Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Gynecol Oncol ; 135(1): 133-41, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25038288

RESUMEN

OBJECTIVE: Constitutive activation of STAT3 is a hallmark of various human cancers, however an increased pSTAT3 expression in high grade human endometrial cancer has not been reported. In the present study, we examine the expression of STAT family of proteins in endometrial cancer cell lines and the efficacy of HO-3867, a novel STAT3 inhibitor designed in our lab. METHODS: Expression of STAT family proteins was evaluated via Western blot. The cell viability, post-treatment with HO-3867, was assessed using MTT, cell-cycle profile and Annexin assay. In vivo efficacy of HO-3867 was evaluated using xenograft mice. RESULTS: Expression of activated STATs was inconsistent among the cell lines and 18 human endometrial cancer specimens tested. While pSTAT3 Tyr705 was not expressed in any of the cell lines, pSTAT3 Ser727 was highly expressed in endometrial cancer cell lines and tumor specimens. HO-3867 decreased the expression of pSTAT3 Ser727 while total STAT3 remained constant; cell viability decreased by 50-80% and induced G2/M arrest in 55% of Ishikawa cells at the G2/M cell cycle checkpoint. There was an increase in p53, a decrease in Bcl2 and Bcl-xL, and cleavage of caspase-3, caspase-7 and PARP. HO-3867 mediated a dosage-dependent inhibition of the growth of xenografted endometrial tumors. CONCLUSIONS: HO-3867 treatment decreases the high levels of pSTAT3 Ser727 in endometrial cancer cells by inducing cell cycle arrest and apoptosis. This suggests a specific role of serine-phosphorylated STAT3, independent of tyrosine phosphorylation in the oncogenesis of endometrial cancer. HO-3867 could potentially serve as an adjunctive targeted therapy.


Asunto(s)
Neoplasias Endometriales/tratamiento farmacológico , Neoplasias Endometriales/metabolismo , Piperidonas/uso terapéutico , Factor de Transcripción STAT3/antagonistas & inhibidores , Factor de Transcripción STAT3/biosíntesis , Animales , Línea Celular Tumoral , Femenino , Humanos , Ratones
2.
PLoS One ; 9(3): e93208, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24675768

RESUMEN

Chemotherapy constitutes the standard modality of treatment for localized head and neck squamous cell carcinomas (HNSCC). However, many patients fail to respond and relapse after this treatments due to the acquisition of chemo-resistance. Therefore, there is an urgent need to develop novel drugs that could reverse the resistant phenotype. Curcumin, the constituent of the spice turmeric has been shown to have anti-inflammatory, anti-oxidant and anti-proliferative properties in several tumor types. However, use of curcumin has been limited due to its poor bio-absorption. Recently, a novel class of curcumin analogs, based on diarylidenylpiperidones (DAP), has been developed by incorporating a piperidone link to the beta-diketone structure and fluoro substitutions on the phenyl groups. In this study, we evaluated the effectiveness of H-4073, a parafluorinated variant of DAP, using both in vitro and in vivo head and neck cancer models. Our results demonstrate that H-4073 is a potent anti-tumor agent and it significantly inhibited cell proliferation in all the HNSCC cell lines tested in a dose-dependent manner. In addition, pretreatment of cisplatin-resistant HNSCC cell lines with H-4073 significantly reversed the chemo-resistance as observed by cell viability assay (MTT), apoptosis assay (Annexin V binding) and cleaved caspase-3 (Western blot). H-4073 mediated its anti-tumor effects by inhibiting JAK/STAT3, FAK, Akt and VEGF signaling pathways that play important roles in cell proliferation, migration, survival and angiogenesis. In the SCID mouse xenograft model, H-4073 significantly enhanced the anti-tumor and anti-angiogenesis effects of cisplatin, with no added systemic toxicity. Interestingly, H-4073 inhibited tumor angiogenesis by blocking VEGF production by tumor cells as well as directly inhibiting endothelial cell function. Taken together, our results suggest that H-4073 is a potent anti-tumor agent and it can be used to overcome chemotherapy resistance in HNSCC.


Asunto(s)
Antineoplásicos/farmacología , Cisplatino/farmacología , Curcumina/farmacología , Neoplasias de Cabeza y Cuello/patología , Animales , Antineoplásicos/administración & dosificación , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cisplatino/administración & dosificación , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/metabolismo , Humanos , Ratones , Neovascularización Patológica/tratamiento farmacológico , Fosforilación , Factor de Transcripción STAT3/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
3.
J Ovarian Res ; 6(1): 35, 2013 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-23663277

RESUMEN

A diagnosis of advanced ovarian cancer is the beginning of a long and arduous journey for a patient. Worldwide, approximately half of the individuals undergoing therapy for advanced cancer will succumb to the disease, or consequences of treatment. Well-known and widely-used chemotherapeutic agents such as cisplatin, paclitaxel, 5-fluorouracil, and doxorubicin are toxic to both cancer and non-cancerous cells, and have debilitating side effects Therefore, development of new targeted anticancer therapies that can selectively kill cancer cells while sparing the surrounding healthy tissues is essential to develop more effective therapies. We have developed a new class of synthetic curcumin analogs, diarylidenyl-piperidones (DAPs), which have higher anticancer activity and enhanced bio-absorption than curcumin. The DAP backbone structure exhibits cytotoxic (anticancer) activity, whereas the N-hydroxypyrroline (-NOH) moiety found on some variants functions as a cellular- or tissue-specific modulator (antioxidant) of cytotoxicity. The anticancer activity of the DAPs has been evaluated using a number of ovarian cancer cell lines, and the safety has been evaluated in a number of non-cancerous cell lines. Both variations of the DAP compounds showed similar levels of cell death in ovarian cancer cells, however the compounds with the -NOH modification were less toxic to non-cancerous cells. The selective cytotoxicity of the DAP-NOH compounds suggests that they will be useful as safe and effective anticancer agents. This article reviews some of the key findings of our work with the DAP compounds, and compares this to some of the targeted therapies currently used in ovarian cancer therapy.

4.
Cardiovasc Res ; 93(1): 89-99, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-22012955

RESUMEN

AIMS: Myocardial infarction (MI) is associated with irreversible loss of viable cardiomyocytes. Cell therapy is a potential option to replace the lost cardiomyocytes and restore cardiac function. However, cell therapy is faced with a number of challenges, including survival of the transplanted cells in the infarct region, which is characterized by abundant levels of oxidants and lack of a pro-survival support mechanism. The goal of the present study was to evaluate the effect of supplemental oxygenation on cell engraftment and functional recovery in a rat model. METHODS AND RESULTS: MI was induced in rats by a 60-min occlusion of the coronary artery, followed by restoration of flow. Mesenchymal stem cells (MSCs), isolated from adult rat bone marrow, were transplanted in the MI region. Rats with transplanted MSCs were exposed to hyperbaric oxygen (HBO: 100% O(2), 2 atmospheres absolute) for 90 min, 5 days/week for 4 weeks. The experimental groups were: MI (control), Ox (MI + HBO), MSC (MI + MSC), and MSC + Ox (MI + MSC + HBO). HBO exposure (oxygenation) was started 3 days after induction of MI. MSCs were transplanted 1 week after induction of MI. Echocardiography showed a significant recovery of cardiac function in the MSC + Ox group, when compared with the MI or MSC group. Oxygenation increased the engraftment of MSCs and vascular density in the infarct region. Molecular analysis of infarct tissue showed a four-fold increase in NOS3 expression in the MSC + Ox group compared with the MI group. CONCLUSIONS: The results showed that post-MI exposure of rats to daily cycles of hyperoxygenation (oxygen cycling) improved stem cell engraftment, cardiac function, and increased NOS3 expression.


Asunto(s)
Oxigenoterapia Hiperbárica , Trasplante de Células Madre Mesenquimatosas , Infarto del Miocardio/terapia , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Animales , Secuencia de Bases , Terapia Combinada , Conexina 43/metabolismo , Cartilla de ADN/genética , Fibrosis , Expresión Génica , Masculino , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Neovascularización Fisiológica , Ratas , Ratas Endogámicas F344
5.
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
6.
Adv Exp Med Biol ; 701: 175-81, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21445785

RESUMEN

Stem-cell transplantation to treat acute myocardial infarction (MI) is gaining importance as a minimally invasive and potent therapy to replace akinetic scar tissue by viable myocardium. Our recent studies have shown that stem-cell transplantation marginally improves myocardial oxygenation in the infarct tissue leading to improvement in cardiac function. The aim of the present study was to determine the effect of hyperbaric oxygen (HBO) treatment on myocardial oxygenation and recovery of function in MI hearts. Fisher-344 rats were subjected to MI by permanently ligating the left-anterior-descending (LAD) coronary artery. The rats were then exposed to 100% O(2) at a pressure of 2 atmospheres for 90 minutes, and the exposure was repeated for 5 days a week for 2 weeks. Adult bone-marrow-derived rat mesenchymal stem cells (MSC, 5x105 cells) were mixed with OxySpin (LiNc- BuO, oxygen sensor) and implanted in the infarct and peri-infarct regions of the heart. M-mode ultrasound echocardiography was performed at baseline and at 2 weeks post-transplantation. The myocardial pO(2) in the MSC+HBO group (16.2±2.2 mmHg) was significantly higher when compared to untreated MI (3.8±1.9 mmHg) or MSC (9.8±2.3 mmHg) groups. In addition, there was a significant improvement in cardiac function, increased vessel density, and VEGF expression in MSC+HBO group compared to MSC group (p < 0.05). In conclusion, the results suggested a beneficial effect of HBO administration on stem-cell therapy for MI.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Oxigenoterapia Hiperbárica , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Infarto del Miocardio/terapia , Oxígeno/metabolismo , Animales , Médula Ósea/metabolismo , Técnicas para Inmunoenzimas , Ratas , Ratas Endogámicas F344 , Factor A de Crecimiento Endotelial Vascular/metabolismo
7.
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
8.
J Cardiovasc Pharmacol ; 56(5): 526-31, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20729753

RESUMEN

The objective of the present study was to evaluate the efficacy and mechanism of Crataegus oxycantha (COC) extract in preventing ischemia-reperfusion (IR) injury in an in vivo rat model of acute myocardial infarction induced by a 30-minute regional ischemia followed by 72 hours of reperfusion. The COC extract [100 mg/(kg body weight)] was administered 12 hours after the surgical procedure and then at 24-hour intervals for 3 days. Animals treated with COC extract showed a significant decrease in creatine kinase activity and infarct size. At the molecular level, COC administration resulted in a significant attenuation of PTEN (phosphatase and tensin homolog deleted on chromosome 10) and upregulation of phospho-Akt and c-Raf levels in the heart. As a consequence, cleaved caspase-9 and cleaved caspase-7 levels were significantly downregulated, indicating negative regulation of apoptosis by COC extract. In part with the hypoxia-inducible factor (HIF) signaling pathway, COC extract administration significantly upregulated the prolyl hydroxylase-2 level. In contrast, other proapoptotic proteins such as nuclear factor-κB, cytochrome c, apoptosis-inducing factor, and cleaved poly(adenosine diphosphate-ribose) polymerase levels were significantly downregulated in the COC-treated group when compared with the untreated control group. The results suggested that COC extract attenuated apoptotic incidence in the experimental myocardial ischemia-reperfusion model by regulating Akt and HIF-1 signaling pathways.


Asunto(s)
Apoptosis/efectos de los fármacos , Crataegus/química , Factor 1 Inducible por Hipoxia/fisiología , Daño por Reperfusión Miocárdica/prevención & control , Extractos Vegetales/uso terapéutico , Proteínas Proto-Oncogénicas c-akt/fisiología , Transducción de Señal , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Creatina Quinasa/metabolismo , Prolina Dioxigenasas del Factor Inducible por Hipoxia , Masculino , Infarto del Miocardio/patología , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Miocardio/patología , Fitoterapia , Procolágeno-Prolina Dioxigenasa/metabolismo , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba
9.
Life Sci ; 87(9-10): 269-74, 2010 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-20600148

RESUMEN

Myocardial infarction (MI) is caused by deprivation of oxygen and nutrients to the cardiac tissue due to blockade of coronary artery. It is a major contributor to chronic heart disease, a leading cause of mortality in the modern world. Oxygen is required to meet the constant energy demands for heart contractility, and also plays an important role in the regulation of heart function. However, reoxygenation of the ischemic myocardium upon restoration of blood flow may lead to further injury. Controlled oxygen delivery during reperfusion has been advocated to prevent this consequence. Monitoring the myocardial oxygen concentration would play a vital role in understanding the pathological changes in the ischemic heart following myocardial infarction. During the last two decades, several new techniques have become available to monitor myocardial oxygen concentration in vivo. Electron paramagnetic resonance (EPR) oximetry would appear to be the most promising and reliable of these techniques. EPR utilizes crystalline probes which yield a single sharp line, the width of which is highly sensitive to oxygen tension. Decreased oxygen tension results in a sharpening of the EPR spectrum, while an increase results in widening. In our recent studies, we have used EPR oximetry as a valuable tool to monitor myocardial oxygenation for several applications like ischemia-reperfusion injury, stem-cell therapy and hyperbaric oxygen therapy. The results obtained from these studies have demonstrated the importance of tissue oxygen in the application of stem-cell therapy to treat ischemic heart tissues. These results have been summarized in this review article.


Asunto(s)
Oxigenoterapia Hiperbárica , Infarto del Miocardio/terapia , Miocardio/metabolismo , Consumo de Oxígeno/fisiología , Trasplante de Células Madre , Animales , Espectroscopía de Resonancia por Spin del Electrón , Humanos , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/cirugía , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/prevención & control , Miocardio/patología , Oximetría/métodos
10.
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
11.
J Cereb Blood Flow Metab ; 30(7): 1275-87, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20145654

RESUMEN

Acute ischemic stroke (AIS) results in focal deprivation of blood-borne factors, one of them being oxygen. The purpose of this study was two-fold: (1) to identify therapeutic conditions for supplemental oxygen in AIS and (2) to use transcriptome-wide screening toward uncovering oxygen-sensitive mechanisms. Transient MCAO in rodents was used to delineate the therapeutic potential of normobaric (NBO, 100% O(2), 1ATA) and hyperbaric oxygen (HBO, 100% O(2), 2ATA) during ischemia (iNBO, iHBO) and after reperfusion (rNBO, rHBO). Stroke lesion was quantified using 4.7 T MRI at 48 h. Supplemental oxygen during AIS significantly attenuated percent stroke hemisphere lesion volume as compared with that in room air (RA) controls, whereas identical treatment immediately after reperfusion exacerbated lesion volume (RA=22.4+/-1.8, iNBO=9.9+/-3.6, iHBO=6.6+/-4.8, rNBO=29.8+/-3.6, rHBO=35.4+/-7.6). iNBO and iHBO corrected penumbra tissue pO(2) during AIS as measured by EPR oxymetry. Unbiased query of oxygen-sensitive transcriptome in stroke-affected tissue identified 5,769 differentially expressed genes. Candidate genes were verified by real-time PCR using neurons laser-captured from the stroke-affected somatosensory cortex. Directed microarray analysis showed that supplemental oxygen limited leukocyte accumulation to the infarct site by attenuation of stroke-inducible proinflammatory chemokine response. The findings provide key information relevant to understanding oxygen-dependent molecular mechanisms in the AIS-affected brain.


Asunto(s)
Isquemia Encefálica , Regulación de la Expresión Génica , Oxigenoterapia Hiperbárica , Oxígeno/metabolismo , Accidente Cerebrovascular , Animales , Biomarcadores/metabolismo , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Isquemia Encefálica/terapia , Modelos Animales de Enfermedad , Humanos , Infarto de la Arteria Cerebral Media , Leucocitos/citología , Leucocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Estrés Oxidativo , Distribución Aleatoria , Ratas , Ratas Wistar , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/patología , Accidente Cerebrovascular/terapia
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.
Phytother Res ; 19(12): 1030-7, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16372368

RESUMEN

The generation of reactive oxygen species and mitochondrial dysfunction has been implicated in doxorubicin (DOX)-induced cardiotoxicity. The aim of the present study was to determine whether Spirulina, a blue-green algae, could serve as a cardioprotective agent during DOX treatment in a mouse model. Mice were treated with DOX (4 mg/kg bw, intraperitoneally), weekly, for 4 weeks. Spirulina was administered orally for 3 days twice daily, then for 7 weeks along with the four equal injections of DOX. Cardiotoxicity was assessed, at 3 weeks after the end of the DOX-treatment period, by mortality, volume of ascites, liver congestion, oxidative stress and ultrastructural changes of heart tissue. The DOX-treated animals showed higher mortality (53%) and more ascites. Myocardial damage, as assessed by ultrastructural changes, showed loss of myofibrils, cytoplasmic vacuolization and mitochondrial swelling. Myocardial superoxide dismutase and glutathione peroxidase activities were decreased and lipid peroxidation was increased. Pretreatment with Spirulina significantly protected the mice from DOX-induced cardiotoxic effects as evidenced from lower mortality (26%), less ascites, lower levels of lipid peroxidation, normalization of antioxidant enzymes and ultrastructural studies showing minimal damage to the heart. In vitro cytotoxic studies using ovarian cancer cells demonstrated that Spirulina did not compromise the anti-tumor activity of doxorubicin. These results suggest that Spirulina has a protective effect against cardiotoxicity induced by DOX and it may, therefore, improve the therapeutic index of DOX.


Asunto(s)
Antibióticos Antineoplásicos/efectos adversos , Proteínas Bacterianas/farmacología , Cardiomiopatía Dilatada/prevención & control , Doxorrubicina/efectos adversos , Corazón/efectos de los fármacos , Animales , Antibióticos Antineoplásicos/farmacología , Proteínas Bacterianas/uso terapéutico , Cardiomiopatía Dilatada/inducido químicamente , Cardiomiopatía Dilatada/patología , Cianobacterias , Doxorrubicina/farmacología , Interacciones Farmacológicas , Femenino , Ratones , Miocardio/metabolismo , Miocardio/patología , Miocardio/ultraestructura , Estrés Oxidativo/efectos de los fármacos , Ficocianina/metabolismo , Spirulina , Superóxidos/metabolismo
14.
Mutat Res ; 579(1-2): 172-81, 2005 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-16105672

RESUMEN

Hypoxia, caused by disrupted vasculature and peripheral vasculopathies, is a key factor that limits dermal wound healing. Factors that can increase oxygen delivery to the regional tissue, such as supplemental oxygen, warmth, and sympathetic blockade, can accelerate healing. Clinical experience with adjunctive hyperbaric oxygen therapy (HBOT) in the treatment of chronic wounds have shown that wound hyperoxia may increase granulation tissue formation and accelerate wound contraction and secondary closure. However, HBOT is not applicable to all wound patients and may pose the risk of oxygen toxicity. Thus, the efficacy of topical oxygen treatment in an experimental setting using the pre-clinical model involving excisional dermal wound in pigs was assessed. Exposure of open dermal wounds to topical oxygen treatment increased tissue pO2 of superficial wound tissue. Repeated treatment accelerated wound closure. Histological studies revealed that the wounds benefited from the treatment. The oxygen treated wounds showed signs of improved angiogenesis and tissue oxygenation. Topically applied pure oxygen has the potential of benefiting some wound types. Further studies testing the potential of topical oxygen in pre-clinical and clinical settings are warranted.


Asunto(s)
Oxígeno/uso terapéutico , Piel/lesiones , Cicatrización de Heridas , Administración Tópica , Animales , Femenino , Oxigenoterapia Hiperbárica/métodos , Queratina-14 , Queratinas/metabolismo , Neovascularización Fisiológica , Oxígeno/administración & dosificación , Piel/irrigación sanguínea , Piel/metabolismo , Procedimientos Quirúrgicos Operativos , Porcinos
15.
Am J Physiol Cell Physiol ; 287(5): C1472-81, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15475520

RESUMEN

Mild and nonlethal heat shock (i.e., hyperthermia) is known to protect the myocardium and cardiomyocytes against ischemic injury. In the present study, we have shown that heat shock regulates the respiration of cultured neonatal cardiomyocytes (cardiac H9c2 cells) through activation of nitric oxide synthase (NOS). The respiration of cultured cardiac H9c2 cells subjected to mild heat shock at 42 degrees C for 1 h was decreased compared with that of control. The O2 concentration at which the rate of O2 consumption is reduced to 50% was increased in heat-shocked cells, indicating a lowering of O2 affinity in the mitochondria. Western blot analyses showed a fourfold increase in the expression of heat shock protein (HSP) 90 and a twofold increase in endothelial NOS (eNOS) expression in the heat-shocked cells. Immunoblots of eNOS, inducible NOS (iNOS), and neuronal NOS (nNOS) in the immunoprecipitate of HSP90 of heat-shocked cells showed that there was a sevenfold increase in eNOS and no changes in iNOS and nNOS. Confocal microscopic analysis of cells stained with the NO-specific fluorescent dye 4,5-diaminofluorescein diacetate showed higher levels of NO production in the heat-shocked cells than in control cells. The results indicate that heat shock-induced HSP90 forms a complex with eNOS and activates it to increase NO concentration in the cardiac H9c2 cells. The generated NO competitively binds to the complexes of the respiratory chain of the mitochondria to downregulate O2 consumption in heat-shocked cells. On the basis of these results, we conclude that myocardial protection by hyperthermia occurs at least partly by the pathway of HSP90-mediated NO production, leading to subsequent attenuation of cellular respiration.


Asunto(s)
Respiración de la Célula/fisiología , Proteínas HSP90 de Choque Térmico/biosíntesis , Miocitos Cardíacos/fisiología , Óxido Nítrico Sintasa/metabolismo , Consumo de Oxígeno/fisiología , Animales , Western Blotting , Células Cultivadas , Hipertermia Inducida , Microscopía Confocal , Mitocondrias , Óxido Nítrico/biosíntesis , Ratas , Transducción de Señal/fisiología , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA