEN

The design, synthesis, and biological evaluation of two diminutive forms of (+)-spongistatin 1, in conjunction with the development of a potentially general design strategy to simplify highly flexible macrocyclic molecules while maintaining biological activity, have been achieved. Examination of the solution conformations of (+)-spongistatin 1 revealed a common conformational preference along the western perimeter comprising the ABEF rings. Exploiting the hypothesis that the small-molecule recognition/binding domains are likely to comprise the conformationally less mobile portions of a ligand led to the design of analogues, incorporating tethers (blue) in place of the CD and the ABCD components of the (+)-spongistatin 1 macrolide, such that the conformation of the retained (+)-spongistatin 1 skeleton would mimic the assigned solution conformations of the natural product. The observed nanomolar cytotoxicity and microtubule destabilizing activity of the ABEF analogue provide support for both the assigned solution conformation of (+)-spongistatin 1 and the validity of the design strategy.


Asunto(s)
Macrólidos/química , Macrólidos/farmacología , Moduladores de Tubulina/química , Moduladores de Tubulina/farmacología , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Diseño de Fármacos , Humanos , Macrólidos/síntesis química , Ratones , Modelos Moleculares , Neoplasias/tratamiento farmacológico , Moduladores de Tubulina/síntesis química
14.
Org Lett ; 12(8): 1792-5, 2010 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-20297810

RESUMEN

The design, synthesis, and biological evaluation of two potential (+)-spongistatin 1 analogues have been achieved. The analogues, incorporating tethers (red) in place of the ABCD and the CD components of the (+)-spongistatin 1 macrolide, were designed such that the conformations of the retained skeleton (blue) would mimic the assigned major solution conformation of the natural product The nanomolar cytotoxicity observed for the ABEF analogue provides strong support for the assigned solution conformation.


Asunto(s)
Diseño de Fármacos , Macrólidos/química , Macrólidos/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Concentración 50 Inhibidora , Macrólidos/síntesis química , Relación Estructura-Actividad
15.
Mol Cancer Ther ; 8(5): 1250-60, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19417157

RESUMEN

We report here that des-methyl, des-amino pateamine A (DMDA-PatA), a structurally simplified analogue of the marine natural product pateamine A, has potent antiproliferative activity against a wide variety of human cancer cell lines while showing relatively low cytotoxicity against nonproliferating, quiescent human fibroblasts. DMDA-PatA retains almost full in vitro potency in P-glycoprotein-overexpressing MES-SA/Dx5-Rx1 human uterine sarcoma cells that are significantly resistant to paclitaxel, suggesting that DMDA-PatA is not a substrate for P-glycoprotein-mediated drug efflux. Treatment of proliferating cells with DMDA-PatA leads to rapid shutdown of DNA synthesis in the S phase of the cell cycle. Cell-free studies show that DMDA-PatA directly inhibits DNA polymerases α and γ in vitro albeit at concentrations considerably higher than those that inhibit cell proliferation. DMDA-PatA shows potent anticancer activity in several human cancer xenograft models in nude mice, including significant regressions observed in the LOX and MDA-MB-435 melanoma models. DMDA-PatA thus represents a promising natural product-based anticancer agent that warrants further investigation.


Asunto(s)
Antineoplásicos/farmacología , Compuestos Epoxi/farmacología , Macrólidos/farmacología , Tiazoles/farmacología , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Animales , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Compuestos Epoxi/química , Compuestos Epoxi/uso terapéutico , Femenino , Humanos , Macrólidos/química , Macrólidos/uso terapéutico , Ratones , Ratones Desnudos , Ratones SCID , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Tiazoles/química , Tiazoles/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Org Lett ; 11(6): 1417-20, 2009 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-19228042

RESUMEN

The first total synthesis of ipomoeassin F was carried out using a convergent approach that relied upon the use of Schmidt glycosidation technology for the coupling of two suitably protected monosaccharide fragments. After two steps, ring-closing metathesis was used to form the macrocyclic ring, and seven more steps then furnished ipomoeassin F. In vitro inhibitory activity against a four-panel cell line showed low nanomolar inhibitory activity.


Asunto(s)
Antineoplásicos Fitogénicos/síntesis química , Productos Biológicos/síntesis química , Glicoconjugados/síntesis química , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/farmacología , Productos Biológicos/química , Productos Biológicos/farmacología , Ensayos de Selección de Medicamentos Antitumorales , Glicoconjugados/química , Glicoconjugados/farmacología , Glicosilación , Humanos , Ipomoea/química , Estructura Molecular
17.
J Biol Chem ; 279(51): 53015-22, 2004 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-15471885

RESUMEN

The p53 tumor suppressor is a key mediator of the cellular response to stress. Phosphorylation induced by multiple stress-activated kinases has been proposed to be essential for p53 stabilization, interaction with transcriptional co-activators, and activation of p53 target genes. However, genetic studies suggest that stress-activated phosphorylation may not be essential for p53 activation. We therefore investigated the role of p53 phosphorylation on six key serine residues (Ser(6), Ser(15), Ser(20), Ser(37), Ser(46), and Ser(392)) for p53 activation using nutlin-3, a recently developed small molecule MDM2 antagonist. We show here that nutlin does not induce the phosphorylation of p53. Comparison of the activity of unphosphorylated and phosphorylated p53 induced by the genotoxic drugs doxorubicin and etoposide in HCT116 and RKO cells revealed no difference in their sequence-specific DNA binding and ability to transactivate p53 target genes and to induce p53-dependent apoptosis. We conclude that p53 phosphorylation on six major serine sites is not required for activation of p53 target genes or biological responses in vivo.


Asunto(s)
Apoptosis , Activación Transcripcional , Proteína p53 Supresora de Tumor/metabolismo , Anexina A5/farmacología , Antibióticos Antineoplásicos/farmacología , Antineoplásicos Fitogénicos/farmacología , Western Blotting , Línea Celular Tumoral , Colorantes/farmacología , ADN/química , Relación Dosis-Respuesta a Droga , Doxorrubicina/farmacología , Ensayo de Inmunoadsorción Enzimática , Etopósido/farmacología , Humanos , Imidazoles/metabolismo , Cinética , Proteínas Nucleares/metabolismo , Fosforilación , Piperazinas/metabolismo , Reacción en Cadena de la Polimerasa , Unión Proteica , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-mdm2 , Serina/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA