Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Biochem Biophys ; 78(2): 191-202, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32449075

RESUMEN

Pancreatic adenocarcinoma is an aggressive cancer with poor clinical prognosis and limited therapeutic options. There is a significant lack of effective, safe, and targeted therapies for successful treatment of pancreatic cancer. In this report, we describe the anticancer efficacy of two novel compounds, N-methylpiperazinyl diarylidenylpiperidone (L-2663) and its pro-nitroxide conjugate (HO-4589) evaluated on human pancreatic adenocarcinoma (AsPC-1) cell line and xenograft tumor in mice. Using flow cytometry, we determined the effect of the L-2663 and HO-4589 drugs in inducing mitochondrial toxicity, triggering cell-cycle arrest, and apoptosis. EPR spectroscopy was used to quantify cellular uptake, metabolic conversion and stability of HO-4589 in cells and in vivo monitoring of tumor oxygenation as a function of growth. The results established different antiproliferative efficacy of the L-2663 and HO-4589 compounds, with a targeted action on cancer cells while being less toxic to noncancerous cells. The study may have important implications in the future designs of safe and effective chemotherapeutic agents for the treatment of pancreatic cancer.


Asunto(s)
Antineoplásicos/farmacología , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Neoplasias Pancreáticas/tratamiento farmacológico , Piperazinas/farmacología , Piperidonas/farmacología , Animales , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , División Celular , Línea Celular Tumoral , Supervivencia Celular , Ensayos de Selección de Medicamentos Antitumorales , Espectroscopía de Resonancia por Spin del Electrón , Fase G2 , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Trasplante de Neoplasias , Oximetría , Especies Reactivas de Oxígeno/metabolismo
3.
Cell Biochem Biophys ; 77(1): 61-67, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30552554

RESUMEN

The synthesis and antiproliferative effect of a novel curcumin analog, 4,4'-disulfonyldiarylidenyl piperidone, are reported. The design of the molecule is based on the fusion of an antiproliferative segment, namely diarylidenyl piperidone (DAP), with N-hyroxypyrroline, which is known to metabolically convert to nitroxide and protect healthy cells. Cellular uptake, metabolic conversion, cytotoxicity and antiproliferative effect of the DAP derivative against HCT-116 human colon cancer cells have been determined. Based on cell viability and proliferation assays as well as western-blot analysis of major transcription factors and inhibitory proteins, it is determined that the DAP compound is cytotoxic by inhibiting cell survival and proliferation pathways. The findings may have important implications in the design and development of effective anticancer agents.


Asunto(s)
Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Curcumina/farmacología , Antineoplásicos/química , Antineoplásicos/metabolismo , Apoptosis/efectos de los fármacos , Biomarcadores de Tumor/metabolismo , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Curcumina/análogos & derivados , Curcumina/metabolismo , Espectroscopía de Resonancia por Spin del Electrón , Células HCT116 , Humanos , Fosforilación/efectos de los fármacos , Piperidonas/química , Piperidonas/metabolismo , Piperidonas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Factor de Transcripción STAT3/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
4.
Front Cardiovasc Med ; 5: 114, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30211171

RESUMEN

Myocardial infarction (MI), which occurs often due to acute ischemia followed by reflow, is associated with irreversible loss (death) of cardiomyocytes. If left untreated, MI will lead to progressive loss of viable cardiomyocytes, deterioration of cardiac function, and congestive heart failure. While supplemental oxygen therapy has long been in practice to treat acute MI, there has not been a clear scientific basis for the observed beneficial effects. Further, there is no rationale for the amount or duration of administration of supplemental oxygenation for effective therapy. The goal of the present study was to determine an optimum oxygenation protocol that can be clinically applicable for treating acute MI. Using EPR oximetry, we studied the effect of exposure to supplemental oxygen cycling (OxCy) administered by inhalation of 21-100% oxygen for brief periods (15-90 min), daily for 5 days, using a rat model of acute MI. Myocardial oxygen tension (pO2), cardiac function and pro-survival/apoptotic signaling molecules were used as markers of treatment outcome. OxCy resulted in a significant reduction of infarct size and improvement of cardiac function. An optimal condition of 30-min OxCy with 95% oxygen + 5% CO2 under normobaric conditions was found to be effective for cardioprotection.

5.
Biomed Microdevices ; 20(2): 29, 2018 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-29549438

RESUMEN

Tissue oxygenation is a critical parameter in various pathophysiological situations including cardiovascular disease and cancer. Hypoxia can significantly influence the prognosis of solid malignancies and the efficacy of their treatment by radiation or chemotherapy. Electron paramagnetic resonance (EPR) oximetry is a reliable method for repeatedly assessing and monitoring oxygen levels in tissues. Lithium octa-n-butoxynaphthalocyanine (LiNc-BuO) has been developed as a probe for biological EPR oximetry, especially for clinical use. However, clinical applicability of LiNc-BuO crystals is hampered by potential limitations associated with biocompatibility, biodegradation, or migration of individual bare crystals in tissue. To overcome these limitations, we have embedded LiNc-BuO crystals in polydimethylsiloxane (PDMS), an oxygen-permeable biocompatible polymer and developed an implantable/retrievable form of chip, called OxyChip. The chip was optimized for maximum spin density (40% w/w of LiNc-BuO in PDMS) and fabricated in a form suitable for implantation using an 18-G syringe needle. In vitro evaluation of the OxyChip showed that it is robust and highly oxygen sensitive. The dependence of its EPR linewidth to oxygen was linear and highly reproducible. In vivo efficacy of the OxyChip was evaluated by implanting it in rat femoris muscle and following its response to tissue oxygenation for up to 12 months. The results revealed preservation of the integrity (size and shape) and calibration (oxygen sensitivity) of the OxyChip throughout the implantation period. Further, no inflammatory or adverse reaction around the implantation area was observed thereby establishing its biocompatibility and safety. Overall, the results demonstrated that the newly-fabricated high-sensitive OxyChip is capable of providing long-term measurements of oxygen concentration in a reliable and repeated manner under clinical conditions.


Asunto(s)
Oximetría/métodos , Animales , Dimetilpolisiloxanos , Espectroscopía de Resonancia por Spin del Electrón , Masculino , Músculos/metabolismo , Oxígeno/metabolismo , Ratas , Ratas Wistar , Factores de Tiempo
6.
Radiat Res ; 189(5): 519-528, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29474156

RESUMEN

Endothelial nitric oxide synthase (eNOS), a constitutive enzyme expressed in vascular endothelial cells, is the main source of nitric oxide (NO), which plays key roles in diverse biological functions, including regulation of vascular tone. Exposure to radiation has been known to generate nitric oxide from eNOS; however, the precise mechanism of its generation and function is not known. The goal of this study was to determine the involvement of radiation-induced DNA damage response (DDR) on eNOS transcription and its effect on cell survival after irradiation. Irradiated bovine aortic endothelial cells showed increased eNOS transcription and NO generation through upregulation of ataxia-telangiectasia mutated (ATM) kinase. Radiation exposure induced NO inhibited cell death, as well as induced cellular senescence postirradiation. This study established that radiation-induced DDR uses ATM kinase to upregulate eNOS transcription and NO generation, leading to cellular senescence, which may play a critical role in radiation-mediated cardiovascular injury.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Células Endoteliales/metabolismo , Células Endoteliales/efectos de la radiación , Regulación Enzimológica de la Expresión Génica/efectos de la radiación , Óxido Nítrico Sintasa de Tipo III/genética , Tolerancia a Radiación , Animales , Bovinos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de la radiación , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Senescencia Celular/efectos de los fármacos , Senescencia Celular/efectos de la radiación , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Citoesqueleto/efectos de la radiación , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Activación Enzimática/efectos de la radiación , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Óxido Nítrico/farmacología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Tolerancia a Radiación/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Transcripción Genética/efectos de la radiación
7.
J Biol Chem ; 293(12): 4262-4276, 2018 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-29382728

RESUMEN

p53 is an important tumor-suppressor protein that is mutated in more than 50% of cancers. Strategies for restoring normal p53 function are complicated by the oncogenic properties of mutant p53 and have not met with clinical success. To counteract mutant p53 activity, a variety of drugs with the potential to reconvert mutant p53 to an active wildtype form have been developed. However, these drugs are associated with various negative effects such as cellular toxicity, nonspecific binding to other proteins, and inability to induce a wildtype p53 response in cancer tissue. Here, we report on the effects of a curcumin analog, HO-3867, on p53 activity in cancer cells from different origins. We found that HO-3867 covalently binds to mutant p53, initiates a wildtype p53-like anticancer genetic response, is exclusively cytotoxic toward cancer cells, and exhibits high anticancer efficacy in tumor models. In conclusion, HO-3867 is a p53 mutant-reactivating drug with high clinical anticancer potential.


Asunto(s)
Apoptosis/efectos de los fármacos , Curcumina/análogos & derivados , Proteínas Mutantes/genética , Mutación , Neoplasias/patología , Piperidonas/farmacología , Proteína p53 Supresora de Tumor/genética , Animales , Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Curcumina/farmacología , Femenino , Humanos , Ratones , Ratones Desnudos , Proteínas Mutantes/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Med Chem ; 13(8): 761-772, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28530545

RESUMEN

BACKGROUND: Natural products and their derivatives are widely used to treat cancer and other diseases associated with ROS- and RNS-induced damages. METHODS: A series of paramagnetic modified curcumin analogs and 3,5-diarylidene-piperidones (DAP) have been designed, synthesized, and characterized on their anti-proliferative and antioxidant activity. RESULTS: Biological characterization of the new compounds supported the earlier results that incorporation of a nitroxide moiety or its precursor into curcumin or diarylidenylpiperidone (DAP) scaffolds resulted in anti-proliferative effect toward cancerous cell-lines in case of aryl hydroxy and/or methoxy substituent containing derivatives, suggesting their potential for targeted therapeutic applications. In case of basic side chain derivatives, nitroxide incorporation gave unambiguous results, however in tendency the more accessible DAP derivatives had stronger anti-proliferative effect. In most cases, the nitroxide incorporation increased the TEAC value (proton and electron donation capability) of DAP derivatives. CONCLUSIONS: Among the compounds synthesized and investigated the spin-labeled curcumin and 3,5-bis(4-hydroxy-3-methoxybenzylidene)piperidin-4-one derivatives were the most effective antiproliferative and antioxidant derivatives.


Asunto(s)
Antineoplásicos/farmacología , Antioxidantes/farmacología , Benzotiazoles/antagonistas & inhibidores , Curcumina/farmacología , Óxidos de Nitrógeno/farmacología , Ácidos Sulfónicos/antagonistas & inhibidores , Antineoplásicos/síntesis química , Antineoplásicos/química , Antioxidantes/síntesis química , Antioxidantes/química , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Curcumina/química , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Estructura Molecular , Óxidos de Nitrógeno/química , Relación Estructura-Actividad
9.
PLoS One ; 9(12): e116281, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25551230

RESUMEN

INTRODUCTION: Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) have recently been shown to express key cardiac proteins and improve in vivo cardiac function when administered following myocardial infarction. However, the efficacy of hiPSC-derived cell therapies, in direct comparison to current, well-established stem cell-based therapies, is yet to be elucidated. The goal of the current study was to compare the therapeutic efficacy of human mesenchymal stem cells (hMSCs) with hiPSC-CMs in mitigating myocardial infarction (MI). METHODS: Male athymic nude hyrats were subjected to permanent ligation of the left-anterior-descending (LAD) coronary artery to induce acute MI. Four experimental groups were studied: 1) control (non-MI), 2) MI, 3) hMSCs (MI+MSC), and 4) hiPSC-CMs (MI+hiPSC-derived cardiomyocytes). The hiPSC-CMs and hMSCs were labeled with superparamagnetic iron oxide (SPIO) in vitro to track the transplanted cells in the ischemic heart by high-field cardiac MRI. These cells were injected into the ischemic heart 30-min after LAD ligation. Four-weeks after MI, cardiac MRI was performed to track the transplanted cells in the infarct heart. Additionally, echocardiography (M-mode) was performed to evaluate the cardiac function. Immunohistological and western blot studies were performed to assess the cell tracking, engraftment and cardiac fibrosis in the infarct heart tissues. RESULTS: Echocardiography data showed a significantly improved cardiac function in the hiPSC-CMs and hMSCs groups, when compared to MI. Immunohistological studies showed expression of connexin-43, α-actinin and myosin heavy chain in engrafted hiPSC-CMs. Cardiac fibrosis was significantly decreased in hiPSC-CMs group when compared to hMSCs or MI groups. Overall, this study demonstrated improved cardiac function with decreased fibrosis with both hiPSC-CMs and hMSCs groups when compared with MI group.


Asunto(s)
Células Madre Pluripotentes Inducidas/fisiología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/fisiología , Infarto del Miocardio/patología , Miocitos Cardíacos/fisiología , Animales , Biomarcadores/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Ecocardiografía , Humanos , Imagen por Resonancia Magnética , Masculino , Infarto del Miocardio/terapia , Ratas Desnudas
10.
J Cell Biochem ; 114(11): 2612-23, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23794477

RESUMEN

Myocardial infarction is a leading cause of mortality and morbidity worldwide. Occlusion of a coronary artery produces ischemia and myocardial necrosis that leads to left ventricular (LV) remodeling, dysfunction, and heart failure. Stem cell therapy may decrease infarct size and improve LV function; the hypoxic environment, however, following a myocardial infarction may result in apoptosis, which in turn decreases survival of transplanted stem cells. Therefore, the effects of preconditioned mesenchymal stem cells (MSC) with hyperoxia (100% oxygen), Z-VAD-FMK pan-caspase inhibitor (CI), or both in a hypoxic environment in order to mimic conditions seen in cardiac tissue post-myocardial infarction were studied in vitro. MSCs preconditioned with hyperoxia or CI significantly decreased apoptosis as suggested by TUNEL assay and Annexin V analysis using fluorescence assisted cell sorting. These effects were more profound when both, hyperoxia and CI, were used. Additionally, gene and protein expression of caspases 1, 3, 6, 7, and 9 were down-regulated significantly in MSCs preconditioned with hyperoxia, CI, or both, while the survival markers Akt1, NF-κB, and Bcl-2 were significantly increased in preconditioned MSCs. These changes ultimately resulted in a significant increase in MSC proliferation in hypoxic environment as determined by BrdU assays compared to MSCs without preconditioning. These effects may prove to be of great clinical significance when transplanting stem cells into the hypoxic myocardium of post-myocardial infarction patients in order to attenuate LV remodeling and improve LV function.


Asunto(s)
Hipoxia de la Célula/fisiología , Proliferación Celular/efectos de los fármacos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/enzimología , Clorometilcetonas de Aminoácidos/farmacología , Animales , Caspasa 1/genética , Caspasa 1/metabolismo , Caspasa 3/genética , Caspasa 3/metabolismo , Caspasa 6/genética , Caspasa 6/metabolismo , Caspasa 7/genética , Caspasa 7/metabolismo , Caspasa 9/genética , Caspasa 9/metabolismo , Células Cultivadas , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Ratas
11.
Oxid Med Cell Longev ; 2013: 279847, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23766850

RESUMEN

The neutrophil elastase inhibitor sivelestat (ONO-5046) possesses unknown mechanisms of cardioprotection when infused following global ischemia, even in the absence of neutrophils. Since myocardial ischemia-reperfusion injury is strongly associated with endothelial dysfunction and reactive oxygen species (ROS) generation during reperfusion, we have tested the hypothesis that infusion of sivelestat during postischemic low flow would preserve endothelial and contractile function and reduce infarct size through an ROS-mediated mechanism. Isolated male rat hearts, subjected to global ischemia of 25 minutes, were reperfused with low flow with or without sivelestat followed by a full flow reperfusion. Hearts treated with sivelestat showed a significant improvement of LV contractile function and a reduction in infarct size. Infusion of L-NAME (nonspecific blocker of endothelial nitric oxide synthase (eNOS)) along with sivelestat during reperfusion reversed the preservation of contractile function and infarct size. In vitro EPR spin trapping experiments showed that sivelestat treatment decreased superoxide adduct formation in bovine aortic endothelial cells (BAECs) subjected to hypoxia-reoxygenation. Similarly, dihydroethidine (DHE) staining showed decreased superoxide production in LV sections from sivelestat-treated hearts. Taken together, these results indicate that sivelestat infusion during postischemic low flow reduces infarct size and preserves vasoreactivity in association with decreased ROS formation and the preservation of nitric oxide.


Asunto(s)
Circulación Coronaria/efectos de los fármacos , Glicina/análogos & derivados , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Daño por Reperfusión Miocárdica/fisiopatología , Sulfonamidas/administración & dosificación , Sulfonamidas/uso terapéutico , Animales , Aorta/patología , Cardiotónicos/farmacología , Cardiotónicos/uso terapéutico , Bovinos , Creatina Quinasa/metabolismo , Espectroscopía de Resonancia por Spin del Electrón , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/patología , Glicina/administración & dosificación , Glicina/farmacología , Glicina/uso terapéutico , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/patología , Ventrículos Cardíacos/fisiopatología , Técnicas In Vitro , Masculino , Modelos Cardiovasculares , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Daño por Reperfusión Miocárdica/enzimología , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo III/metabolismo , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Sulfonamidas/farmacología , Superóxidos/metabolismo , Vasoconstricción/efectos de los fármacos , Función Ventricular Izquierda/efectos de los fármacos
12.
J Pharmacol Exp Ther ; 343(1): 62-71, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22739507

RESUMEN

Adult stem cells have shown great promise toward repairing infarcted heart and restoring cardiac function. Mesenchymal stem cells (MSCs), because of their inherent multipotent nature and their ability to secrete a multitude of growth factors and cytokines, have been used for cardiac repair with encouraging results. Preclinical studies showed that MSCs injected into infarcted hearts improve cardiac function and attenuate fibrosis. Although stem cell transplantation is a promising therapeutic option to repair the infarcted heart, it is faced with a number of challenges, including the survival of the transplanted cells in the ischemic region, due to excessive oxidative stress present in the ischemic region. The objective of this study was to determine the effect of Carvedilol (Carv), a nonselective ß-blocker with antioxidant properties, on the survival and engraftment of MSCs in the infarcted heart. MSCs were subjected to a simulated host-tissue environment, similar to the one present in the infarcted myocardium, by culturing them in the presence of hydrogen peroxide (H(2)O(2)) to induce oxidative stress. MSCs were treated with 2.5 µM Carv for 1 h in serum-free medium, followed by treatment with H(2)O(2) for 2 h. The treated cells exhibited significant protection against H(2)O(2)-induced cell death versus untreated controls as determined by 3-(4,5-dimethylthiazolyl-2)-2,5-diphenyltetrazolium bromide and terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling assays. Likewise, transplantation of MSCs after permanent left coronary artery ligation and treatment of animals after myocardial infarction (MI) with Carv (5 mg/kg b.wt.) led to significant improvement in cardiac function, decreased fibrosis, and caspase-3 expression compared with the MI or MSC-alone groups.


Asunto(s)
Carbazoles/uso terapéutico , Caspasa 3/biosíntesis , Inhibidores de Caspasas/uso terapéutico , Regulación Enzimológica de la Expresión Génica , Trasplante de Células Madre Mesenquimatosas/métodos , Infarto del Miocardio/terapia , Propanolaminas/uso terapéutico , Animales , Carbazoles/farmacología , Carvedilol , Inhibidores de Caspasas/farmacología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Terapia Combinada , Regulación hacia Abajo/fisiología , Sinergismo Farmacológico , Infarto del Miocardio/enzimología , Infarto del Miocardio/patología , Propanolaminas/farmacología , Distribución Aleatoria , Ratas , Ratas Endogámicas F344 , Regulación hacia Arriba/fisiología
13.
Cancer Biol Ther ; 12(9): 837-45, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-21885917

RESUMEN

Cisplatin resistance is a major obstacle in the treatment of ovarian cancer. Drug combinations with synergistic or complementary functions are a promising strategy to overcome this issue. We studied the anticancer efficacy of a novel compound, HO-3867, used in combination with cisplatin against chemotherapy-resistant ovarian cancer. A2780R cells, a cisplatin-resistant human ovarian cancer cell line, were exposed to 1, 5, or 10 uM of HO-3867 alone or in combination with cisplatin (10 ug/ml) for 24 hours. Cell viability (MTT), proliferation (BrdU), cell-cycle analysis (FACS), and protein expression (western blot) were used for in vitro studies. STAT3 overexpression was performed using transfected STAT3 cDNA. In vivo studies used cisplatin-resistant xenograft tumors grown in nude mice and treated with 100-ppm HO-3867 and weekly injections of 4-mg/kg cisplatin. HO-3867/cisplatin combination treatment significantly inhibited cisplatin-resistant cell proliferation in a concentration-dependent manner. The inhibition was associated with increased expression of p53 and p21, and decreased expression of cdk5 and cyclin D1. Apoptosis was induced by activation of Bax, cytochrome c release, and stimulated cleavage of caspase-9, caspase-3, and PARP. Overexpression of STAT3 decreased the HO-3867-induced apoptosis. The combination treatment significantly inhibited the growth of cisplatin-resistant xenograft tumors with significant downregulation of pSTAT3, and without apparent toxicity to healthy tissues. The combination treatment exhibited synergistic anticancer efficacy, which appears largely due to HO-3867-induced downregulation of pSTAT3. The results, combined with the previously-reported safety features of HO-3867, suggest the potential use of this compound as a safe and effective adjuvant for the treatment of ovarian cancer.


Asunto(s)
Antineoplásicos/farmacología , Cisplatino/farmacología , Curcumina/farmacología , Neoplasias Ováricas/metabolismo , Piperidonas/farmacología , Factor de Transcripción STAT3/antagonistas & inhibidores , Animales , Antineoplásicos/uso terapéutico , Antineoplásicos/toxicidad , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/metabolismo , Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cisplatino/uso terapéutico , Cisplatino/toxicidad , Curcumina/análogos & derivados , Curcumina/uso terapéutico , Curcumina/toxicidad , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias Ováricas/tratamiento farmacológico , Piperidonas/uso terapéutico , Piperidonas/toxicidad , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
14.
J Pharmacol Exp Ther ; 339(2): 350-7, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21799049

RESUMEN

Doxorubicin (DOX) is a drug commonly used for the treatment of cancer. The development of resistance to DOX is common, and high cumulative doses cause potentially lethal cardiac side effects. HO-3867 (3,5-bis(4-fluorobenzylidene)-1-[(2,2,5,5-tetramethyl-2,5-dihydro-1-hydroxy-pyrrol-3-yl)methyl]piperidin-4-one), a synthetic curcumin analog, has been shown to exhibit both anticancer and cardioprotective effects. However, its cardioprotection in the setting of a conventional cancer therapy has not been established. This work investigated the use of HO-3867 and DOX to achieve a complementary outcome, i.e., increased toxicity toward cancer cells, and reduced cardiac toxicity. Combination treatment was investigated using DOX-resistant MCF-7 breast cancer cells [MCF-7 multidrug-resistant (MDR)] and BALB/c mice. Lower doses of HO-3867 and DOX (5 and 2.5 µM, respectively) reduced viability of MCF-7 MDR cells to an extent significantly greater than that when either drug was used alone, an effect equivalent to that induced by exposure to 50 µM DOX. In normal cardiac cells, the loss of viability from combination treatment was significantly lower than that induced by 50 µM DOX. Increases in apoptotic markers, e.g., cleaved caspase-3, and decreases in fatty acid synthase and pAkt expressions were observed by Western blotting. Mice treated with both HO-3867 and DOX showed significant improvement in cardiac functional parameters compared with mice treated with DOX alone. Reduced expression of Bcl-2 and pAkt was observed in mice treated with DOX alone, whereas mice given combination treatment showed levels similar to control. The study indicates that combination treatment of HO-3867 and DOX is a viable option for treatment of cancer with reduced cardiotoxic side effects.


Asunto(s)
Antineoplásicos/toxicidad , Cardiotónicos/farmacología , Doxorrubicina/toxicidad , Piperidonas/farmacología , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/uso terapéutico , Antioxidantes/administración & dosificación , Antioxidantes/farmacología , Antioxidantes/uso terapéutico , Aorta/citología , Aorta/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Cardiotónicos/administración & dosificación , Cardiotónicos/uso terapéutico , Caspasas/biosíntesis , Línea Celular Tumoral , Ciclinas/antagonistas & inhibidores , Ciclinas/biosíntesis , Doxorrubicina/administración & dosificación , Doxorrubicina/uso terapéutico , Resistencia a Múltiples Medicamentos , Resistencia a Antineoplásicos , Ecocardiografía , Femenino , Corazón/efectos de los fármacos , Cardiopatías/tratamiento farmacológico , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Piperidonas/administración & dosificación , Piperidonas/uso terapéutico , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/biosíntesis , Receptor fas/antagonistas & inhibidores , Receptor fas/biosíntesis
15.
J Med Chem ; 54(15): 5414-21, 2011 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-21702507

RESUMEN

A series of 3,5-bis(arylidene)-4-piperidone (DAP) compounds are considered as synthetic analogues of curcumin for anticancer properties. We performed structure-activity relationship studies by synthesizing a number of DAPs N-alkylated or acylated with nitroxides or their amine precursors as potent antioxidant moieties. Both subtituents on arylidene rings and on piperidone nitrogen (five- or six-membered, 2- or 3-substituted or 3,4-disubstituted isoindoline nitroxides) were varied. The anticancer efficacy of the new DAP compounds was tested by measuring their cytotoxicity to cancer cell lines A2780 and MCF-7 and to the H9c2 cell line. The results showed that all DAP compounds induced a significant loss of cell viability in the human cancer cell lines tested; however, only pyrroline appended nitroxides (5c (Selvendiran, K.; Tong, L.; Bratasz, A.; Kuppusamy, L. M.; Ahmed, S.; Ravi, Y.; Trigg, N. J.; Rivera, B. K.; Kálai, T.; Hideg, K.; Kuppusamy, P. Mol. Cancer Ther. 2010, 9, 1169-1179), 5e, 7, 9) showed limited toxicity toward noncancerous cell lines. Computer docking simulations support the biological activity tested. These results suggest that antioxidant-conjugated DAPs will be useful as a safe and effective anticancer agent for cancer therapy.


Asunto(s)
Antineoplásicos/síntesis química , Antioxidantes/síntesis química , Curcumina/análogos & derivados , Piperidonas/síntesis química , Antineoplásicos/farmacología , Antioxidantes/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Simulación por Computador , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Piperidonas/farmacología , Relación Estructura-Actividad
16.
Adv Exp Med Biol ; 701: 29-36, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21445766

RESUMEN

A new paramagnetic crystalline material, namely, lithium naphthalocyanine (LiNc), whose electron-paramagnetic-resonance (EPR) line width is highly sensitive to oxygen content, has been evaluated for use as oximetry probe in cells and tissues. Previously,we reported on the synthesis, structural framework,magnetic and oxygen-sensing properties of LiNc microcrystalline powder (Pandian et al, J. Mater. Chem. 19, 4138, 2009). The material exhibited a single, sharp EPR line that showed a highly linear response of its width to surrounding molecular oxygen (pO(2)) with a sensitivity of 31.2 mG/mmHg. In the present study, we evaluated the suitability of this material for in vivo oximetry in biological systems. We observed that the probe was stable in tissues for more than two months without any adverse effect on its oxygen-sensing properties. We further demonstrated that the probe can be prepared in sub-micron sizes for uptake by stem cells. Thus, the high oxygen sensitivity, biocompatibility, and long-term stability in tissues may be useful for high-resolution EPR oximetry.


Asunto(s)
Espectroscopía de Resonancia por Spin del Electrón , Oximetría/instrumentación , Oxígeno/metabolismo , Porfirinas/química , Marcadores de Spin , Animales , Cristalización , Femenino , Ratones , Ratones Endogámicos C3H , Ratas , Ratas Endogámicas F344
17.
Am J Physiol Cell Physiol ; 299(6): C1562-70, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20861473

RESUMEN

Stem cells transplanted to the ischemic myocardium usually encounter massive cell death within a few days of therapy. Hypoxic preconditioning (HPC) is currently employed as a strategy to prepare stem cells for increased survival and engraftment in the heart. However, HPC of stem cells has provided varying results, supposedly due to the differences in the oxygen concentration, duration of exposure, and passage conditions. In the present study, we determined the effect of HPC on rat mesenchymal stem cells (MSCs) exposed to 0.5% oxygen concentration for 24, 48, or 72 h. We evaluated the expression of prosurvival, proangiogenic, and functional markers such as hypoxia-inducible factor-1α, VEGF, phosphorylated Akt, survivin, p21, cytochrome c, caspase-3, caspase-7, CXCR4, and c-Met. MSCs exposed to 24-h hypoxia showed reduced apoptosis on being subjected to severe hypoxic conditions. They also had significantly higher levels of prosurvival, proangiogenic, and prodifferentiation proteins when compared with longer exposure (72 h). Cells taken directly from the cryopreserved state did not respond effectively to the 24-h HPC as those that were cultured under normoxia before HPC. Cells cultured under normoxia before HPC showed decreased apoptosis, enhanced expression of connexin-43, cardiac myosin heavy chain, and CD31. The preconditioned cells were able to differentiate into the cardiovascular lineage. The results suggest that MSCs cultured under normoxia before 24-h HPC are in a state of optimal expression of prosurvival, proangiogenic, and functional proteins that may increase the survival and engraftment in the infarct heart. These results could provide further insights into optimal preparation of MSCs which would greatly influence the effectiveness of cell therapy in vivo.


Asunto(s)
Precondicionamiento Isquémico Miocárdico , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/metabolismo , Neovascularización Fisiológica , Animales , Apoptosis , Caspasa 3/análisis , Caspasa 7/análisis , Hipoxia de la Célula , Línea Celular , Conexina 43/análisis , Citocromos c/análisis , Corazón , Subunidad alfa del Factor 1 Inducible por Hipoxia/análisis , Proteínas Asociadas a Microtúbulos/análisis , Cadenas Pesadas de Miosina/análisis , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/análisis , Proteínas Proto-Oncogénicas c-akt/análisis , Proteínas Proto-Oncogénicas c-met , Ratas , Receptores CXCR4/análisis , Survivin , Factor A de Crecimiento Endotelial Vascular/análisis , Proteínas de Unión al GTP rho/análisis
18.
Cancer Biol Ther ; 10(10): 1027-32, 2010 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-20798598

RESUMEN

Curcumin, a naturally-occurring compound found in the rhizome of Curcuma longa plant, is known for its antitumor activities. However, its clinical efficacy is limited due to poor bioabsorption. A new class of synthetic analogs of curcumin, namely diarylidenylpiperidone (DAP), has been developed with substantially higher anticancer activity than curcumin. However, its cellular uptake and bioabsorption have not been evaluated. In this study we have determined the absorption of a representative DAP compound, HO-3867, using optical and electron paramagnetic resonance spectrometry. The cellular uptake of HO-3867 was measured in a variety of cancer cell lines. HO-3867 was taken in cells within 15 minutes of exposure and its uptake was more than 100-fold higher than curcumin. HO-3867 was also retained in cells in an active form for 72 hours and possibly longer. HO-3867 was substantially cytotoxic to all the cancer cells tested. However, there was no direct correlation between cellular uptake and cytotoxicity suggesting that the cytotoxic mechanisms could be cell-type specific. When administered to rats by intraperitoneal injection, significantly high levels of HO-3867 were found in the liver, kidney, stomach, and blood after 3 hours. Also, significant accumulation of HO-3867 was found in murine tumor xenografts with a dose-dependent inhibition of tumor growth. The results suggest that the curcumin analog has substantially higher bioabsorption when compared to curcumin.


Asunto(s)
Antineoplásicos/uso terapéutico , Curcumina/uso terapéutico , Neoplasias Ováricas/tratamiento farmacológico , Piperidonas/farmacocinética , Piperidonas/uso terapéutico , Animales , Espectroscopía de Resonancia por Spin del Electrón , Femenino , Halogenación , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Ratas , Ratas Wistar , Distribución Tisular , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Mol Cancer Res ; 8(9): 1188-97, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20713491

RESUMEN

Fatty acid synthase (FAS) and focal adhesion kinase (FAK), which are overexpressed in a variety of human epithelial tumors, play a key role in the migration and invasion of cancer cells. Hence, strategies targeted at inhibiting the FAS/FAK proteins may have therapeutic potential for cancer treatment. The goal of the present study was to determine the effect of HO-3867, a synthetic compound, on the migratory ability of ovarian cancer cells and to understand the mechanistic pathways including the involvement of FAS, FAK, and associated signaling proteins. The study was done using two established human ovarian cancer cell lines, A2780 and SKOV3. Incubation with 10 µmol/L HO-3867 for 24 hours significantly inhibited the native as well as the vascular endothelial growth factor (VEGF)-mediated migration and invasion of the cells. HO-3867 significantly attenuated FAS and FAK protein levels apparently through accelerated ubiquitin-dependent degradation, as shown by a clear downregulation of isopeptidase USP2a. Exposure of cells to HO-3867 also significantly inhibited FAS activity and mRNA levels and a number of downstream proteins, including phospho-extracellular signal-regulated kinase 1/2, phospho-human epidermal growth factor receptor 1, sterol regulatory element binding protein 1, VEGF, and matrix metalloproteinase 2. Western blot and immunohistochemical analyses of A2780 xenograft tumors in mice treated with HO-3867 showed significant reduction in FAS, FAK, VEGF, and downstream protein levels when compared with the untreated control. Collectively, the results showed that HO-3867 suppressed the migration and invasion of ovarian cancer cells by inhibiting the expression or activity of FAS and FAK proteins. The study suggests that molecular targeting of FAS and FAK by HO-3867 may be a potential strategy for ovarian cancer therapy.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Ácido Graso Sintasas/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Neoplasias Ováricas/enzimología , Neoplasias Ováricas/patología , Piperidonas/farmacología , Animales , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Invasividad Neoplásica/prevención & control , Neoplasias Ováricas/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Cancer Biol Ther ; 10(4): 386-90, 2010 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-20562529

RESUMEN

Hypoxia, which is commonly observed in many solid tumors, is a major impediment to chemo- or radiation therapy. Hypoxia is also known to overexpress/activate signal transducer and activator of transcription 3 (STAT3) leading to tumor progression as well as drug resistance. We hypothesized that increased oxygenation of the hypoxic tumor may have an inhibitory effect on STAT3 activation and hence tumor-growth inhibition. Mice containing human ovarian cancer xenograft tumor were exposed to hyperbaric oxygen (HBO; 100% oxygen; 2 atm; 90-min duration) daily, for up to 21 days. Mice exposed to HBO showed a significant reduction in tumor volume, with no effect on body weight. STAT3 (Tyr 705) activation and cyclin-D1 protein/mRNA levels were significantly decreased up on HBO exposure. Interestingly, HBO exposure, in combination with weekly administration of cisplatin, also significantly reduced the tumor volume; however, this group of mice had drastically reduced body weight when compared to other groups. While conventional wisdom might suggest that increased oxygenation of tumors would promote tumor growth, the results of the present study indicated otherwise. Hyperoxia appears to inhibit STAT3 activation, which is a key step in the ovarian tumor progression. The study may have important implications for the treatment of ovarian cancer in the clinic.


Asunto(s)
Proliferación Celular , Ciclina D1/metabolismo , Oxigenoterapia Hiperbárica , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/terapia , Factor de Transcripción STAT3/metabolismo , Animales , Antineoplásicos/uso terapéutico , Hipoxia de la Célula , Línea Celular Tumoral , Cisplatino/uso terapéutico , Terapia Combinada , Ciclina D1/genética , Regulación hacia Abajo , Resistencia a Antineoplásicos , Femenino , Humanos , Ratones , Fosforilación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT3/antagonistas & inhibidores , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA