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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
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
2.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
Mol Cell Biochem ; 333(1-2): 9-26, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19585224

RESUMEN

Vascular endothelium is vulnerable to the attack of glucose-derived oxoaldehydes (glyoxal and methylglyoxal) during diabetes, through the formation of advanced glycation end products (AGEs). Although aminoguanidine (AG) has been shown to protect against the AGE-induced adverse effects, its protection against the glyoxal-induced alterations in vascular endothelial cells (ECs) such as cytotoxicity, barrier dysfunction, and inhibition of angiogenesis has not been reported and we investigated this in the bovine pulmonary artery ECs (BPAECs). The results showed that glyoxal (1-10 mM) significantly induced cytotoxicity and mitochondrial dysfunction in a dose- and time-dependent (4-12 h) fashion in ECs. Glyoxal was also observed to significantly inhibit EC proliferation. The study also revealed that glyoxal induced EC barrier dysfunction (loss of trans-endothelial electrical resistance), actin cytoskeletal rearrangement, and tight junction alterations in BPAECs. Furthermore, the results revealed that glyoxal significantly inhibited in vitro angiogenesis on the Matrigel. For the first time, this study demonstrated that AG significantly protected against the glyoxal-induced cytotoxicity, barrier dysfunction, cytoskeletal rearrangement, and inhibition of angiogenesis in BPAECs. Therefore, AG appears as a promising protective agent in the treatment of AGE-induced vascular endothelial alterations and dysfunction during diabetes, presumably by blocking the reactivity of the sugar-derived dicarbonyls such as glyoxal and preventing the formation of AGEs.


Asunto(s)
Citoesqueleto/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Glioxal/farmacología , Guanidinas/farmacología , Neovascularización Fisiológica/efectos de los fármacos , Animales , Bovinos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Citoesqueleto/patología , Diabetes Mellitus/tratamiento farmacológico , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patología , Células Endoteliales/ultraestructura , Endotelio Vascular/citología , Productos Finales de Glicación Avanzada , Guanidinas/uso terapéutico , Óxido Nítrico Sintasa/antagonistas & inhibidores , Sustancias Protectoras
10.
Bioorg Med Chem ; 18(8): 2954-63, 2010 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-20347319

RESUMEN

New derivatives of verapamil (1) modified with nitroxides and their precursors were synthesized and screened for reactive oxygen species (ROS)-scavenging activities. The basic structure was modified by changing the nitrile group to an amide or the methyl substituent on tertiary nitrogen with nitroxides and their reduced forms (hydroxylamine and secondary amines). Among the new verapamil derivatives compound 16B [Mohan, I. K.; Kahn, M.; Wisel, S.; Selvendiran, K.; Sridhar, A.; Carnes, C.A.; Bognár, B.; Kálai, T.; Hideg, K.; Kuppusamy, P. Am. J. Physiol. Heart Circ. Physiol.2009, 296, 140], modified with hydroxylamine salt of 2,2,6,6-tetramethyl-1,2,3,6-tetrahydropyridine-1-yloxyl proved to be the best ROS scavenger in vitro and protected HSMC and CHO cells against H(2)O(2) induced damage.


Asunto(s)
Cardiotónicos/síntesis química , Depuradores de Radicales Libres/síntesis química , Magnetismo , Verapamilo/química , Animales , Células CHO , Cardiotónicos/química , Cardiotónicos/farmacología , Línea Celular , Cricetinae , Cricetulus , Espectroscopía de Resonancia por Spin del Electrón , Depuradores de Radicales Libres/química , Depuradores de Radicales Libres/farmacología , Humanos , Peróxido de Hidrógeno/farmacología , Verapamilo/análogos & derivados , Verapamilo/síntesis química , Verapamilo/farmacología
11.
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
12.
J Mol Cell Cardiol ; 47(2): 275-87, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19376124

RESUMEN

A major limitation to the application of stem-cell therapy to repair ischemic heart damage is the low survival of transplanted cells in the heart, possibly due to poor oxygenation. We hypothesized that hyperbaric oxygenation (HBO) can be used as an adjuvant treatment to augment stem-cell therapy. Therefore, the goal of this study was to evaluate the effect of HBO on the engraftment of rat bone marrow-derived mesenchymal stem cells (MSCs) transplanted in infarct rat hearts. Myocardial infarction (MI) was induced in Fisher-344 rats by permanently ligating the left-anterior-descending coronary artery. MSCs, labeled with fluorescent superparamagnetic iron oxide (SPIO) particles, were transplanted in the infarct and peri-infarct regions of the MI hearts. HBO (100% oxygen at 2 ATA for 90 min) was administered daily for 2 weeks. Four MI groups were used: untreated (MI); HBO; MSC; MSC+HBO. Echocardiography, electro-vectorcardiography, and magnetic resonance imaging were used for functional evaluations. The engraftment of transplanted MSCs in the heart was confirmed by SPIO fluorescence and Prussian-blue staining. Immunohistochemical staining was used to identify key cellular and molecular markers including CD29, troponin-T, connexin-43, VEGF, alpha-smooth-muscle actin, and von Willebrand factor in the tissue. Compared to MI and MSC groups, the MSC+HBO group showed a significantly increased recovery of cardiac function including left-ventricular (LV) ejection fraction, fraction shortening, LV wall thickness, and QRS vector. Further, HBO treatment significantly increased the engraftment of CD29-positive cells, expression of connexin-43, troponin-T and VEGF, and angiogenesis in the infarct tissue. Thus, HBO appears to be a potential and clinically-viable adjuvant treatment for myocardial stem-cell therapy.


Asunto(s)
Oxigenoterapia Hiperbárica , Trasplante de Células Madre Mesenquimatosas , Infarto del Miocardio/fisiopatología , Infarto del Miocardio/terapia , Recuperación de la Función , Potenciales de Acción , Animales , Vasos Sanguíneos/patología , Conexina 43/metabolismo , Compuestos Férricos/metabolismo , Fibrosis , Integrina beta1/metabolismo , Miocardio/patología , Neovascularización Fisiológica , Presión Parcial , Ratas , Ratas Endogámicas F344 , Troponina T/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Función Ventricular Izquierda
13.
Int J Cancer ; 125(9): 2198-204, 2009 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-19623660

RESUMEN

Signal transducer and activator of transcription 3 (STAT3) is activated in a variety of human cancers, including ovarian cancer. The molecular mechanism by which the STAT3 is activated in cancer cells is poorly understood. We observed that human ovarian xenograft tumors (A2780) in mice were severely hypoxic (pO(2) approximately 2 mmHg). We further observed that hypoxic exposure significantly increased the phosphorylation of STAT3 (pSTAT3) at the Tyr705 residue in A2780 cell line. The pSTAT3 (Tyr705) level was highly dependent on cellular oxygenation levels, with a significant increase at <2% O(2), and without any change in the pSTAT3 (Ser727) or total STAT3 levels. The pSTAT3 (Tyr705) elevation following hypoxic exposure could be reversed within 12 hr after returning the cells to normoxia. The increased level of pSTAT3 was partly mediated by increased levels of reactive oxygen species generation in the hypoxic cancer cells. Conventional chemotherapeutic drugs cisplatin and taxol were far less effective in eliminating the hypoxic ovarian cancer cells suggesting a role for pSTAT3 in cellular resistance to chemotherapy. Inhibition of STAT3 by AG490 followed by treatment with cisplatin or taxol resulted in a significant increase in apoptosis suggesting that hypoxia-induced STAT3 activation is responsible for chemoresistance. The results have important clinical implications for the treatment of hypoxic ovarian tumors using STAT3-specific inhibitors.


Asunto(s)
Hipoxia de la Célula , Neoplasias Ováricas/tratamiento farmacológico , Factor de Transcripción STAT3/fisiología , Línea Celular Tumoral , Proliferación Celular , Resistencia a Antineoplásicos , Femenino , Humanos , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Oxígeno/farmacología , Especies Reactivas de Oxígeno/metabolismo , Factor de Transcripción STAT3/antagonistas & inhibidores
14.
J Pharmacol Exp Ther ; 329(2): 543-50, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19218529

RESUMEN

Stem cell transplantation is a possible therapeutic option to repair ischemic damage to the heart. However, it is faced with a number of challenges including the survival of the transplanted cells in the ischemic region. The present study was designed to use stem cells preconditioned with trimetazidine (1-[2,3,4-trimethoxybenzyl]piperazine; TMZ), a widely used anti-ischemic drug for treating angina in cardiac patients, to increase the rate of their survival after transplantation. Bone marrow-derived rat mesenchymal stem cells (MSCs) were subjected to a simulated host tissue environment by culturing them under hypoxia (2% O(2)) and using hydrogen peroxide (H(2)O(2)) to induce oxidative stress. MSCs were preconditioned with 10 microM TMZ for 6 h followed by treatment with 100 microM H(2)O(2) for 1 h and characterized for their cellular viability and metabolic activity. The preconditioned cells showed a significant protection against H(2)O(2)-induced loss of cellular viability, membrane damage, and oxygen metabolism accompanied by a significant increase in HIF-1alpha, survivin, phosphorylated Akt (pAkt), and Bcl-2 protein levels and Bcl-2 gene expression. The therapeutic efficacy of the TMZ-preconditioned MSCs was evaluated in an in vivo rat model of myocardial infarction induced by permanent ligation of left anterior descending coronary artery. A significant increase in the recovery of myocardial function and up-regulation of pAkt and Bcl-2 levels were observed in hearts transplanted with TMZ-preconditioned cells. This study clearly demonstrated the potential benefits of pharmacological preconditioning of MSCs with TMZ for stem cell therapy for repairing myocardial ischemic damage.


Asunto(s)
Ciclina D1/biosíntesis , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/efectos de los fármacos , Infarto del Miocardio/terapia , Estrés Oxidativo/efectos de los fármacos , Trimetazidina/uso terapéutico , Vasodilatadores/uso terapéutico , Animales , Western Blotting , Hipoxia de la Célula/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Células Madre Mesenquimatosas/metabolismo , Infarto del Miocardio/metabolismo , Consumo de Oxígeno , Ratas , Ratas Endogámicas F344 , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trimetazidina/administración & dosificación , Trimetazidina/farmacología , Vasodilatadores/administración & dosificación , Vasodilatadores/farmacología
15.
J Pharmacol Exp Ther ; 329(3): 959-66, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19276401

RESUMEN

Phosphatase and tensin homolog (PTEN), a tumor suppressor gene, has been shown to play a vital role in vascular smooth muscle cell (SMC) proliferation and hence is a potential therapeutic target to inhibit vascular remodeling. The goal of this study was to evaluate the efficacy and mechanism of HO-3867 [((3E,5E)-3,5-bis[(4-fluorophenyl)methylidene]-1-[(1-hydroxy-2,2,5,5-tetramethyl-2,5-dihydro-1H-pyrrol-3-yl)methyl]piperidin-4-one)], a new synthetic curcuminoid, in the inhibition of vascular SMC proliferation and restenosis. Experiments were performed using human aortic SMCs and a rat carotid artery balloon injury model. HO-3867 (10 microM) significantly inhibited the proliferation of serum-stimulated SMCs by inducing cell cycle arrest at the G(1) phase (72% at 24 h) and apoptosis (at 48 h). HO-3867 significantly increased the phosphorylated and total levels of PTEN in SMCs. Suppression of PTEN expression by PTEN-small interfering RNA transfection reduced p53 and p21 levels and increased extracellular signal-regulated kinase 1/2 phosphorylation, resulting in decreased apoptosis. Conversely, overexpression of PTEN by cDNA transfection activated caspase-3 and increased apoptosis. Furthermore, HO-3867 significantly down-regulated matrix metalloproteinase (MMP)-2, MMP-9, and nuclear factor (NF)-kappaB expressions in SMCs. Finally, HO-3867 inhibited arterial neointimal hyperplasia through overexpression of PTEN and down-regulation of MMPs and NF-kappaB proteins. HO-3867 is a potent drug, capable of overexpressing PTEN, which is a key target in the prevention of vascular remodeling, including restenosis.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Reestenosis Coronaria/prevención & control , Curcumina/análogos & derivados , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/efectos de los fármacos , Fosfohidrolasa PTEN/metabolismo , Piperidonas/farmacología , Factor de Transcripción Activador 2/metabolismo , Animales , Apoptosis/fisiología , Compuestos de Bencilideno/farmacología , Compuestos de Bencilideno/uso terapéutico , Arterias Carótidas/efectos de los fármacos , Arterias Carótidas/metabolismo , Arterias Carótidas/patología , Arterias Carótidas/cirugía , Caspasa 3/metabolismo , Ciclo Celular/efectos de los fármacos , Células Cultivadas , Reestenosis Coronaria/metabolismo , Reestenosis Coronaria/patología , Curcumina/farmacología , Curcumina/uso terapéutico , Fase G1/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Inhibidores de Crecimiento/farmacología , Inhibidores de Crecimiento/uso terapéutico , Humanos , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Estructura Molecular , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , FN-kappa B/metabolismo , Fosfohidrolasa PTEN/genética , Piperidonas/uso terapéutico , ARN Interferente Pequeño/genética , Ratas , Ratas Sprague-Dawley
16.
Int J Toxicol ; 28(3): 190-206, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19546257

RESUMEN

Earlier, we reported that mercury, the environmental risk factor for cardiovascular diseases, activates vascular endothelial cell (EC) phospholipase D (PLD). Here, we report the novel and significant finding that calcium and calmodulin regulated mercury-induced PLD activation in bovine pulmonary artery ECs (BPAECs). Mercury (mercury chloride, 25 microM; thimerosal, 25 microM; methylmercury, 10 microM) significantly activated PLD in BPAECs. Calcium chelating agents and calcium depletion of the medium completely attenuated the mercury-induced PLD activation in ECs. Calmodulin inhibitors significantly attenuated mercury-induced PLD activation in BPAECs. Despite the absence of L-type calcium channels in ECs, nifedipine, nimodipine, and diltiazem significantly attenuated mercury-induced PLD activation and cytotoxicity in BPAECs. This study demonstrated the importance of calcium and calmodulin in the regulation of mercury-induced PLD activation and the protective action of L-type calcium channel blockers against mercury cytotoxicity in vascular ECs, suggesting mechanisms of mercury vasculotoxicity and mercury-induced cardiovascular diseases.


Asunto(s)
Calcio/fisiología , Calmodulina/metabolismo , Células Endoteliales/efectos de los fármacos , Cloruro de Mercurio/toxicidad , Compuestos de Metilmercurio/toxicidad , Fosfolipasa D/metabolismo , Timerosal/toxicidad , Animales , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo L/efectos de los fármacos , Canales de Calcio Tipo L/metabolismo , Señalización del Calcio/efectos de los fármacos , Calmodulina/antagonistas & inhibidores , Bovinos , Muerte Celular , Células Cultivadas , Quelantes/farmacología , Células Endoteliales/metabolismo , Activación Enzimática
17.
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
18.
Adv Exp Med Biol ; 614: 45-52, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18290313

RESUMEN

We have developed a noninvasive EPR (electron paramagnetic resonance) oximetry, based on a new class of oxygen-sensing nano-particulate probe (LiNc-BuO), for simultaneous monitoring of stem-cell therapy and in situ oxygenation (partial pressure of oxygen, pO2) in a mouse model of acute myocardial infarction (AMI). AMI was induced by a permanent occlusion of left-anterior-descending (LAD) coronary artery. Skeletal myoblast (SM) cells were used for therapy. The oximetry probe was implanted in the mid-ventricular region using a needle. Tissue histological studies after 3 weeks of implantation of the probe revealed significant fibrosis, which was solely due to the needle track and not due to the probe particles. The feasibility of long-term monitoring of pO2 was established in control (non-infarct) group of hearts (> 3 months; pO2 = 15.0 +/- 1.2 mmHg,). A mixture of the probe with/without SM cells (1 x 10(5)) was implanted as a single injection in the infarcted region and the myocardial tissue pO2 at the site of cell therapy was measured for 4 weeks. The pO2 was significantly higher in infarcted hearts treated with SM cells (pO2 = 3.5 +/- 0.9 mmHg) compared to untreated hearts (pO2 = 1.6 +/- 0.7 mmHg). We have demonstrated, for the first time, the feasibility of monitoring pO2 in mouse hearts after stem cell therapy.


Asunto(s)
Modelos Animales de Enfermedad , Espectroscopía de Resonancia por Spin del Electrón/métodos , Infarto del Miocardio/fisiopatología , Oxígeno/análisis , Trasplante de Células Madre , Animales , Técnicas de Cultivo de Célula , Células Cultivadas , Medios de Cultivo , Estudios de Factibilidad , Miembro Posterior , Masculino , Ratones , Ratones Endogámicos C57BL , Mioblastos Esqueléticos/metabolismo , Mioblastos Esqueléticos/trasplante , Infarto del Miocardio/etiología , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Miocardio/metabolismo , Miocardio/patología , Oximetría , Consumo de Oxígeno , Presión Parcial , Factores de Tiempo
19.
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.

20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA