Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Drug Deliv ; 28(1): 1962-1971, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34565273

RESUMEN

Betulinic acid (3ß-Hydroxy-20(29)-lupaene-28-oic acid, BA) has excellent anti-cancer activity but poor solubility and low bioavailability. To improve the antitumor activity of BA, a polyvinyl caprolactam-polyvinyl acetate-polyethylene glycol (PVCL-PVA-PEG) graft copolymer (Soluplus) encapsulated BA micelle (Soluplus-BA) was fabricated. The Soluplus-BA micelles presented a mean size of 54.77 ± 1.26 nm and a polydispersity index (PDI) of 0.083. The MTT assay results showed that Soluplus-BA micelles increased the inhibitory effect of BA on MDA-MB-231 cells, mainly due to the enhanced accumulation of reactive oxygen species (ROS) and the destruction of mitochondrial membrane potential (MMP). Soluplus-BA micelles induced the DNA double-strand breaks (DSBs) as the γH2AX foci increased. Moreover, Soluplus-BA also inhibited the tube formation and migration of human umbilical vein endothelial cells (HUVECs), and inhibited the neovascularization of the chicken chorioallantoic membrane (CAM). This angiogenesis inhibitory effect may be accomplished by regulating the HIF-1/VEGF-FAK signaling pathway. The in vivo study confirmed the improved anti-tumor effect of Soluplus-BA and its inhibitory effect on angiogenesis, demonstrating the possibility of Soluplus-BA as an effective anti-breast cancer drug delivery system.


Asunto(s)
Antineoplásicos/administración & dosificación , Neoplasias de la Mama/patología , Micelas , Triterpenos Pentacíclicos/administración & dosificación , Polietilenglicoles/química , Polivinilos/química , Animales , Animales no Consanguíneos , Antineoplásicos/farmacología , Línea Celular Tumoral , Química Farmacéutica/métodos , Pollos , Portadores de Fármacos/química , Células Endoteliales de la Vena Umbilical Humana , Humanos , Factor 1 Inducible por Hipoxia/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Neovascularización Patológica/metabolismo , Triterpenos Pentacíclicos/farmacología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Propiedades de Superficie , Factor A de Crecimiento Endotelial Vascular/efectos de los fármacos , Ácido Betulínico
2.
Pharmacology ; 106(9-10): 509-519, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34412054

RESUMEN

INTRODUCTION: Intrahepatic cholangiocarcinoma (ICC), which is difficult to diagnose and is usually fatal due to its late clinical presentation and a lack of effective treatment, has risen over the past decades but without much improvement in prognosis. OBJECTIVE: The study aimed to investigate the role of apatinib that targets vascular endothelial growth factor receptor-2 (VEGFR2) in ICC. METHODS: MTT assays, cell scratch assays, and tube formation assays were used to assess the effect of apatinib on human ICC cell line (HuCCT-1) and RBE cells proliferation, migration, and angiogenic capacity, respectively. Expression of vascular endothelial growth factor (VEGF), VEGFR2, signal transducer and activator of transcription factor 3 (STAT3), pSTAT3, and hypoxia inducible factor 1 subunit alpha (HIF-1α) pathway proteins was assessed using Western blotting and mRNA expression analysis in HuCCT-1 was performed using RT-qPCR assays. The pcDNA 3.1(-)-VEGFR2 and pcDNA 3.1(-)-HIF-1α were transfected into HuCCT-1 and RBE cells using Lipofectamine 2,000 to obtain overexpressed HuCCT-1 and RBE cells. RESULTS: We found that apatinib-inhibited proliferation, migration, and angiogenesis of HuCCT-1 and RBE cells in vitro in a dose-dependent manner. We also proved that apatinib effectively inhibits angiogenesis in tumor cells by blocking the expression of VEGF and VEGFR2 in these cells. In addition, we demonstrated that apatinib regulates the expression of STAT3 phosphorylation by inhibiting VEGFR2. Finally, we showed that apatinib regulates ICC angiogenesis and HIF-1α/VEGF expression via STAT3. CONCLUSIONS: Based on the above findings, we conclude that apatinib inhibits HuCCT-1 and RBE cell proliferation, migration, and tumor angiogenesis by inhibiting the VEGFR2/STAT3/HIF-1α axis signaling pathway. Apatinib can be a promising drug for ICC-targeted molecular therapy.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de los Conductos Biliares/patología , Colangiocarcinoma/patología , Neovascularización Patológica/patología , Piridinas/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Factor 1 Inducible por Hipoxia/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Factor de Transcripción 3/efectos de los fármacos , Receptor 2 de Factores de Crecimiento Endotelial Vascular/efectos de los fármacos
3.
Anesthesiology ; 135(1): 136-150, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-33914856

RESUMEN

BACKGROUND: Sepsis is one of the leading causes of mortality in intensive care units, and sedation in the intensive care unit during sepsis is usually performed intravenously. The inhalative anesthetic sevoflurane has been shown to elicit protective effects in various inflammatory studies, but its role in peritonitis-induced sepsis remains elusive. The hypothesis was that sevoflurane controls the neutrophil infiltration by stabilization of hypoxia-inducible factor 1α and elevated adenosine A2B receptor expression. METHODS: In mouse models of zymosan- and fecal-induced peritonitis, male mice were anesthetized with sevoflurane (2 volume percent, 30 min) after the onset of inflammation. Control animals received the solvent saline. The neutrophil counts and adhesion molecules on neutrophils in the peritoneal lavage of wild-type, adenosine A2B receptor -/-, and chimeric animals were determined by flow cytometry 4 h after stimulation. Cytokines and protein release were determined in the lavage. Further, the adenosine A2B receptor and its transcription factor hypoxia-inducible factor 1α were evaluated by real-time polymerase chain reaction and Western blot analysis 4 h after stimulation. RESULTS: Sevoflurane reduced the neutrophil counts in the peritoneal lavage (mean ± SD, 25 ± 17 × 105vs. 12 ± 7 × 105 neutrophils; P = 0.004; n = 19/17) by lower expression of various adhesion molecules on neutrophils of wild-type animals but not of adenosine A2B receptor -/- animals. The cytokines concentration (means ± SD, tumor necrosis factor α [pg/ml], 523 ± 227 vs. 281 ± 101; P = 0.002; n = 9/9) and protein extravasation (mean ± SD [mg/ml], 1.4 ± 0.3 vs. 0.8 ± 0.4; P = 0.002; n = 12/11) were also lower after sevoflurane only in the wild-type mice. Chimeric mice showed the required expression of the adenosine A2B receptor on the hematopoietic and nonhematopoietic compartments for the protective effects of the anesthetic. Sevoflurane induced the expression of hypoxia-inducible factor 1α and adenosine A2B receptor in the intestine, liver, and lung. CONCLUSIONS: Sevoflurane exerts various protective effects in two murine peritonitis-induced sepsis models. These protective effects were linked with a functional adenosine A2B receptor.


Asunto(s)
Factor 1 Inducible por Hipoxia/efectos de los fármacos , Peritonitis/complicaciones , Receptor de Adenosina A2B/efectos de los fármacos , Sepsis/etiología , Sepsis/prevención & control , Sevoflurano/farmacología , Transducción de Señal/efectos de los fármacos , Anestésicos por Inhalación/farmacología , Animales , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL
4.
Drug Test Anal ; 13(4): 794-816, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33458935

RESUMEN

The transcriptional activator hypoxia-inducible factor (HIF) is a vital arbitrator in the performance of cellular responses lacking oxygen supply in aerobic organisms. Because these compounds are capable of enhancing the organism's capacity for molecular oxygen transport, they possess great potential for abuse as a performance-enhancing agent in sports. A comprehensive study of the metabolic conversion of the most popular HIF stabilisers such as IOX2, IOX3 and IOX4 using equine liver microsomes (in vitro) is reported. The parents and their metabolites were identified and characterised by liquid chromatography-mass spectrometry in negative ionisation mode using a QExactive high-resolution mass spectrometer. Under the current experimental condition, a total of 10 metabolites for IOX2 (three phase I and seven phase II), nine metabolites for IOX3 (four phase I and five phase II) and five metabolites for IOX4 (three phase I and two phase II) were detected. The outcome of the present study is as follows: (1) all the three IOX candidates are prone to oxidation, results in subsequent monohydroxylated, and some dihydroxylated metabolites. (2) Besides oxidation, there is a possibility of hydrolysis and de-alkylation, which results in corresponding carboxylic acid and amide, respectively. (3) The glucuronide and sulphate conjugate of the parent drugs as well as the monohydroxylated analogues were observed in this study. The characterised in vitro metabolites can potentially serve as target analytes for doping control analysis.


Asunto(s)
Doping en los Deportes/prevención & control , Glicina/análogos & derivados , Isoquinolinas/metabolismo , Sustancias para Mejorar el Rendimiento/metabolismo , Detección de Abuso de Sustancias/métodos , Animales , Cromatografía Liquida/métodos , Glicina/análisis , Glicina/metabolismo , Caballos , Factor 1 Inducible por Hipoxia/efectos de los fármacos , Factor 1 Inducible por Hipoxia/metabolismo , Isoquinolinas/análisis , Microsomas Hepáticos/metabolismo , Sustancias para Mejorar el Rendimiento/análisis , Espectrometría de Masas en Tándem/métodos
5.
Am J Respir Crit Care Med ; 202(8): 1146-1158, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-32551816

RESUMEN

Rationale: Antenatal inflammation with placental dysfunction is strongly associated with high bronchopulmonary dysplasia (BPD) risk in preterm infants. Whether antenatal or postnatal HIF (hypoxia-inducible factor) augmentation can preserve lung structure and function and prevent pulmonary hypertension after intrauterine inflammation is controversial.Objectives: To determine whether antenatal or postnatal prolyl-hydroxylase inhibitor (PHi) therapy increases lung HIF expression, preserves lung growth and function, and prevents pulmonary hypertension in a rat model of chorioamnionitis-induced BPD caused by antenatal inflammation.Methods: Endotoxin (ETX) was administered to pregnant rats by intraamniotic injection at Embryonic Day 20, and pups were delivered by cesarean section at Embryonic Day 22. Selective PHi drugs, dimethyloxalylglycine or GSK360A, were administered into the amniotic space at Embryonic Day 20 or after birth by intraperitoneal injection for 2 weeks. Placentas and lung tissue were collected at birth for morphometric and Western blot measurements of HIF-1a, HIF-2a, VEGF (vascular endothelial growth factor), and eNOS (endothelial nitric oxide synthase) protein contents. At Day 14, lung function was assessed, and tissues were harvested to determine alveolarization by radial alveolar counts, pulmonary vessel density, and right ventricle hypertrophy (RVH).Measurements and Main Results: Antenatal PHi therapy preserves lung alveolar and vascular growth and lung function and prevents RVH after intrauterine ETX exposure. Antenatal administration of PHi markedly upregulates lung HIF-1a, HIF-2a, VEGF, and eNOS expression after ETX exposure.Conclusions: HIF augmentation improves lung structure and function, prevents RVH, and improves placental structure following antenatal ETX exposure. We speculate that antenatal or postnatal PHi therapy may provide novel strategies to prevent BPD due to antenatal inflammation.


Asunto(s)
Displasia Broncopulmonar/tratamiento farmacológico , Factor 1 Inducible por Hipoxia/metabolismo , Pulmón/efectos de los fármacos , Péptido PHI/farmacología , Preñez , Aminoácidos Dicarboxílicos/farmacología , Animales , Animales Recién Nacidos , Western Blotting , Displasia Broncopulmonar/metabolismo , Displasia Broncopulmonar/patología , Modelos Animales de Enfermedad , Endotoxinas/efectos adversos , Endotoxinas/farmacología , Femenino , Factor 1 Inducible por Hipoxia/efectos de los fármacos , Inmunohistoquímica , Técnicas In Vitro , Inyecciones Intralesiones , Pulmón/embriología , Embarazo , Atención Prenatal , Alveolos Pulmonares/efectos de los fármacos , Alveolos Pulmonares/crecimiento & desarrollo , Circulación Pulmonar/efectos de los fármacos , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Valores de Referencia , Pruebas de Función Respiratoria , Técnicas de Cultivo de Tejidos
6.
Cell ; 180(1): 6, 2020 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-31951520

RESUMEN

Erythropoietin (EPO) production in the kidney is regulated by the oxygen-sensing transcription factor HIF-1α, which is degraded under normoxic conditions by HIF-prolyl hydroxylase (HIF-PHD). Inhibition of HIF-PHD by roxadustat leads to increased EPO production, better iron absorption, and amelioration of anemia in chronic kidney disease (CKD).


Asunto(s)
Anemia/terapia , Glicina/análogos & derivados , Factor 1 Inducible por Hipoxia/metabolismo , Isoquinolinas/uso terapéutico , Anemia/metabolismo , Glicina/uso terapéutico , Humanos , Factor 1 Inducible por Hipoxia/efectos de los fármacos , Riñón/metabolismo , Riñón/patología , Prolil Hidroxilasas/efectos de los fármacos , Prolil Hidroxilasas/metabolismo , Insuficiencia Renal Crónica/tratamiento farmacológico , Insuficiencia Renal Crónica/metabolismo
7.
Mol Cell Biol ; 39(14)2019 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-31061092

RESUMEN

Intestinal epithelial cells (IECs) are exposed to the low-oxygen environment present in the lumen of the gut. These hypoxic conditions on one hand are fundamental for the survival of the commensal microbiota and, on the other hand, favor the formation of a selective semipermeable barrier, allowing IECs to transport essential nutrients/water while keeping the sterile internal compartments separated from the lumen containing commensals. The hypoxia-inducible factor (HIF) complex, which allows cells to respond and adapt to fluctuations in oxygen levels, has been described as a key regulator in maintaining IEC barrier function by regulating their tight junction integrity. In this study, we sought to better evaluate the mechanisms by which low oxygen conditions impact the barrier function of human IECs. By profiling miRNA expression in IECs under hypoxia, we identified microRNA 320a (miRNA-320a) as a novel barrier formation regulator. Using pharmacological inhibitors and short hairpin RNA-mediated silencing, we could demonstrate that expression of this microRNA (miRNA) was HIF dependent. Importantly, using overexpression and knockdown approaches of miRNA-320a, we could confirm its direct role in the regulation of barrier function in human IECs. These results reveal an important link between miRNA expression and barrier integrity, providing a novel insight into mechanisms of hypoxia-driven epithelial homeostasis.


Asunto(s)
Factor 1 Inducible por Hipoxia/metabolismo , Intestinos/citología , MicroARNs/genética , Hipoxia de la Célula/efectos de los fármacos , Línea Celular Tumoral , Células Epiteliales/química , Células Epiteliales/citología , Regulación de la Expresión Génica , Células HEK293 , Células HeLa , Humanos , Factor 1 Inducible por Hipoxia/efectos de los fármacos , Intestinos/química , ARN Interferente Pequeño/farmacología , Uniones Estrechas/metabolismo
8.
J Nephrol ; 32(4): 539-547, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30635875

RESUMEN

Acute kidney injury (AKI) as a result of ischaemia-reperfusion represents a major healthcare burden worldwide. Mortality rates from AKI in hospitalized patients are extremely high and have changed little despite decades of research and medical advances. In 1986, Murry et al. demonstrated for the first time the phenomenon of ischaemic preconditioning to protect against ischaemia-reperfusion injury (IRI). This seminal finding paved the way for a broad body of research, which attempted to understand and ultimately harness this phenomenon for human application. The ability of preconditioning to limit renal IRI has now been demonstrated in multiple different animal models. However, more than 30 years later, a safe and consistent method of protecting human organs, including the kidneys, against IRI is still not available. This review highlights agents which, despite strong preclinical data, have recently failed to reduce AKI in human trials. The multiple reasons which may have contributed to the failure to translate some of the promising findings to clinical therapies are discussed. Agents which hold promise in the clinic because of their recent efficacy in preclinical large animal models are also reviewed.


Asunto(s)
Lesión Renal Aguda/prevención & control , Precondicionamiento Isquémico , Riñón/irrigación sanguínea , Daño por Reperfusión/prevención & control , Acetilcisteína/uso terapéutico , Lesión Renal Aguda/etiología , Animales , Quelantes/farmacología , Modelos Animales de Enfermedad , Diuréticos Osmóticos/uso terapéutico , Determinación de Punto Final , Depuradores de Radicales Libres/uso terapéutico , Humanos , Factor 1 Inducible por Hipoxia/efectos de los fármacos , Precondicionamiento Isquémico/métodos , Manitol/uso terapéutico , Oligopéptidos/uso terapéutico , Daño por Reperfusión/complicaciones , Reproducibilidad de los Resultados , Investigación Biomédica Traslacional
9.
Life Sci ; 213: 126-133, 2018 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-30336147

RESUMEN

AIM: This study investigated the effects of atorvastatin (ATS) on the paraquat (PQ)-induced epithelial-mesenchymal transition (EMT) and the potential mechanism through hypoxia-inducible factor-1 alpha (HIF-1α). MAIN METHODS: Sprague-Dawley (SD) rats were randomly divided into a control group (n = 5), PQ group (n = 20), PQ + ATS L group (n = 20, ATS 20 mg/kg daily) and PQ + ATS H group (n = 20, ATS 40 mg/kg daily). All treated rats were given a 20% PQ solution (50 mg/kg) once by gavage and then sacrificed 12, 24, 72 and 168 h after PQ exposure. The A549 and RLE-6TN cell lines were treated with ATS, PQ or both for 24 h. Mesenchymal (α-SMA and vimentin) and epithelial (E-cadherin and ZO-1) cell marker expression was tested both in vivo and in vitro. The effects of ATS on HIF-1α and ß­catenin expression were also evaluated. KEY FINDINGS: ATS alleviated PQ poisoning-induced lung injury and pulmonary fibrosis in vivo. This effect was dose-dependent. ATS treatment attenuated the EMT by increasing the levels of the epithelial markers E-cadherin and ZO-1 and by decreasing the expression of the mesenchymal markers α-SMA and vimentin in both lung tissues and in vitro cell culture. In addition, ATS treatment may decrease the HIF-1α and ß­catenin levels both in vivo and in vitro. SIGNIFICANCE: In conclusion, ATS can attenuate PQ-induced pulmonary fibrosis. The mechanism may involve the downregulation of the HIF-1α/ß­catenin pathway and the inhibition of the PQ-induced EMT by ATS. ATS may be considered as a therapeutic agent for PQ poisoning-induced pulmonary fibrosis.


Asunto(s)
Atorvastatina/farmacología , Factor 1 Inducible por Hipoxia/efectos de los fármacos , Fibrosis Pulmonar/tratamiento farmacológico , Células A549 , Animales , Atorvastatina/metabolismo , Cadherinas/metabolismo , Línea Celular , Transición Epitelial-Mesenquimal/efectos de los fármacos , Humanos , Hipoxia , Factor 1 Inducible por Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/efectos de los fármacos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Pulmón/metabolismo , Lesión Pulmonar/tratamiento farmacológico , Lesión Pulmonar/metabolismo , Masculino , Paraquat/efectos adversos , Fibrosis Pulmonar/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Proteína de la Zonula Occludens-1/metabolismo
10.
Neuropharmacology ; 105: 228-240, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26801077

RESUMEN

Oxidative stress is a hallmark of ischemic stroke pathogenesis causing neuronal malfunction and cell death. Up-regulation of anti-oxidative genes through activation of the NF-E2-related transcription factor 2 (Nrf2) is one of the key mechanisms in cellular defense against oxidative stress. Fumaric acid esters (FAEs) represent a class of anti-oxidative and anti-inflammatory molecules that are already in clinical use for multiple sclerosis therapy. Purpose of this study was to investigate whether FAEs promote neuronal survival upon ischemia, and analyze putative underlying molecular mechanisms in neurons. Murine organotypic hippocampal slice cultures, and two neuronal cell lines were treated with dimethyl fumarate (DMF) and monomethyl fumarate (MMF). Ischemic conditions were generated by exposing cells and slice cultures to oxygen-glucose deprivation (OGD), and cell death was determined through propidium iodide staining. Treatment with both DMF and MMF immediately after OGD during reoxygenation strongly reduced cell death in hippocampal cultures ex vivo. Both DMF and MMF promoted neuronal survival in HT-22 and SH-SY5Y cell lines exposed to ischemic stress. DMF but not MMF activated the anti-oxidative Nrf2 pathway in neurons. Accordingly, Nrf2 knockdown in murine neurons abrogated the protective effect of DMF but not MMF. Moreover, FAEs did not activate the hypoxia-inducible factor (HIF) pathway suggesting that this pathway may not significantly contribute to FAE mediated neuroprotection. Our results may provide the basis for a new therapeutic approach to treat ischemic pathologies such as stroke with a drug that already has a broad safety record in humans.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Fumaratos/farmacología , Factor 1 Inducible por Hipoxia/efectos de los fármacos , Factor 2 Relacionado con NF-E2/efectos de los fármacos , Neuronas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Animales , Antiinflamatorios no Esteroideos/farmacología , Antioxidantes/farmacología , Isquemia Encefálica/patología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Ésteres/farmacología , Glucosa/deficiencia , Hipocampo/citología , Hipocampo/efectos de los fármacos , Humanos , Hipoxia/tratamiento farmacológico , Factor 1 Inducible por Hipoxia/genética , Ratones , Ratones Endogámicos C57BL , Factor 2 Relacionado con NF-E2/genética
11.
Stroke ; 46(9): 2576-83, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26304864

RESUMEN

BACKGROUND AND PURPOSE: Iron chelation therapy is emerging as a novel neuroprotective strategy. The mechanisms of neuroprotection are diverse and include both neuronal and vascular pathways. We sought to examine the effect of iron chelation on cerebrovascular function in healthy aging and to explore whether hypoxia-inducible transcription factor-1 activation may be temporally correlated with vascular changes. METHODS: We assessed cerebrovascular function (autoregulation, vasoreactivity, and neurovascular coupling) and serum concentrations of vascular endothelial growth factor and erythropoietin, as representative measures of hypoxia-inducible transcription factor-1 activation, during 6 hours of deferoxamine infusion in 24 young and 24 older healthy volunteers in a randomized, blinded, placebo-controlled cross-over study design. Cerebrovascular function was assessed using the transcranial Doppler ultrasound. Vascular endothelial growth factor and erythropoietin serum protein assays were conducted using the Meso Scale Discovery platform. RESULTS: Deferoxamine elicited a strong age- and time-dependent increase in the plasma concentrations of erythropoietin and vascular endothelial growth factor, which persisted ≤3 hours post infusion (age effect P=0.04; treatment×time P<0.01). Deferoxamine infusion also resulted in a significant time- and age-dependent improvement in cerebral vasoreactivity (treatment×time P<0.01; age P<0.01) and cerebral autoregulation (gain: age×time×treatment P=0.04). CONCLUSIONS: Deferoxamine infusion improved cerebrovascular function, particularly in older individuals. The temporal association between improved cerebrovascular function and increased serum vascular endothelial growth factor and erythropoietin concentrations is supportive of shared hypoxia-inducible transcription factor-1-regulated pathways. Therefore, pharmacological activation of hypoxia-inducible transcription factor-1 to enhance cerebrovascular function may be a promising neuroprotective strategy in acute and chronic ischemic syndromes, especially in elderly patients. CLINICAL TRIAL REGISTRATION: URL: http://www.clinicaltrials.gov. Unique identifier: NCT013655104.


Asunto(s)
Circulación Cerebrovascular/efectos de los fármacos , Terapia por Quelación/métodos , Deferoxamina/farmacología , Eritropoyetina/sangre , Factor 1 Inducible por Hipoxia/efectos de los fármacos , Sideróforos/farmacología , Factores de Crecimiento Endotelial Vascular/efectos de los fármacos , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Envejecimiento/efectos de los fármacos , Estudios Cruzados , Deferoxamina/administración & dosificación , Femenino , Hemodinámica/efectos de los fármacos , Humanos , Masculino , Persona de Mediana Edad , Sideróforos/administración & dosificación , Transducción de Señal , Resultado del Tratamiento , Ultrasonografía Doppler Transcraneal , Factores de Crecimiento Endotelial Vascular/sangre , Adulto Joven
13.
Int J Radiat Oncol Biol Phys ; 87(4): 753-60, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24035332

RESUMEN

PURPOSE: We explored changes in hypoxia-inducible factor 1 (HIF1) signaling during androgen deprivation therapy (ADT) of androgen-sensitive prostate cancer xenografts under conditions in which no significant change in immunostaining of the hypoxia marker pimonidazole had occurred. METHODS AND MATERIALS: Gene expression profiles of volume-matched androgen-exposed and androgen-deprived CWR22 xenografts, with similar pimonidazole-positive fractions, were compared. Direct targets of androgen receptor (AR) and HIF1 transcription factors were identified among the differentially expressed genes by using published lists. Biological processes affected by ADT were determined by gene ontology analysis. HIF1α protein expression in xenografts and biopsy samples from 35 patients receiving neoadjuvant ADT was assessed by immunohistochemistry. RESULTS: A total of 1344 genes showed more than 2-fold change in expression by ADT, including 35 downregulated and 5 upregulated HIF1 targets. Six genes were shared HIF1 and AR targets, and their downregulation was confirmed with quantitative RT-PCR. Significant suppression of the biological processes proliferation, metabolism, and stress response in androgen-deprived xenografts was found, consistent with tumor regression. Nineteen downregulated HIF1 targets were involved in those significant biological processes, most of them in metabolism. Four of these were shared AR and HIF1 targets, including genes encoding the regulatory glycolytic proteins HK2, PFKFB3, and SLC2A1. Most of the downregulated HIF1 targets were induced by hypoxia in androgen-responsive prostate cancer cell lines, confirming their role as hypoxia-responsive HIF1 targets in prostate cancer. Downregulation of HIF1 targets was consistent with the absence of HIF1α protein in xenografts and downregulation in patients by ADT (P<.001). CONCLUSIONS: AR repression by ADT may lead to downregulation of HIF1 signaling independently of hypoxic fraction, and this may contribute to tumor regression. HIF1α expression is probably not a useful hypoxia biomarker during ADT in prostate cancer.


Asunto(s)
Antagonistas de Andrógenos/uso terapéutico , Antineoplásicos Hormonales/uso terapéutico , Biomarcadores de Tumor , Hipoxia de la Célula/fisiología , Perfilación de la Expresión Génica/métodos , Factor 1 Inducible por Hipoxia/efectos de los fármacos , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/efectos de los fármacos , Anilidas/uso terapéutico , Animales , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Terapia Combinada/métodos , Regulación hacia Abajo/efectos de los fármacos , Perfilación de la Expresión Génica/estadística & datos numéricos , Goserelina/uso terapéutico , Humanos , Factor 1 Inducible por Hipoxia/genética , Factor 1 Inducible por Hipoxia/metabolismo , Quimioterapia de Inducción/métodos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Nitrilos/uso terapéutico , Antígeno Prostático Específico/sangre , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/radioterapia , Distribución Aleatoria , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Estrés Fisiológico/efectos de los fármacos , Estrés Fisiológico/fisiología , Compuestos de Tosilo/uso terapéutico , Transcripción Genética/efectos de los fármacos , Transcripción Genética/genética , Regulación hacia Arriba/efectos de los fármacos
14.
J Nat Prod ; 76(6): 1175-81, 2013 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-23731014

RESUMEN

The hypoxia-inducible factor-1 (HIF-1) transcription factor regulates cellular oxygen homeostasis. Agents that activate HIF-1 and downstream HIF targets represent potential drug leads for the prevention and/or treatment of ischemic disorders. In a search for small-molecule HIF-1 activators, 1936 marine invertebrate and algal extract samples (U.S. National Cancer Institute's Open Repository) were evaluated for HIF-1 activation activity in a cell-based reporter assay. Bioassay-guided fractionation of two active extracts of the sponge Dactylospongia elegans afforded four new sesquiterpene quinones (2-5), one new sesquiterpene phenol (6), the known Golgi disruptor ilimaquinone (1), and three previously reported ilimaquinone analogues (7-9). While antiproliferative activity was observed at higher concentrations, the sesquiterpene quinones (1-3) possessing a 2-hydroxy-5-methoxy-1,4-benzoquinone moiety activated HIF-1 and increased the expression of HIF-1 target gene vascular endothelial growth factor (VEGF) in T47D cells.


Asunto(s)
Factor 1 Inducible por Hipoxia/efectos de los fármacos , Poríferos/química , Quinonas/aislamiento & purificación , Quinonas/farmacología , Sesquiterpenos/aislamiento & purificación , Sesquiterpenos/farmacología , Animales , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Estructura Molecular , Resonancia Magnética Nuclear Biomolecular , Océanos y Mares , Quinonas/química , Sesquiterpenos/química , Factor A de Crecimiento Endotelial Vascular/efectos de los fármacos
15.
Toxicol Lett ; 219(2): 143-50, 2013 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-23528251

RESUMEN

Hypoxia inducible factor (HIF) and 5'-AMP-activated protein kinase are often activated under similar physiological conditions. Constitutive androstane receptor (CAR) translocates into the nucleus in accordance with 5'-AMP-activated protein kinase and thus confers transactivation. The aim of the present study was to investigate a possible link between CAR and HIFα. Phenobarbital (PB), a typical CAR activator, increased the gene expression of HIF-target genes in the livers of mice, including erythropoietin, heme oxygenase-1 and vascular endothelial growth factor-a. PB induced an accumulation of nuclear HIF-1α and an increase in the HIF-responsive element-mediated transactivation in HepG2 cells. Cobalt chloride, a typical HIF activator, induced the gene expression of CAR-target genes, including cyp2b9 and cyp2b10, an accumulation of nuclear CAR and an increase in the PB-responsive enhancer module-mediated transactivation in the mouse liver. Immunoprecipitation-immunoblot and chromatin immunoprecipitation analyses suggest that CAR binds to the PB-responsive enhancer module with HIF-1α in the liver of untreated mice and that the complex dissociates upon PB treatment. Taken together these results suggest that CAR and HIF-α interact and reciprocally modulate the functions of each other.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Factor 1 Inducible por Hipoxia/efectos de los fármacos , Receptor Cross-Talk/efectos de los fármacos , Receptores Citoplasmáticos y Nucleares/efectos de los fármacos , Animales , Western Blotting , Inmunoprecipitación de Cromatina , Cobalto/farmacología , Receptor de Androstano Constitutivo , Regulación de la Expresión Génica/genética , Genes Reporteros/efectos de los fármacos , Humanos , Factor 1 Inducible por Hipoxia/genética , Inmunoprecipitación , Masculino , Ratones , Ratones Endogámicos C3H , Fenobarbital/farmacología , Plásmidos/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Citoplasmáticos y Nucleares/genética
16.
Apoptosis ; 17(9): 938-49, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22684842

RESUMEN

Tumor hypoxia is considered the best validated target in clinical oncology because of its significant contribution to chemotherapy failure and drug resistance. As an approach to target hypoxia, we assessed the potential of quercetin, a flavonoid widely distributed in plants, as a anticancer agent under hypoxic conditions and examined its pharmacological mechanisms by primarily focusing on the role of AMP-activated protein kinase (AMPK). Quercetin significantly attenuated tumor growth in an HCT116 cancer xenograft in vivo model with a substantial reduction of AMPK activity. In a cell culture system, quercetin more dramatically induced apoptosis of HCT116 cancer cells under hypoxic conditions than normoxic conditions, and this was tightly associated with inhibition of hypoxia-induced AMPK activity. An in vitro kinase assay demonstrated that quercetin directly inhibits AMPK activity. Inhibition of AMPK by expressing a dominant-negative form resulted in an increase of apoptosis under hypoxia, and a constitutively active form of AMPK effectively blocked quercetin-induced apoptosis under hypoxia. Collectively, our data suggest that quercetin directly inhibits hypoxia-induced AMPK, which plays a protective role against hypoxia. Quercetin also reduced the activity of hypoxia-inducible factor-1 (HIF-1), a major transcription factor for adaptive cellular response to hypoxia. Moreover, quercetin sensitized HCT116 cancer cells to the anticancer drugs cisplatin and etoposide under hypoxic conditions. Our findings suggest that AMPK may serve as a novel target for overcoming tumor hypoxia-associated negative aspects.


Asunto(s)
Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Hipoxia de la Célula/efectos de los fármacos , Neoplasias del Colon/tratamiento farmacológico , Quercetina/farmacología , Animales , Antineoplásicos/farmacología , Antioxidantes/farmacología , Línea Celular Tumoral , Cisplatino/farmacología , Etopósido/farmacología , Genes Reporteros , Humanos , Factor 1 Inducible por Hipoxia/efectos de los fármacos , Factor 1 Inducible por Hipoxia/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Drug Chem Toxicol ; 35(1): 57-70, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22145808

RESUMEN

Berberine, a naturally occurring isoquinoline alkaloid, is present in a number of important medicinal plants. Berberine has a wide range of biochemical and pharmacological effects, including anticancer effects. In this study, we elucidated the mechanism of antiangiogenic activity of berberine using in vivo and in vitro models. In vivo antiangiogenic activity was studied using B16F-10 melanoma cells and induced capillary formation in C57BL/6 mice. Berberine, at 10 mg/kg body weight, showed significant inhibition in tumor-directed capillary formation and in various proangiogenic factors, such as vascular endothelial growth factor (VEGF), and proinflammatory mediators, such as interleukin (IL)-1ß, IL-6, tumor necrosis factor alpha (TNF-α), and granulocyte macrophage colony-stimulating factor (GM-CSF), which are involved in tumor angiogenesis. At the same time, it could also increase antitumor factors, such as IL-2 and tissue-inhibitor metalloproteinase (TIMP) levels in the serum. Berberine could also inhibit endothelial motility, migration, tube formation, and vessel sprouting from rat aortic ring in vitro. Further, berberine inhibited various transcription factors involved in tumor development and angiogenesis, such as NF-ĸB, c-Fos, CREB, and ATF-2. mRNA expression levels of proangiogenic factors, such as cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS), and hypoxia-inducible factor (HIF), were also downregulated in tumor cells after treatment with berberine. Drastically elevated expressions of HIF and VEGF mRNA by tumor cells under hypoxic conditions were also decreased after treatment with berberine. This result clearly demonstrates that the antiangiogenic activity of berberine is mainly mediated through the inhibition of various proinflammatory and pro-angiogenic factors and the major ones are HIF, VEGF, COX-2, NO, NF-ĸB, and proinflammatory cytokines.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Berberina/farmacología , Melanoma Experimental/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Animales , Línea Celular Tumoral , Regulación hacia Abajo/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Factor 1 Inducible por Hipoxia/efectos de los fármacos , Factor 1 Inducible por Hipoxia/metabolismo , Mediadores de Inflamación/metabolismo , Masculino , Melanoma Experimental/irrigación sanguínea , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Factor A de Crecimiento Endotelial Vascular/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/metabolismo
18.
J Pharmacol Exp Ther ; 340(1): 176-84, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22010210

RESUMEN

Wound and subsequent healing are frequently associated with hypoxia. Although hypoxia induces angiogenesis for tissue remodeling during wound healing, it may also affect the healing response of parenchymal cells. Whether and how wound healing is affected by hypoxia in kidney cells and tissues is currently unknown. Here, we used scratch-wound healing and transwell migration models to examine the effect of hypoxia in cultured renal proximal tubular cells (RPTC). Wound healing and migration were significantly slower in hypoxic (1% oxygen) RPTC than normoxic (21% oxygen) cells. Hypoxia-inducible factor-1α (HIF-1α) was induced during scratch-wound healing in normoxia, and the induction was more evident in hypoxia. Nevertheless, HIF-1α-null and wild-type cells healed similarly after scratch wounding. Moreover, activation of HIF-1α with dimethyloxalylglycine in normoxic cells did not suppress wound healing, negating a major role of HIF-1α in wound healing in this model. Scratch-wound healing was also associated with glycogen synthase kinase 3ß (GSK3ß)/ß-catenin signaling, which was further enhanced by hypoxia. Pharmacological inhibition of GSK3ß resulted in ß-catenin expression, accompanied by the suppression of wound healing and transwell cell migration. Ectopic expression of ß-catenin in normoxic cells could also suppress wound healing, mimicking the effect of hypoxia. Conversely, inhibition of ß-catenin via dominant negative mutants or short hairpin RNA improved wound healing and transwell migration in hypoxic cells. The results suggest that GSK3ß/ß-catenin signaling may contribute to defective wound healing in hypoxic renal cells and tissues.


Asunto(s)
Hipoxia de la Célula/fisiología , Glucógeno Sintasa Quinasa 3/fisiología , Factor 1 Inducible por Hipoxia/fisiología , Túbulos Renales Proximales/fisiología , Transducción de Señal/fisiología , Cicatrización de Heridas/fisiología , beta Catenina/fisiología , Animales , Animales Modificados Genéticamente , Western Blotting , Hipoxia de la Célula/efectos de los fármacos , Movimiento Celular , Células Cultivadas , Técnica del Anticuerpo Fluorescente , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3 beta , Factor 1 Inducible por Hipoxia/efectos de los fármacos , Factor 1 Inducible por Hipoxia/genética , Lentivirus/genética , Ratones , ARN/biosíntesis , ARN/genética , Ratas , Transfección , Cicatrización de Heridas/efectos de los fármacos , beta Catenina/biosíntesis , beta Catenina/genética
19.
J Nat Prod ; 74(2): 240-8, 2011 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-21214226

RESUMEN

In an effort to identify natural product-based molecular-targeted antitumor agents, mammea-type coumarins from the tropical/subtropical plant Mammea americana were found to inhibit the activation of HIF-1 (hypoxia-inducible factor-1) in human breast and prostate tumor cells. In addition to the recently reported mammea E/BB (15), bioassay-guided fractionation of the active extract yielded 14 mammea-type coumarins including three new compounds, mammea F/BB (1), mammea F/BA (2), and mammea C/AA (3). The absolute configuration of C-1' in 1 was determined by the modified Mosher's method on a methylated derivative. These coumarins were evaluated for their effects on mitochondrial respiration, HIF-1 signaling, and tumor cell proliferation/viability. Acetylation of 1 afforded a triacetoxylated product (A-2) that inhibited HIF-1 activation with increased potency in both T47D (IC(50) 0.83 µM for hypoxia-induced) and PC-3 cells (IC(50) 0.94 µM for hypoxia-induced). Coumarins possessing a 6-prenyl-8-(3-methyloxobutyl) substituent pattern exhibited enhanced HIF-1 inhibitory effects. The O-methylated derivatives were less active at inhibiting HIF-1 and suppressing cell proliferation/viability. Mechanistic studies indicate that these compounds act as anionic protonophores that potently uncouple mitochondrial electron transport and disrupt hypoxic signaling.


Asunto(s)
Antineoplásicos Fitogénicos , Respiración de la Célula/efectos de los fármacos , Cumarinas , Factor 1 Inducible por Hipoxia/efectos de los fármacos , Mammea/química , Algoritmos , Antineoplásicos Fitogénicos/síntesis química , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/aislamiento & purificación , Antineoplásicos Fitogénicos/farmacología , Cumarinas/síntesis química , Cumarinas/química , Cumarinas/aislamiento & purificación , Cumarinas/farmacología , Dominica , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Masculino , Estructura Molecular , Corteza de la Planta/química , Prenilación , Relación Estructura-Actividad
20.
J Toxicol Environ Health A ; 74(1): 43-51, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21120747

RESUMEN

The aim of this study was to examine the effects of pyocyanin exposure on mitochondrial GSH, other cellular thiols (thioredoxin-1, Trx-1), and oxidant-sensitive signaling pathways hypoxia inducible factor (HIF-1α) and heme oxygenase (HO-1) in A549 and HBE cell lines. A549 human type II alveolar epithelial cells and human bronchial epithelial (HBE) cells were treated with varying concentrations of pyocyanin extracted from Pseudomonas aeruginosa bacteria. Cytoplasmic and mitochondrial thiols and oxidant sensitive signal transduction proteins (HIF-1α and HO-1) were measured. Exposure to pyocyanin generated reactive oxygen species (ROS) in cellular mitochondria and altered total cellular glutathione (GSH). Pyocyanin, at concentrations present in conditions in vivo, increased oxidized Trx-1 in A549 human type II alveolar epithelial cells and HBE cells by 184 and 74%, respectively. Oxidized mitochondrial glutathione (GSSG) was elevated more than twofold in both cell types. Pyocyanin also increased the cellular oxidant-sensitive proteins HIF-1α and HO-1. Data indicate that pyocyanin-induced alterations in mitochondrial and cytosolic thiols, as well as oxidant-sensitive proteins, may contribute to P. aeruginosa-mediated lung injury.


Asunto(s)
Pulmón/efectos de los fármacos , Oxidación-Reducción/efectos de los fármacos , Pseudomonas aeruginosa/metabolismo , Piocianina/farmacología , Mucosa Respiratoria/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Línea Celular , Inducción Enzimática/efectos de los fármacos , Glutatión/efectos de los fármacos , Glutatión/metabolismo , Hemo Oxigenasa (Desciclizante)/biosíntesis , Hemo Oxigenasa (Desciclizante)/efectos de los fármacos , Hemo Oxigenasa (Desciclizante)/metabolismo , Humanos , Factor 1 Inducible por Hipoxia/efectos de los fármacos , Factor 1 Inducible por Hipoxia/metabolismo , Immunoblotting , Pulmón/metabolismo , Pulmón/microbiología , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Infecciones por Pseudomonas/metabolismo , Alveolos Pulmonares/efectos de los fármacos , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/microbiología , Piocianina/efectos adversos , Especies Reactivas de Oxígeno/metabolismo , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/microbiología , Tiorredoxinas/efectos de los fármacos , Tiorredoxinas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...