Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 22(6)2021 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-33805714

RESUMEN

Trifluoperazine (TFP), an antipsychotic drug approved by the Food and Drug Administration, has been show to exhibit anti-cancer effects. Pulmonary arterial hypertension (PAH) is a devastating disease characterized by a progressive obliteration of small pulmonary arteries (PAs) due to exaggerated proliferation and resistance to apoptosis of PA smooth muscle cells (PASMCs). However, the therapeutic potential of TFP for correcting the cancer-like phenotype of PAH-PASMCs and improving PAH in animal models remains unknown. PASMCs isolated from PAH patients were exposed to different concentrations of TFP before assessments of cell proliferation and apoptosis. The in vivo therapeutic potential of TFP was tested in two preclinical models with established PAH, namely the monocrotaline and sugen/hypoxia-induced rat models. Assessments of hemodynamics by right heart catheterization and histopathology were conducted. TFP showed strong anti-survival and anti-proliferative effects on cultured PAH-PASMCs. Exposure to TFP was associated with downregulation of AKT activity and nuclear translocation of forkhead box protein O3 (FOXO3). In both preclinical models, TFP significantly lowered the right ventricular systolic pressure and total pulmonary resistance and improved cardiac function. Consistently, TFP reduced the medial wall thickness of distal PAs. Overall, our data indicate that TFP could have beneficial effects in PAH and support the view that seeking new uses for old drugs may represent a fruitful approach.


Asunto(s)
Fármacos Cardiovasculares/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Hipertensión Pulmonar/tratamiento farmacológico , Hipoxia/prevención & control , Miocitos del Músculo Liso/efectos de los fármacos , Trifluoperazina/farmacología , Animales , Antipsicóticos/farmacología , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Reposicionamiento de Medicamentos , Femenino , Proteína Forkhead Box O3/genética , Proteína Forkhead Box O3/metabolismo , Hemodinámica/efectos de los fármacos , Humanos , Hipertensión Pulmonar/inducido químicamente , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/fisiopatología , Hipoxia/inducido químicamente , Hipoxia/genética , Hipoxia/fisiopatología , Indoles/administración & dosificación , Monocrotalina/administración & dosificación , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Cultivo Primario de Células , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Arteria Pulmonar/citología , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/metabolismo , Pirroles/administración & dosificación , Ratas , Ratas Sprague-Dawley , Survivin/genética , Survivin/metabolismo
2.
Int J Mol Sci ; 21(18)2020 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-32947968

RESUMEN

Pulmonary arterial hypertension (PAH) is a progressive disease which causes right ventricular (RV) failure. Canstatin, a C-terminal fragment of type IV collagen α2 chain, is expressed in various rat organs. However, the expression level of canstatin in plasma and organs during PAH is still unclear. We aimed to clarify it and further investigated the protective effects of canstatin in a rat model of monocrotaline-induced PAH. Cardiac functions were assessed by echocardiography. Expression levels of canstatin in plasma and organs were evaluated by enzyme-linked immunosorbent assay and Western blotting, respectively. PAH was evaluated by catheterization. RV remodeling was evaluated by histological analyses. Real-time polymerase chain reaction was performed to evaluate RV remodeling-related genes. The plasma concentration of canstatin in PAH rats was decreased, which was correlated with a reduction in acceleration time/ejection time ratio and an increase in RV weight/body weight ratio. The protein expression of canstatin in RV, lung and kidney was decreased in PAH rats. While recombinant canstatin had no effect on PAH, it significantly improved RV remodeling, including hypertrophy and fibrosis, and prevented the increase in RV remodeling-related genes. We demonstrated that plasma canstatin is decreased in PAH rats and that administration of canstatin exerts cardioprotective effects.


Asunto(s)
Cardiotónicos/uso terapéutico , Colágeno Tipo IV/biosíntesis , Colágeno Tipo IV/uso terapéutico , Hipertensión Pulmonar/metabolismo , Fragmentos de Péptidos/uso terapéutico , Remodelación Ventricular/efectos de los fármacos , Animales , Peso Corporal/efectos de los fármacos , Colágeno Tipo IV/sangre , Colágeno Tipo IV/genética , Evaluación Preclínica de Medicamentos , Ensayo de Inmunoadsorción Enzimática , Fibrosis , Ventrículos Cardíacos/efectos de los fármacos , Hipertensión Pulmonar/inducido químicamente , Hipertensión Pulmonar/genética , Hipertrofia , Riñón/metabolismo , Pulmón/metabolismo , Pulmón/patología , Masculino , Monocrotalina/toxicidad , Tamaño de los Órganos/efectos de los fármacos , Ratas , Ratas Wistar , Proteínas Recombinantes/uso terapéutico
3.
J Mol Med (Berl) ; 97(11): 1557-1566, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31529142

RESUMEN

The VARS2 gene encodes a mitochondrial valyl-transfer RNA synthetase which is used in mitochondrial translation. To date, several patients with VARS2 pathogenic variants have been described in the literature. These patients have features of lactic acidosis with encephalomyopathy. We present a case of an infant with lactic acidosis, failure to thrive, and severe primary pulmonary hypertension who was found to be a compound heterozygote for two novel VARS2 variants (c.1940C>T, p.(Thr647Met) and c.2318G>A, p.(Arg773Gln)). The patient was treated with vitamin supplements and a carbohydrate-restricted diet. The lactic acidosis and failure to thrive resolved, and he showed good growth and development. Functional studies and molecular analysis employed a yeast model system and the VAS1 gene (yeast homolog of VARS2). VAS1 genes harboring either one of two mutations corresponding to the two novel variants in the VARS2 gene, exhibited partially reduced function in haploid yeast strains. A combination of both VAS1 variant alleles in a diploid yeast cell exhibited a more significant decrease in oxidative metabolism-dependent growth and in the oxygen consumption rate (reminiscent of the patient who carries two mutant VARS2 alleles). Our results demonstrate the pathogenicity of the biallellic novel VARS2 variants. KEY MESSAGES: • A case of an infant who is a compound heterozygote for two novel VARS2 variants. • This infant displayed lactic acidosis, failure to thrive, and pulmonary hypertension. • Treatment of the patient with a carbohydrate-restricted diet resulted in good growth and development. • Studies with the homologous yeast VAS1 gene showed reduced function of corresponding single mutant in haploid yeast strains. • A combination of both VAS1 variant alleles in diploid yeast exhibited a more significant decrease in function, thereby confirming the pathogenicity of the biallellic novel VARS2 variants.


Asunto(s)
Insuficiencia de Crecimiento/genética , Insuficiencia de Crecimiento/metabolismo , Antígenos HLA/genética , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/metabolismo , Enfermedades Mitocondriales/genética , Enfermedades Mitocondriales/metabolismo , Valina-ARNt Ligasa/genética , Alelos , Secuencia de Aminoácidos , Heterocigoto , Humanos , Lactante , Masculino , Mutación/genética , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Análisis de Secuencia de ADN
4.
Circulation ; 139(19): 2238-2255, 2019 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-30759996

RESUMEN

BACKGROUND: Deficiencies of iron-sulfur (Fe-S) clusters, metal complexes that control redox state and mitochondrial metabolism, have been linked to pulmonary hypertension (PH), a deadly vascular disease with poorly defined molecular origins. BOLA3 (BolA Family Member 3) regulates Fe-S biogenesis, and mutations in BOLA3 result in multiple mitochondrial dysfunction syndrome, a fatal disorder associated with PH. The mechanistic role of BOLA3 in PH remains undefined. METHODS: In vitro assessment of BOLA3 regulation and gain- and loss-of-function assays were performed in human pulmonary artery endothelial cells using siRNA and lentiviral vectors expressing the mitochondrial isoform of BOLA3. Polymeric nanoparticle 7C1 was used for lung endothelium-specific delivery of BOLA3 siRNA oligonucleotides in mice. Overexpression of pulmonary vascular BOLA3 was performed by orotracheal transgene delivery of adeno-associated virus in mouse models of PH. RESULTS: In cultured hypoxic pulmonary artery endothelial cells, lung from human patients with Group 1 and 3 PH, and multiple rodent models of PH, endothelial BOLA3 expression was downregulated, which involved hypoxia inducible factor-2α-dependent transcriptional repression via histone deacetylase 1-mediated histone deacetylation. In vitro gain- and loss-of-function studies demonstrated that BOLA3 regulated Fe-S integrity, thus modulating lipoate-containing 2-oxoacid dehydrogenases with consequent control over glycolysis and mitochondrial respiration. In contexts of siRNA knockdown and naturally occurring human genetic mutation, cellular BOLA3 deficiency downregulated the glycine cleavage system protein H, thus bolstering intracellular glycine content. In the setting of these alterations of oxidative metabolism and glycine levels, BOLA3 deficiency increased endothelial proliferation, survival, and vasoconstriction while decreasing angiogenic potential. In vivo, pharmacological knockdown of endothelial BOLA3 and targeted overexpression of BOLA3 in mice demonstrated that BOLA3 deficiency promotes histological and hemodynamic manifestations of PH. Notably, the therapeutic effects of BOLA3 expression were reversed by exogenous glycine supplementation. CONCLUSIONS: BOLA3 acts as a crucial lynchpin connecting Fe-S-dependent oxidative respiration and glycine homeostasis with endothelial metabolic reprogramming critical to PH pathogenesis. These results provide a molecular explanation for the clinical associations linking PH with hyperglycinemic syndromes and mitochondrial disorders. These findings also identify novel metabolic targets, including those involved in epigenetics, Fe-S biogenesis, and glycine biology, for diagnostic and therapeutic development.


Asunto(s)
Endotelio Vascular/fisiología , Glicina/metabolismo , Hipertensión Pulmonar/genética , Proteínas Mitocondriales/metabolismo , Adolescente , Adulto , Animales , Respiración de la Célula , Células Cultivadas , Niño , Preescolar , Modelos Animales de Enfermedad , Femenino , Humanos , Hipertensión Pulmonar/metabolismo , Lactante , Proteínas Hierro-Azufre/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Mitocondriales/genética , Mutación/genética , Oxidación-Reducción , ARN Interferente Pequeño/genética , Adulto Joven
5.
J Cell Physiol ; 233(12): 9750-9762, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30078229

RESUMEN

Excessive proliferation, migration, and antiapoptosis of pulmonary artery (PA) smooth muscle cells (PASMCs) underlies the development of pulmonary vascular remodeling. The innervation of the PA is predominantly sympathetic, and increased levels of circulating catecholamines have been detected in pulmonary arterial hypertension (PAH), suggesting that neurotransmitters released by sympathetic overactivation may play an essential role in PAH. However, the responsible mechanism remains unclear. Here, to investigate the effects of norepinephrine (NE) on PASMCs and the related mechanism, we used 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide, the proliferating cell nuclear antigen and the cell counting kit-8 assay to evaluate the proliferation of PASMCs, Boyden chamber migration, and wound-healing assays to assess migration and western blot analysis to investigate protein expression. We demonstrated that the phosphorylation level of the protein phosphatase 2A (PP2A) catalytic subunit (Y307) was higher in PAH patients and PAH models than in controls, both in vivo and in vitro. In addition, NE induced the proliferation and migration of PASMCs, which was attenuated by berberine (BBR), a Chinese herbal medicine, and/or PP2A overexpression. PP2A inhibition worsened NE-induced PAH and could not be reversed by BBR. Thus, PP2A is critical in driving PAH, and BBR may alleviate PAH via PP2A signaling pathways, thereby offering a potential therapeutic option for PAH.


Asunto(s)
Berberina/administración & dosificación , Hipertensión Pulmonar/tratamiento farmacológico , Proteína Fosfatasa 2/genética , Arteria Pulmonar/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Hipertensión Pulmonar/inducido químicamente , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/patología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/patología , Norepinefrina/toxicidad , Arteria Pulmonar/patología , Ratas , Transducción de Señal/efectos de los fármacos , Remodelación Vascular/efectos de los fármacos , Remodelación Vascular/genética
6.
Hum Mutat ; 38(6): 692-703, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28247525

RESUMEN

COX5A is a nuclear-encoded subunit of mitochondrial respiratory chain complex IV (cytochrome c oxidase). We present patients with a homozygous pathogenic variant in the COX5A gene. Clinical details of two affected siblings suffering from early-onset pulmonary arterial hypertension, lactic acidemia, failure to thrive, and isolated complex IV deficiency are presented. We show that the variant lies within the evolutionarily conserved COX5A/COX4 interface domain, suggesting that it alters the interaction between these two subunits during complex IV biogenesis. In patient skin fibroblasts, the enzymatic activity and protein levels of complex IV and several of its subunits are reduced. Lentiviral complementation rescues complex IV deficiency. The monomeric COX1 assembly intermediate accumulates demonstrating a function of COX5A in complex IV biogenesis. A potential therapeutic lead is demonstrated by showing that copper supplementation leads to partial rescue of complex IV deficiency in patient fibroblasts.


Asunto(s)
Acidosis Láctica/genética , Ciclooxigenasa 1/genética , Grupo Citocromo c/genética , Insuficiencia de Crecimiento/genética , Hipertensión Pulmonar/genética , Acidosis Láctica/patología , Núcleo Celular/genética , Ciclooxigenasa 1/química , Grupo Citocromo c/química , Deficiencia de Citocromo-c Oxidasa , Complejo IV de Transporte de Electrones , Insuficiencia de Crecimiento/patología , Fibroblastos , Predisposición Genética a la Enfermedad , Homocigoto , Humanos , Hipertensión Pulmonar/patología , Mitocondrias/genética , Mutación , Subunidades de Proteína/genética
7.
Circulation ; 135(5): 460-475, 2017 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-27881557

RESUMEN

BACKGROUND: Pulmonary arterial hypertension (PAH) is a heterogeneous disorder with high mortality. METHODS: We conducted a comprehensive study of plasma metabolites using ultraperformance liquid chromatography mass spectrometry to identify patients at high risk of early death, to identify patients who respond well to treatment, and to provide novel molecular insights into disease pathogenesis. RESULTS: Fifty-three circulating metabolites distinguished well-phenotyped patients with idiopathic or heritable PAH (n=365) from healthy control subjects (n=121) after correction for multiple testing (P<7.3e-5) and confounding factors, including drug therapy, and renal and hepatic impairment. A subset of 20 of 53 metabolites also discriminated patients with PAH from disease control subjects (symptomatic patients without pulmonary hypertension, n=139). Sixty-two metabolites were prognostic in PAH, with 36 of 62 independent of established prognostic markers. Increased levels of tRNA-specific modified nucleosides (N2,N2-dimethylguanosine, N1-methylinosine), tricarboxylic acid cycle intermediates (malate, fumarate), glutamate, fatty acid acylcarnitines, tryptophan, and polyamine metabolites and decreased levels of steroids, sphingomyelins, and phosphatidylcholines distinguished patients from control subjects. The largest differences correlated with increased risk of death, and correction of several metabolites over time was associated with a better outcome. Patients who responded to calcium channel blocker therapy had metabolic profiles similar to those of healthy control subjects. CONCLUSIONS: Metabolic profiles in PAH are strongly related to survival and should be considered part of the deep phenotypic characterization of this disease. Our results support the investigation of targeted therapeutic strategies that seek to address the alterations in translational regulation and energy metabolism that characterize these patients.


Asunto(s)
Hipertensión Pulmonar/genética , Metabolómica/métodos , ARN de Transferencia/metabolismo , Adulto , Anciano , Metabolismo Energético , Femenino , Humanos , Hipertensión Pulmonar/metabolismo , Masculino , Persona de Mediana Edad , Pronóstico , Resultado del Tratamiento , Adulto Joven
8.
Zhongguo Zhong Yao Za Zhi ; 40(7): 1355-61, 2015 Apr.
Artículo en Chino | MEDLINE | ID: mdl-26281561

RESUMEN

OBJECTIVE: To observe the effect of sesamin (Ses) on pulmonary vascular remodeling in rats with monocrotaline ( MCT)-induced pulmonary hypertension (PH). METHOD: Totally 48 male Sprague-Dawley (SD) rats were fed adaptively for one week and then divided into the normal control group, the MCT group, the MCT +Ses (50 mg x kg(-1)) group and the MCT + Ses (100 mg x kg(-1)) group, with 12 rats in each group. The PH rat model was induced through the subcutaneous injection with MCT(60 mg x kg(-1)). After the administration for four weeks, efforts were made to measure the right ventricular systolic pressure( RVSP) and mean pulmonary artery pressure (mPAP) through right jugular vein catheterization, and isolate right ventricle( RV) and left ventricle( LV) +septum (S) and measure their length to calculate RV/ ( LV + S) and ratio of RV to tibial length. Pathologic changes in arterioles were observed by HE staining. Masson's trichrome stain was used to demonstrate changes in collagen deposition of arterioles. The alpha-smooth muscle actin (alpha-SMA) expression in pulmonary arteries was measured by immunohistochemisty. The total antioxidative capacity (T-AOC) and malondialdehyde (MDA) content in pulmonary arteries were determined by the colorimetric method. The protein expressions of collagen I, NOX2 and NOX4 were analyzed by Real-time PCR and Western blot. RESULT: After the administration for 4 weeks, Ses could attenuate RVSP and mPAP induced by MCT, RV/ (LV + S) and ratio of RV to Tibial length, alpha-SMA and collagen I expressions and remodeling of pulmonary vessels and right ventricle. Meanwhile, Ses could obviously inhibit the expressions of NOX2, NOX4 and MDA content and increase T-AOC. CONCLUSION: Sesamin could ameliorate pulmonary vascular remodeling induced by monocrotaline in PH rats. Its mechanism may be related to expressions of NOX2 and NOX4 expression and reduction in oxidative stress injury.


Asunto(s)
Dioxoles/administración & dosificación , Medicamentos Herbarios Chinos/administración & dosificación , Hipertensión Pulmonar/tratamiento farmacológico , Lignanos/administración & dosificación , Remodelación Vascular/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Humanos , Hipertensión Pulmonar/enzimología , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/fisiopatología , Pulmón/irrigación sanguínea , Pulmón/enzimología , Pulmón/metabolismo , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Monocrotalina/efectos adversos , NADPH Oxidasa 2 , NADPH Oxidasa 4 , NADPH Oxidasas/genética , NADPH Oxidasas/metabolismo , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/metabolismo , Arteria Pulmonar/fisiopatología , Ratas , Ratas Sprague-Dawley
9.
Am J Respir Cell Mol Biol ; 48(1): 125-34, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23065131

RESUMEN

Danshen, the dried root of Salvia miltiorrhiza, is widely used in clinics in China for treating various diseases, including cardiovascular diseases. Sodium tanshinone IIA sulfonate (STS), a water-soluble derivative of tanshinone IIA isolated as the major active component from Danshen, was recently reported to be effective in attenuating the characteristic pulmonary vascular changes associated with chronically hypoxic pulmonary hypertension (CHPH); however, the underlying detailed mechanisms are poorly understood. In this study, we investigated the effects of STS on basal intracellular Ca(2+) concentration ([Ca(2+)](i)) and store-operated Ca(2+) entry (SOCE) in distal pulmonary arterial smooth muscle cells (PASMCs) exposed to prolonged hypoxia or isolated from CHPH rats. SOCE measured by Mn(2+) quenching of Fura-2 fluorescence in PASMCs from rats exposed to chronic hypoxia (10% O(2), 21 d) was increased by 59%, and basal [Ca(2+)](i) was increased by 119%; this effect was inhibited by intraperitoneal injection of STS. These inhibitory effects of STS on hypoxic increases of SOCE and basal [Ca(2+)](i) were associated with reduced expression of canonical transient receptor potential (TRPC)1 and TRPC6 in distal pulmonary arterial smooth muscle and decreases on right ventricular pressure, right ventricular hypertrophy, and peripheral pulmonary vessel thickening. In ex vivo cultured distal PASMCs from normoxic rats, STS (0-25 µM) dose-dependently inhibited hypoxia-induced cell proliferation and migration, paralleled with attenuation in increases of basal [Ca(2+)](i), SOCE, mRNA, and protein expression of TRPC1 and TRPC6. STS also relieved right ventricular systolic pressure, right ventricular hypertrophy, and TRPC1 and TRPC6 protein expression in distal pulmonary arteries in a monocrotaline-induced rat model of pulmonary arterial hypertension. These results indicate that STS prevents pulmonary arterial hypertension development likely by inhibiting TRPC1 and TRPC6 expression, resulting in normalized basal [Ca(2+)](i) and attenuated proliferation and migration of PASMCs.


Asunto(s)
Hipertensión Pulmonar/tratamiento farmacológico , Fenantrenos/farmacología , Fitoterapia , Canales de Potencial de Receptor Transitorio/antagonistas & inhibidores , Animales , Calcio/metabolismo , Canales de Calcio/metabolismo , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Medicamentos Herbarios Chinos/farmacología , Hemodinámica/efectos de los fármacos , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/patología , Hipertensión Pulmonar/fisiopatología , Hipoxia/tratamiento farmacológico , Hipoxia/genética , Hipoxia/patología , Hipoxia/fisiopatología , Masculino , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/patología , Músculo Liso Vascular/fisiopatología , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/patología , Arteria Pulmonar/fisiopatología , Ratas , Ratas Sprague-Dawley , Salvia miltiorrhiza , Canales Catiónicos TRPC/antagonistas & inhibidores , Canales Catiónicos TRPC/genética , Canales Catiónicos TRPC/metabolismo , Canales de Potencial de Receptor Transitorio/genética , Canales de Potencial de Receptor Transitorio/metabolismo
10.
Am J Cardiol ; 105(10): 1485-9, 2010 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-20451700

RESUMEN

Before 2001, intravenous epoprostenol was the only approved drug for patients with idiopathic pulmonary arterial hypertension (IPAH) or familial pulmonary arterial hypertension (FPAH) who were nonresponsive to high-dose calcium channel blockade. The investigators report transitioning select pediatric patients with IPAH or FPAH from intravenous epoprostenol to oral and/or inhaled agents for pulmonary arterial hypertension. A retrospective review was performed on all pediatric patients with IPAH or FPAH treated at Columbia University (1987 to 2008) who transitioned off epoprostenol to oral or inhaled drugs. Criteria for transition included functional class I or II, mean pulmonary arterial pressure <35 mm Hg, normal cardiac index, and age >6 years. Hemodynamic and clinical data were obtained at baseline (before epoprostenol), at peak epoprostenol dose, and after epoprostenol discontinuation. Fourteen of 104 pediatric patients with IPAH or FPAH transitioned off epoprostenol to oral or inhaled drugs from April 2003 to July 2008. Of the 14 subjects, 13 transitioned off epoprostenol successfully to oral or inhaled drugs. No significant changes in functional class, hemodynamics, or exercise data were seen after epoprostenol (mean follow-up duration 7.0 +/- 5.8 months) compared to peak epoprostenol, but further improvement was reported in World Health Organization functional class (p <0.005) after epoprostenol discontinuation. After successful epoprostenol discontinuation, 77% of patients were treated with endothelin receptor antagonists, 69% with phosphodiesterase-5 inhibitors, 38% with calcium channel blockers, and 8% inhaled iloprost. At the cut-off date (May 2009), there was 100% survival and 93% transition success. In conclusion, in carefully selected children with IPAH or FPAH initiated on intravenous epoprostenol before the availability of nonparenteral therapy, transition to oral or inhaled therapy for pulmonary arterial hypertension appears safe, with efficacy maintained when performed with close follow-up at a pulmonary hypertension specialty center.


Asunto(s)
Antihipertensivos/administración & dosificación , Epoprostenol/administración & dosificación , Hipertensión Pulmonar/diagnóstico , Hipertensión Pulmonar/tratamiento farmacológico , Administración por Inhalación , Administración Oral , Adolescente , Niño , Preescolar , Estudios de Cohortes , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Femenino , Estudios de Seguimiento , Humanos , Hipertensión Pulmonar/genética , Lactante , Inyecciones Intravenosas , Masculino , Probabilidad , Estudios Retrospectivos , Medición de Riesgo , Índice de Severidad de la Enfermedad , Resultado del Tratamiento
11.
Adv Exp Med Biol ; 661: 419-34, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20204746

RESUMEN

Pulmonary hypertension (PH) is a disorder characterized by vascular remodeling and proliferation, a phenotype dependent upon unimpeded growth factor and kinase pathway activation with strong similarities to malignant tumors. This chapter details our novel application of the multikinase inhibitor, sorafenib, in rodent models of PH to improved hemodynamic parameters and attenuates PH structural changes1. Sorafenib is a Raf kinase inhibitor and our biochemical and genomic evidence supported the potential involvement of the MAPK cascade system and TGFB3 in PH development and the response to therapy. Integration of expression genomic analyses coupled with intense bioinformatics identified gene expression and ontology signatures in the development of PH and implicated the role of cytoskeletal protein such as caldesmon or nmMLCK as potentially key participants in PH-induced vascular remodeling and proliferation. Our studies suggest the PKI sorafenib as a potentially novel treatment for severe PH with the MAPK cascade a potential canonical target profoundly effecting vascular cytoskeletal -rearrangements and remodeling1.


Asunto(s)
Bencenosulfonatos/uso terapéutico , Hipertensión Pulmonar/tratamiento farmacológico , Hipertensión Pulmonar/fisiopatología , Inhibidores de Proteínas Quinasas/uso terapéutico , Piridinas/uso terapéutico , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Animales , Bencenosulfonatos/farmacología , Proteínas de Unión a Calmodulina/genética , Proteínas de Unión a Calmodulina/metabolismo , Expresión Génica/efectos de los fármacos , Perfilación de la Expresión Génica , Hemodinámica/efectos de los fármacos , Humanos , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/patología , Pulmón/irrigación sanguínea , Pulmón/patología , Pulmón/fisiopatología , Análisis por Micromatrices , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Niacinamida/análogos & derivados , Compuestos de Fenilurea , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Transducción de Señal/efectos de los fármacos , Sorafenib
12.
Endocrinology ; 149(9): 4367-73, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18511508

RESUMEN

The pathogenesis of pulmonary hypertension (PH) includes an inflammatory response. Thymulin, a zinc-dependent thymic hormone, has important immunobiological effects by inhibiting various proinflammatory cytokines and chemokines. We investigated morphological and hemodynamic effects of thymulin administration in a rat model of monocrotaline (MCT)-induced PH, as well as the pattern of proinflammatory cytokine gene expression and the intracellular pathways involved. Adult Wistar rats received an injection of MCT (60 mg/kg, sc) or an equal volume of saline. One day after, the animals randomly received during 3 wk an injection of saline, vehicle (zinc plus carboxymethyl cellulose), or thymulin (100 ng/kg, sc, daily). At d 23-25, the animals were anesthetized for hemodynamic recordings, whereas heart and lungs were collected for morphometric and molecular analysis. Thymulin prevented morphological, hemodynamic, and inflammatory cardiopulmonary profile characteristic of MCT-induced PH, whereas part of these effects were also observed in MCT-treated animals injected with the thymulin's vehicle containing zinc. The pulmonary thymulin effect was likely mediated through suppression of p38 pathway.


Asunto(s)
Hipertensión Pulmonar/inducido químicamente , Hipertensión Pulmonar/prevención & control , Interleucina-6/genética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Monocrotalina , Factor Tímico Circulante/farmacología , Animales , Citocinas/genética , Citocinas/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Corazón/anatomía & histología , Corazón/efectos de los fármacos , Hemodinámica/efectos de los fármacos , Hipertensión Pulmonar/genética , Mediadores de Inflamación/metabolismo , Interleucina-6/metabolismo , Pulmón/anatomía & histología , Pulmón/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/genética , Masculino , Ratas , Ratas Wistar , Factor Tímico Circulante/uso terapéutico , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología
13.
J Pharmacol Sci ; 107(1): 8-14, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18490853

RESUMEN

The most important strategies in pharmacogenomics are gene expression profiling and the network analysis of human disease models. We have previously discovered novel drug target candidates in cardiovascular diseases through investigations of these pharmacogenomics. The significant induction of S100C mRNA and protein expression was detected in the rat pulmonary hypertension and myocardial infarction model. We also found increased taurine in hypoxia, a calcium-associated cytoprotective compound, to suppress the hypoxia-induced S100C gene expression and vascular remodeling. These results suggest that S100C may be one of the potential novel drug targets in hypoxic or ischemic diseases. Delayed cerebral vasospasm after aneurysmal subarachnoid hemorrhage causes cerebral ischemia and infarction. Using a DNA microarray, a prominant upregulation of heme oxygenase-1 (HO-1) and heat shock protein (HSP) 72 mRNAs were observed in the basilar artery of a murine vasospasm model. Antisense HO-1 and HSP 72 oligodeoxynucleotide inhibited HO-1 and HSP 72 induction, respectively, and significantly aggravated cerebral vasospasm. Moreover, we have also developed a unique heart failure model in zebrafish and identified several candidate genes as novel drug targets. These results suggest that pharmacogenomic network analysis has the potential to bridge the gap between in vitro and in vivo studies and could define strategies for identifying novel drug targets in various cardiovascular diseases.


Asunto(s)
Fármacos Cardiovasculares/farmacología , Enfermedades Cardiovasculares/terapia , Perfilación de la Expresión Génica , Redes Reguladoras de Genes/efectos de los fármacos , Terapia Genética , Farmacogenética , Animales , Fármacos Cardiovasculares/uso terapéutico , Enfermedades Cardiovasculares/tratamiento farmacológico , Enfermedades Cardiovasculares/genética , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Proteínas del Choque Térmico HSP72/genética , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/terapia , Hemo-Oxigenasa 1/genética , Humanos , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/terapia , Ratas , Proteínas S100/genética , Vasoespasmo Intracraneal/genética , Vasoespasmo Intracraneal/terapia , Pez Cebra/genética
14.
Endocrinology ; 149(1): 237-44, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17916633

RESUMEN

Chronic exposure to hypoxia, a common adverse consequence of most pulmonary disorders, can lead to a sustained increase in pulmonary arterial pressure (PAP), right ventricular hypertrophy, and is, therefore, closely associated with heart failure and increased mortality. Ghrelin, originally identified as an endogenous GH secretagogue, has recently been shown to possess potent vasodilator properties, likely involving modulation of the vascular endothelium and its associated vasoactive peptides. In this study we hypothesized that ghrelin would impede the pathogenesis of pulmonary arterial hypertension during chronic hypoxia (CH). PAP was continuously measured using radiotelemetry, in conscious male Sprague Dawley rats, in normoxia and during 2-wk CH (10% O(2)). During this hypoxic period, rats received a daily sc injection of either saline or ghrelin (150 microg/kg). Subsequently, heart and lung samples were collected for morphological, histological, and molecular analyses. CH significantly elevated PAP in saline-treated rats, increased wall thickness of peripheral pulmonary arteries, and, consequently, induced right ventricular hypertrophy. In these rats, CH also led to the overexpression of endothelial nitric oxide synthase mRNA and protein, as well as endothelin-1 mRNA within the lung. Exogenous ghrelin administration attenuated the CH-induced overexpression of endothelial nitric oxide synthase mRNA and protein, as well as endothelin-1 mRNA. Consequently, ghrelin significantly attenuated the development of pulmonary arterial hypertension, pulmonary vascular remodeling, and right ventricular hypertrophy. These results demonstrate the therapeutic benefits of ghrelin for impeding the pathogenesis of pulmonary hypertension and right ventricular hypertrophy, particularly in subjects prone to CH (e.g. pulmonary disorders).


Asunto(s)
Ghrelina/uso terapéutico , Hipertensión Pulmonar/tratamiento farmacológico , Hipertensión Pulmonar/etiología , Hipertensión Pulmonar/patología , Hipoxia/complicaciones , Hipoxia/patología , Algoritmos , Animales , Glucemia/análisis , Peso Corporal/efectos de los fármacos , Enfermedad Crónica , Estado de Conciencia , Progresión de la Enfermedad , Evaluación Preclínica de Medicamentos , Endotelina-1/genética , Endotelina-1/metabolismo , Ácidos Grasos no Esterificados/sangre , Ghrelina/farmacología , Hemodinámica/efectos de los fármacos , Hipertensión Pulmonar/genética , Hipoxia/genética , Masculino , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley
15.
Am J Pathol ; 167(6): 1497-509, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16314465

RESUMEN

Immediately after birth the adluminal vascular SMCs of the pulmonary elastic arteries undergo transient actin cytoskeletal remodeling as well as cellular de-differentiation and proliferation. Vascular smooth muscle phenotype is regulated by serum response factor, which is itself regulated in part by the negative regulator YY1. We therefore studied the subcellular localization of YY1 in arteries of normal newborn piglets and piglets affected by neonatal pulmonary hypertension. We found that YY1 localization changed during development and that expression of gamma-smooth muscle actin correlated with expression of cytoplasmic rather than nuclear YY1. Analysis of the regulation of YY1 localization in vitro demonstrated that polymerized gamma-actin sequestered EGFP-YY1 in the cytoplasm and that YY1 activation of c-myc promoter activity was inhibited by LIM kinase, which increases actin polymerization. Consistent with these data siRNA-mediated down-regulation of YY1 in C2C12 cells increased SM22-alpha expression and inhibited cell proliferation. Thus, actin polymerization controls subcellular YY1 localization, which contributes to vascular SMC proliferation and differentiation in normal pulmonary artery development. In the absence of actin depolymerization, YY1 does not relocate to the nucleus, and this lack of relocation may contribute to the pathobiology of pulmonary hypertension.


Asunto(s)
Regulación de la Expresión Génica , Hipertensión Pulmonar/genética , Músculo Liso Vascular/fisiología , Factor de Transcripción YY1/fisiología , Actinas/genética , Animales , Animales Recién Nacidos , Secuencia de Bases , Línea Celular , Clonación Molecular , Cartilla de ADN , Genes myc , Mutación , Regiones Promotoras Genéticas , ARN Interferente Pequeño/genética , Porcinos , Transfección
16.
Physiol Genomics ; 21(3): 314-23, 2005 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-15728335

RESUMEN

Myocardial right ventricular (RV) hypertrophy due to pulmonary hypertension is aimed at normalizing ventricular wall stress. Depending on the degree of pressure overload, RV hypertrophy may progress to a state of impaired contractile function and heart failure, but this cannot be discerned during the early stages of ventricular remodeling. We tested whether critical differences in gene expression profiles exist between ventricles before the ultimate development of either a compensated or decompensated hypertrophic phenotype. Both phenotypes were selectively induced in Wistar rats by a single subcutaneous injection of either a low or a high dose of the pyrrolizidine alkaloid monocrotaline (MCT). Spotted oligonucleotide microarrays were used to investigate pressure-dependent cardiac gene expression profiles at 2 wk after the MCT injections, between control rats and rats that would ultimately develop either compensated or decompensated hypertrophy. Clustering of significantly regulated genes revealed specific expression profiles for each group, although the degree of hypertrophy was still similar in both. The ventricles destined to progress to failure showed activation of pro-apoptotic pathways, particularly related to mitochondria, whereas the group developing compensated hypertrophy showed blocked pro-death effector signaling via p38-MAPK, through upregulation of MAPK phosphatase-1. In summary, we show that, already at an early time point, pivotal differences in gene expression exist between ventricles that will ultimately develop either a compensated or a decompensated phenotype, depending on the degree of pressure overload. These data reveal genes that may provide markers for the early prediction of clinical outcome as well as potential targets for early intervention.


Asunto(s)
Insuficiencia Cardíaca/genética , Hipertrofia Ventricular Izquierda/genética , Hipertrofia Ventricular Derecha/genética , ARN Mensajero/genética , Animales , Factor Natriurético Atrial/genética , ATPasas Transportadoras de Calcio/genética , Cartilla de ADN , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Hipertensión/genética , Hipertensión Pulmonar/etiología , Hipertensión Pulmonar/genética , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa , ARN/genética , ARN/aislamiento & purificación , Ratas , Ratas Wistar , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico
17.
Heart Fail Rev ; 8(1): 5-21, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12652155

RESUMEN

Pulmonary Arterial Hypertension (PAH) is a disease of the pulmonary vasculature leading to vasoconstriction and remodeling of the pulmonary arteries. The resulting increase in the right ventricular afterload leads to right ventricular failure and death. The treatment options are limited, expensive and associated with significant side effects. The nitric oxide (NO) pathway in the pulmonary circulation provides several targets for the development of new therapies for this disease. However, the NO pathway is modulated at multiple levels including transcription and expression of the NO synthase gene, regulation of the NO synthase activity, regulation of the production of cyclic guanomonophosphate (cGMP) by phosphodiesterases, postsynthetic oxidation of NO, etc. This makes the study of the role of the NO pathway very difficult, unless one uses multiple complementary techniques. Furthermore, there are significant differences between the pulmonary and the systemic circulation which make extrapolation of data from one circulation to the other very difficult. In addition, the role of NO in the development of pulmonary hypertension varies among different models of the disease. This paper reviews the role of the NO pathway in both the healthy and diseased pulmonary circulation and in several animal models and human forms of the disease. It focuses on the role of recent therapies that target the NO pathway, including L-Arginine, inhaled NO, the phosphodiesterase inhibitor sildenafil and gene therapy.


Asunto(s)
Hipertensión Pulmonar/tratamiento farmacológico , Hipertensión Pulmonar/fisiopatología , Óxido Nítrico/fisiología , Óxido Nítrico/uso terapéutico , Arteria Pulmonar/fisiopatología , Animales , Humanos , Hipertensión Pulmonar/genética , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa/metabolismo , Arteria Pulmonar/metabolismo , Circulación Pulmonar/fisiología , Transcripción Genética/genética , Resistencia Vascular/fisiología
18.
Zhonghua Yi Xue Za Zhi ; 79(9): 654-6, 1999 Sep.
Artículo en Chino | MEDLINE | ID: mdl-11715451

RESUMEN

OBJECTIVE: To investigate the collagen expression effect of intra-acinar pulmonary arteries and the right ventricle and to observe the intervention effect of Radix Astragali in rats with hypoxic pulmonary hypertension. METHODS: Wistar male rats were divided into three groups: hypoxia group, which was fed in hypoxic environment (oxygen concentration was 9%-12%) under normal atmospheric pressure for three weeks (12 hours every day); intervention group, which was abdomenly injected with Radix Astragali and fed in the same environment as hypoxia group; and control group, which was fed in normal environment. Pulmonary artery pressure and cardiac output of the right ventricle were measured with catheterization and Fick's method. Protein expression of collagen in intra-acinar pulmonary arteries was assessed with histopathologic method. mRNA expressions of collagen in the right ventricle, and endothelin-1 and nitric oxide synthase in lung tissue were evaluated by RT-PCR technique. RESULTS: Compared with those in control group, pulmonary artery pressure and protein expression of collagen in intra-acinar pulmonary arteries and mRNA expression of type I collagen in the right ventricle and endothelin-1 in lung tissue were obviously increased in hypoxia group (mean pulmonary artery pressure: (24.8 +/- 3.6) mm Hg vs (18.8 +/- 1.7) mm Hg, P < 0.01; protein expression of collagen: (20.4 +/- 4.5) AU vs (3.4 +/- 1.1) AU (P < 0.01), but mRNA expression of nitric oxide synthase in lung tissue was declined. Compared with the data obtained in hypoxia group, cardiac output and mRNA expression of nitric oxide synthase in lung tissue were raised in intervention group (cardiac output: [(141 +/- 20) ml/min vs (118 +/- 40) ml/min, P < 0.01], but protein expression of collagen in intra-acinar pulmonary arteries and mRNA expression of type I collagen in the right ventricle and endothelin-1 in lung tissue were markedly decreased (protein expression of collagen: [(5.6 +/- 1.2) AU vs (20.4 +/- 4.5) AU, P < 0.001]. CONCLUSION: Changes of collagen expression in intra-acinar pulmonary arteries and right ventricle are one of the most important features in remodeling of pulmonary arteries and right ventricle in hypoxic pulmonary hypertension. Radix Astragali can partly inhibit the remodeling of pulmonary arteries and right ventricle, and regulate mRNA expression of endothelin-1, nitric oxide synthase, and collagen, thus contributing to the treatment of hypoxic pulmonary hypertension.


Asunto(s)
Colágeno/biosíntesis , Medicamentos Herbarios Chinos/farmacología , Ventrículos Cardíacos/metabolismo , Hipertensión Pulmonar/genética , Arteria Pulmonar/metabolismo , Animales , Astragalus propinquus , Colágeno/genética , Hipertensión Pulmonar/tratamiento farmacológico , Hipoxia/complicaciones , Masculino , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo III , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Distribución Aleatoria , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA