Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Am J Physiol Lung Cell Mol Physiol ; 318(4): L773-L786, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-32159369

RESUMEN

Metabolic reprogramming is considered important in the pathogenesis of the occlusive vasculopathy observed in pulmonary hypertension (PH). However, the mechanisms that link reprogrammed metabolism to aberrant expression of genes, which modulate functional phenotypes of cells in PH, remain enigmatic. Herein, we demonstrate that, in mice, hypoxia-induced PH was prevented by glucose-6-phosphate dehydrogenase deficiency (G6PDDef), and further show that established severe PH in Cyp2c44-/- mice was attenuated by knockdown with G6PD shRNA or by G6PD inhibition with an inhibitor (N-ethyl-N'-[(3ß,5α)-17-oxoandrostan-3-yl]urea, NEOU). Mechanistically, G6PDDef, knockdown and inhibition in lungs: 1) reduced hypoxia-induced changes in cytoplasmic and mitochondrial metabolism, 2) increased expression of Tet methylcytosine dioxygenase 2 (Tet2) gene, and 3) upregulated expression of the coding genes and long noncoding (lnc) RNA Pint, which inhibits cell growth, by hypomethylating the promoter flanking region downstream of the transcription start site. These results suggest functional TET2 is required for G6PD inhibition to increase gene expression and to reverse hypoxia-induced PH in mice. Furthermore, the inhibitor of G6PD activity (NEOU) decreased metabolic reprogramming, upregulated TET2 and lncPINT, and inhibited growth of control and diseased smooth muscle cells isolated from pulmonary arteries of normal individuals and idiopathic-PAH patients, respectively. Collectively, these findings demonstrate a previously unrecognized function for G6PD as a regulator of DNA methylation. These findings further suggest that G6PD acts as a link between reprogrammed metabolism and aberrant gene regulation and plays a crucial role in regulating the phenotype of cells implicated in the pathogenesis of PH, a debilitating disorder with a high mortality rate.


Asunto(s)
Metilación de ADN/genética , Glucosafosfato Deshidrogenasa/genética , Hipertensión Pulmonar/genética , Hipoxia/genética , Animales , Proliferación Celular/genética , Familia 2 del Citocromo P450/genética , Femenino , Expresión Génica/genética , Regulación de la Expresión Génica/genética , Pulmón/metabolismo , Masculino , Ratones , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Fenotipo , Arteria Pulmonar/metabolismo , ARN Largo no Codificante/genética , Regulación hacia Arriba/genética
2.
J Mol Cell Cardiol ; 117: 88-99, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29428638

RESUMEN

Arterial stiffness plays a causal role in development of systolic hypertension. 20-hydroxyeicosatetraeonic acid (20-HETE), a cytochrome P450 (CYP450)-derived arachidonic acid metabolite, is known to be elevated in resistance arteries in hypertensive animal models and loosely associated with obesity in humans. However, the role of 20-HETE in the regulation of large artery remodeling in metabolic syndrome has not been investigated. We hypothesized that elevated 20-HETE in metabolic syndrome increases matrix metalloproteinase 12 (MMP12) activation leading to increased degradation of elastin, increased large artery stiffness and increased systolic blood pressure. 20-HETE production was increased ~7 fold in large, conduit arteries of metabolic syndrome (JCR:LA-cp, JCR) vs. normal Sprague-Dawley (SD) rats. This correlated with increased elastin degradation (~7 fold) and decreased arterial compliance (~75% JCR vs. SD). 20-HETE antagonists blocked elastin degradation in JCR rats concomitant with blocking MMP12 activation. 20-HETE antagonists normalized, and MMP12 inhibition (pharmacological and MMP12-shRNA-Lnv) significantly improved (~50% vs. untreated JCR) large artery compliance in JCR rats. 20-HETE antagonists also decreased systolic (182 ±â€¯3 mmHg JCR, 145 ±â€¯3 mmHg JCR + 20-HETE antagonists) but not diastolic blood pressure in JCR rats. Whereas diastolic pressure was fully angiotensin II (Ang II)-dependent, systolic pressure was only partially Ang II-dependent, and large artery stiffness was Ang II-independent. Thus, 20-HETE-dependent regulation of systolic blood pressure may be a unique feature of metabolic syndrome related to high 20-HETE production in large, conduit arteries, which results in increased large artery stiffness and systolic blood pressure. These findings may have implications for management of systolic hypertension in patients with metabolic syndrome.


Asunto(s)
Presión Sanguínea , Ácidos Hidroxieicosatetraenoicos/metabolismo , Hipertensión/enzimología , Hipertensión/fisiopatología , Metaloproteinasa 12 de la Matriz/metabolismo , Síndrome Metabólico/enzimología , Síndrome Metabólico/fisiopatología , Rigidez Vascular , Animales , Colágeno Tipo I/metabolismo , Colágeno Tipo III/metabolismo , Adaptabilidad , Citocromo P-450 CYP4A/metabolismo , Familia 4 del Citocromo P450/metabolismo , Diástole/efectos de los fármacos , Elastina/metabolismo , Activación Enzimática/efectos de los fármacos , Hipertensión/complicaciones , Losartán/farmacología , Masculino , Síndrome Metabólico/complicaciones , Proteolisis/efectos de los fármacos , ARN Interferente Pequeño/metabolismo , Ratas Sprague-Dawley , Rigidez Vascular/efectos de los fármacos
3.
Am J Physiol Heart Circ Physiol ; 313(2): H293-H303, 2017 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-28550179

RESUMEN

We have recently demonstrated that disruption of the murine cytochrome P-450 2c44 gene (Cyp2c44) exacerbates chronic hypoxia-induced pulmonary artery remodeling and hypertension in mice. Subsequently, we serendipitously found that Cyp2c44 gene disruption also increases hematopoietic stem cell (HSC) numbers in bone marrow and blood. Therefore, the objective of the present study was to investigate whether CYP2C44-derived eicosanoids regulate HSC proliferation/cell growth and whether increased HSCs contribute to chronic hypoxia-induced remodeling of pulmonary arteries in Cyp2c44 knockout mice. Our findings demonstrated that lack of CYP2C44 epoxygenase, which catalyzed the oxidation of arachidonic acid to epoxyeicosatrienoic (EETs) and hydroxyeicosatetraenoic (HETE) acids, increases the numbers of 1) HSCs (CD34+, CD117+, and CD133+), 2) proangiogenic (CD34+CD133+ and CD34+CD117+CD133+) cells, and 3) immunogenic/inflammatory (CD34+CD11b+, CD133+CD11b+, F4/80+, CD11b+, and F4/80+CD11b+) macrophages in bone marrow and blood compared with wild-type mice. Among the various CYP2C44-derived arachidonic acids, only 15-HETE decreased CD117+ cell numbers when applied to bone marrow cell cultures. Interestingly, CD133+ and von Willebrand factor-positive cells, which are derived from proangiogenic stem cells, are increased in the bone marrow, blood, and lungs of mice exposed to chronic hypoxia and in remodeled and occluded pulmonary arteries of CYP2C44-deficient mice. In conclusion, our results demonstrate that CYP2C44-derived 15-HETE plays a critical role in downregulating HSC proliferation and growth, because disruption of the Cyp2c44 gene increased HSCs that potentially contribute to chronic hypoxia-induced pulmonary arterial remodeling and occlusion.NEW & NOTEWORTHY This study demonstrates that cytochrome P-450 2C44 plays a critical role in controlling the phenotype of hematopoietic stem cells and that when this enzyme is knocked out, stem cells are differentiated. These stem cells give rise to increased circulating monocytes and macrophages and contribute to the pathogenesis of chronic hypoxia-induced pulmonary artery remodeling and hypertension.


Asunto(s)
Proliferación Celular , Familia 2 del Citocromo P450/deficiencia , Células Madre Hematopoyéticas/enzimología , Ácidos Hidroxieicosatetraenoicos/metabolismo , Hipertensión Pulmonar/enzimología , Hipoxia/complicaciones , Arteria Pulmonar/enzimología , Remodelación Vascular , Antígeno AC133/metabolismo , Animales , Antígenos CD34/metabolismo , Antígenos de Diferenciación/metabolismo , Antígeno CD11b/metabolismo , Diferenciación Celular , Células Cultivadas , Enfermedad Crónica , Familia 2 del Citocromo P450/genética , Modelos Animales de Enfermedad , Femenino , Predisposición Genética a la Enfermedad , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/patología , Hipertensión Pulmonar/fisiopatología , Macrófagos/enzimología , Masculino , Ratones de la Cepa 129 , Ratones Noqueados , Monocitos/enzimología , Fenotipo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Arteria Pulmonar/patología , Arteria Pulmonar/fisiopatología , Transducción de Señal
4.
Am J Physiol Heart Circ Physiol ; 311(3): H689-98, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27422986

RESUMEN

Heart failure, a major cause of morbidity and mortality in patients with pulmonary arterial hypertension (PAH), is an outcome of complex biochemical processes. In this study, we determined changes in microRNAs (miRs) in the right and left ventricles of normal and PAH rats. Using an unbiased quantitative miR microarray analysis, we found 1) miR-21-5p, miR-31-5 and 3p, miR-140-5 and 3p, miR-208b-3p, miR-221-3p, miR-222-3p, miR-702-3p, and miR-1298 were upregulated (>2-fold; P < 0.05) in the right ventricle (RV) of PAH compared with normal rats; 2) miR-31-5 and 3p, and miR-208b-3p were upregulated (>2-fold; P < 0.05) in the left ventricle plus septum (LV+S) of PAH compared with normal rats; 3) miR-187-5p, miR-208a-3p, and miR-877 were downregulated (>2-fold; P < 0.05) in the RV of PAH compared with normal rats; and 4) no miRs were up- or downregulated with >2-fold in LV+S compared with RV of PAH and normal. Upregulation of miR-140 and miR-31 in the hypertrophic RV was further confirmed by quantitative PCR. Interestingly, compared with control rats, expression of mitofusin-1 (MFN1), a mitochondrial fusion protein that regulates apoptosis, and which is a direct target of miR-140, was reduced in the RV relative to LV+S of PAH rats. We found a correlation between increased miR-140 and decreased MFN1 expression in the hypertrophic RV. Our results also demonstrated that upregulation of miR-140 and downregulation of MFN1 correlated with increased RV systolic pressure and hypertrophy. These results suggest that miR-140 and MFN1 play a role in the pathogenesis of PAH-associated RV dysfunction.


Asunto(s)
Ventrículos Cardíacos/metabolismo , Hipertensión Pulmonar/metabolismo , Hipertrofia Ventricular Derecha/metabolismo , Proteínas de la Membrana/metabolismo , MicroARNs/metabolismo , Proteínas Mitocondriales/metabolismo , Disfunción Ventricular Derecha/metabolismo , Animales , Apoptosis , Western Blotting , Línea Celular , ADN Mitocondrial/metabolismo , Modelos Animales de Enfermedad , Regulación hacia Abajo , Masculino , Potencial de la Membrana Mitocondrial , Reacción en Cadena de la Polimerasa , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba
5.
Am J Physiol Heart Circ Physiol ; 311(4): H904-H912, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27521420

RESUMEN

Homeostatic control of vascular smooth muscle cell (VSMC) differentiation is critical for contractile activity and regulation of blood flow. Recently, we reported that precontracted blood vessels are relaxed and the phenotype of VSMC is regulated from a synthetic to contractile state by glucose-6-phosphate dehydrogenase (G6PD) inhibition. In the current study, we investigated whether the increase in the expression of VSMC contractile proteins by inhibition and knockdown of G6PD is mediated through a protein kinase G (PKG)-dependent pathway and whether it regulates blood pressure. We found that the expression of VSMC-restricted contractile proteins, myocardin (MYOCD), and miR-1 and miR-143 are increased by G6PD inhibition or knockdown. Importantly, RNA-sequence analysis of aortic tissue from G6PD-deficient mice revealed uniform increases in VSMC-restricted genes, particularly those regulated by the MYOCD-serum response factor (SRF) switch. Conversely, expression of Krüppel-like factor 4 (KLF4) is decreased by G6PD inhibition. Interestingly, the G6PD inhibition-induced expression of miR-1 and contractile proteins was blocked by Rp-ß-phenyl-1,N2-etheno-8-bromo-guanosine-3',5'-cyclic monophosphorothioate, a PKG inhibitor. On the other hand, MYOCD and miR-143 levels are increased by G6PD inhibition through a PKG-independent manner. Furthermore, blood pressure was lower in the G6PD-deficient compared with wild-type mice. Therefore, our results suggest that the expression of VSMC contractile proteins induced by G6PD inhibition occurs via PKG1α-dependent and -independent pathways.


Asunto(s)
Aorta/metabolismo , Proteínas Contráctiles/genética , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/metabolismo , Glucosafosfato Deshidrogenasa/antagonistas & inhibidores , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Animales , Aorta/efectos de los fármacos , Western Blotting , Bovinos , Cromatografía Liquida , Proteínas Contráctiles/efectos de los fármacos , Proteínas Contráctiles/metabolismo , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/antagonistas & inhibidores , Proteínas Quinasas Dependientes de GMP Cíclico/antagonistas & inhibidores , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Técnicas de Silenciamiento del Gen , Glucosafosfato Deshidrogenasa/genética , Inmunoprecipitación , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/efectos de los fármacos , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , MicroARNs/efectos de los fármacos , MicroARNs/genética , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Proteínas Nucleares/efectos de los fármacos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Reacción en Cadena de la Polimerasa , Ratas , Factor de Respuesta Sérica/efectos de los fármacos , Factor de Respuesta Sérica/genética , Factor de Respuesta Sérica/metabolismo , Espectrometría de Masas en Tándem , Transactivadores/efectos de los fármacos , Transactivadores/genética , Transactivadores/metabolismo
6.
Am J Physiol Heart Circ Physiol ; 310(9): H1107-17, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-26921441

RESUMEN

20-Hydroxyeicosatetraeonic acid (20-HETE) produced by cytochrome P-450 monooxygenases in NADPH-dependent manner is proinflammatory, and it contributes to the pathogenesis of systemic and pulmonary hypertension. In this study, we tested the hypothesis that inhibition of glucose-6-phosphate dehydrogenase (G6PD), a major source of NADPH in the cell, prevents 20-HETE synthesis and 20-HETE-induced proinflammatory signaling that promotes secretory phenotype of vascular smooth muscle cells. Lipidomic analysis indicated that G6PD inhibition and knockdown decreased 20-HETE levels in pulmonary arteries as well as 20-HETE-induced 1) mitochondrial superoxide production, 2) activation of mitogen-activated protein kinase 1 and 3, 3) phosphorylation of ETS domain-containing protein Elk-1 that activate transcription of tumor necrosis factor-α gene (Tnfa), and 4) expression of tumor necrosis factor-α (TNF-α). Moreover, inhibition of G6PD increased protein kinase G1α activity, which, at least partially, mitigated superoxide production and Elk-1 and TNF-α expression. Additionally, we report here for the first time that 20-HETE repressed miR-143, which suppresses Elk-1 expression, and miR-133a, which is known to suppress synthetic/secretory phenotype of vascular smooth muscle cells. In summary, our findings indicate that 20-HETE elicited mitochondrial superoxide production and promoted secretory phenotype of vascular smooth muscle cells by activating MAPK1-Elk-1, all of which are blocked by inhibition of G6PD.


Asunto(s)
Antiinflamatorios/farmacología , Inhibidores Enzimáticos/farmacología , Glucosafosfato Deshidrogenasa/antagonistas & inhibidores , Ácidos Hidroxieicosatetraenoicos/metabolismo , Inflamación/prevención & control , Mitocondrias Musculares/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Superóxidos/metabolismo , Animales , Aorta Torácica/efectos de los fármacos , Aorta Torácica/enzimología , Bovinos , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Citocromo P-450 CYP4A/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Genotipo , Glucosafosfato Deshidrogenasa/genética , Glucosafosfato Deshidrogenasa/metabolismo , Inflamación/enzimología , Inflamación/genética , Mediadores de Inflamación/metabolismo , Masculino , Ratones Mutantes , MicroARNs/genética , MicroARNs/metabolismo , Mitocondrias Musculares/enzimología , Músculo Liso Vascular/enzimología , Mutación , Miocitos del Músculo Liso/enzimología , Fenotipo , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/enzimología , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Proteína Elk-1 con Dominio ets/metabolismo
7.
Am J Physiol Lung Cell Mol Physiol ; 307(7): L545-56, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25063801

RESUMEN

Although hypoxia is detrimental to most cell types, it aids survival of progenitor cells and is associated with diseases like cancer and pulmonary hypertension in humans. Therefore, understanding the underlying mechanisms that promote survival of progenitor cells in hypoxia and then developing novel therapies to stop their growth in hypoxia-associated human diseases is important. Here we demonstrate that the proliferation and growth of human CD133(+) progenitor cells, which contribute to tumorigenesis and the development of pulmonary hypertension, are increased when cultured under hypoxic conditions. Furthermore, glucose-6-phosphate dehydrogenase (G6PD) activity was increased threefold in hypoxic CD133(+) cells. The increased G6PD activity was required for CD133(+) cell proliferation, and their growth was arrested by G6PD inhibition or knockdown. G6PD activity upregulated expression of HIF1α, cyclin A, and phospho-histone H3, thereby promoting CD133(+) cell dedifferentiation and self-renewal and altering cell cycle regulation. When CD133(+) cells were cocultured across a porous membrane from pulmonary artery smooth muscle cells (PASMCs), G6PD-dependent H2O2 production and release by PASMCs recruited CD133(+) cells to the membrane, where they attached and expressed smooth muscle markers (α-actin and SM22α). Inhibition of G6PD reduced smooth muscle marker expression in CD133(+) cells under normoxia but not hypoxia. In vivo, CD133(+) cells colocalized with G6PD(+) cells in the perivascular region of lungs from rats with hypoxia-induced pulmonary hypertension. Finally, inhibition of G6PD by dehydroepiandrosterone in pulmonary arterial hypertensive rats nearly abolished CD133(+) cell accumulation around pulmonary arteries and the formation of occlusive lesions. These observations suggest G6PD plays a key role in increasing hypoxia-induced CD133(+) cell survival in hypertensive lungs that differentiate to smooth muscle cells and contribute to pulmonary arterial remodeling during development of pulmonary hypertension.


Asunto(s)
Antígenos CD/metabolismo , Proliferación Celular , Glucosafosfato Deshidrogenasa/fisiología , Glicoproteínas/metabolismo , Hipertensión Pulmonar/enzimología , Péptidos/metabolismo , Células Madre/enzimología , Antígeno AC133 , Administración Oral , Animales , Diferenciación Celular , Hipoxia de la Célula , Técnicas de Cocultivo , Deshidroepiandrosterona/administración & dosificación , Glucosafosfato Deshidrogenasa/antagonistas & inhibidores , Humanos , Hipertensión Pulmonar/tratamiento farmacológico , Hipertensión Pulmonar/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Pulmón/patología , Masculino , Transporte de Proteínas , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/enzimología , Arteria Pulmonar/patología , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Células Madre/fisiología , Factor de Crecimiento Transformador beta/metabolismo
9.
Pulm Circ ; 6(3): 360-8, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27683613

RESUMEN

Epoxyeicosatrienoicacids (EETs), synthesized from arachidonic acid by epoxygenases of the CYP2C and CYP2J gene subfamilies, contribute to hypoxic pulmonary vasoconstriction (HPV) in mice. Despite their roles in HPV, it is controversial whether EETs mediate or ameliorate pulmonary hypertension (PH). A recent study showed that deficiency of Cyp2j did not protect male and female mice from hypoxia-induced PH. Since CYP2C44 is a functionally important epoxygenase, we hypothesized that knockout of the Cyp2c44 gene would protect both sexes of mice from hypoxia-induced PH. We tested this hypothesis in wild-type (WT) and Cyp2c44 knockout (Cyp2c44 (-/-)) mice exposed to normoxia (room air) and hypoxia (10% O2) for 5 weeks. Exposure of WT and Cyp2c44 (-/-) mice to hypoxia resulted in pulmonary vascular remodeling, increased pulmonary artery resistance, and decreased cardiac function in both sexes. However, in female Cyp2c44 (-/-) mice, compared with WT mice, (1) pulmonary artery resistance and right ventricular hypertrophy were greater, (2) cardiac index was lower, (3) left ventricular and arterial stiffness were higher, and (4) plasma aldosterone levels were higher, but (5) there was no difference in levels of EET in lungs and heart. Paradoxically and unexpectedly, we found that Cyp2c44 disruption exacerbated hypoxia-induced PH in female but not male mice. We attribute exacerbated PH in female Cyp2c44 (-/-) mice to elevated aldosterone and as-yet-unknown systemic factors. Therefore, we suggest a role for the human CYP2C genes in protecting women from severe PH and that this could be one of the underlying causes for a better 5-year survival rate in women than in men.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA