Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Arterioscler Thromb Vasc Biol ; 44(4): 826-842, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38328937

RESUMEN

BACKGROUND: Coronary microvascular dysfunction (CMD) has been shown to contribute to cardiac hypertrophy and heart failure (HF) with preserved ejection fraction. At this point, there are no proven treatments for CMD. METHODS: We have shown that histone acetylation may play a critical role in the regulation of CMD. By using a mouse model that replaces lysine with arginine at residues K98, K117, K161, and K162R of p53 (p534KR), preventing acetylation at these sites, we test the hypothesis that acetylation-deficient p534KR could improve CMD and prevent the progression of hypertensive cardiac hypertrophy and HF. Wild-type and p534KR mice were subjected to pressure overload by transverse aortic constriction to induce cardiac hypertrophy and HF. RESULTS: Echocardiography measurements revealed improved cardiac function together with a reduction of apoptosis and fibrosis in p534KR mice. Importantly, myocardial capillary density and coronary flow reserve were significantly improved in p534KR mice. Moreover, p534KR upregulated the expression of cardiac glycolytic enzymes and Gluts (glucose transporters), as well as the level of fructose-2,6-biphosphate; increased PFK-1 (phosphofructokinase 1) activity; and attenuated cardiac hypertrophy. These changes were accompanied by increased expression of HIF-1α (hypoxia-inducible factor-1α) and proangiogenic growth factors. Additionally, the levels of SERCA-2 were significantly upregulated in sham p534KR mice, as well as in p534KR mice after transverse aortic constriction. In vitro, p534KR significantly improved endothelial cell glycolytic function and mitochondrial respiration and enhanced endothelial cell proliferation and angiogenesis. Similarly, acetylation-deficient p534KR significantly improved coronary flow reserve and rescued cardiac dysfunction in SIRT3 (sirtuin 3) knockout mice. CONCLUSIONS: Our data reveal the importance of p53 acetylation in coronary microvascular function, cardiac function, and remodeling and may provide a promising approach to improve hypertension-induced CMD and to prevent the transition of cardiac hypertrophy to HF.


Asunto(s)
Insuficiencia Cardíaca , Hipertensión , Isquemia Miocárdica , Animales , Ratones , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Acetilación , Cardiomegalia/metabolismo , Miocardio/metabolismo , Isquemia Miocárdica/metabolismo , Ratones Noqueados , Hipertensión/metabolismo
2.
Chaos ; 34(8)2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-39088350

RESUMEN

Human games are inherently diverse, involving more than mere identity interactions. The diversity of game tasks offers a more authentic explanation in the exploration of social dilemmas. Human behavior is also influenced by conformity, and prosociality is a crucial factor in addressing social dilemmas. This study proposes a generalized prisoner's dilemma model of task diversity that incorporates a conformity-driven interaction. Simulation findings indicate that the diversity of multi-tasks and the path dependence contribute to the flourishing of cooperation in games. Conformity-driven interactions also promote cooperation. However, this promotion effect does not increase linearly, and only appropriate task sizes and suitable proportions of conformity-driven interactions yield optimal results. From a broader group perspective, the interplay of network adaptation, task size, and conformity-driven interaction can form a structure of attractors or repellents.


Asunto(s)
Conducta Cooperativa , Teoría del Juego , Humanos , Evolución Biológica , Dilema del Prisionero , Simulación por Computador
3.
Int J Mol Sci ; 25(4)2024 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-38397106

RESUMEN

Hypertension is the key contributor to pathological cardiac hypertrophy. Growing evidence indicates that glucose metabolism plays an essential role in cardiac hypertrophy. TP53-induced glycolysis and apoptosis regulator (TIGAR) has been shown to regulate glucose metabolism in pressure overload-induced cardiac remodeling. In the present study, we investigated the role of TIGAR in cardiac remodeling during Angiotensin II (Ang-II)-induced hypertension. Wild-type (WT) and TIGAR knockout (KO) mice were infused with Angiotensin-II (Ang-II, 1 µg/kg/min) via mini-pump for four weeks. The blood pressure was similar between the WT and TIGAR KO mice. The Ang-II infusion resulted in a similar reduction of systolic function in both groups, as evidenced by the comparable decrease in LV ejection fraction and fractional shortening. The Ang-II infusion also increased the isovolumic relaxation time and myocardial performance index to the same extent in WT and TIGAR KO mice, suggesting the development of similar diastolic dysfunction. However, the knockout of TIGAR significantly attenuated hypertension-induced cardiac hypertrophy. This was associated with higher levels of fructose 2,6-bisphosphate, PFK-1, and Glut-4 in the TIGAR KO mice. Our present study suggests that TIGAR is involved in the control of glucose metabolism and glucose transporters by Ang-II and that knockout of TIGAR attenuates the development of maladaptive cardiac hypertrophy.


Asunto(s)
Angiotensina II , Proteínas Reguladoras de la Apoptosis , Cardiomegalia , Hipertensión , Animales , Ratones , Angiotensina II/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Cardiomegalia/genética , Cardiomegalia/inducido químicamente , Fibrosis , Glucosa/metabolismo , Glucólisis , Hipertensión/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Remodelación Ventricular/fisiología
4.
Chaos ; 33(2): 023114, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36859219

RESUMEN

Human behaviors are often subject to conformity, but little research attention has been paid to social dilemmas in which players are assumed to only pursue the maximization of their payoffs. The present study proposed a generalized prisoner dilemma model in a signed network considering conformity. Simulation shows that conformity helps promote the imitation of cooperative behavior when positive edges dominate the network, while negative edges may impede conformity from fostering cooperation. The logic of homophily and xenophobia allows for the coexistence of cooperators and defectors and guides the evolution toward the equality of the two strategies. We also find that cooperation prevails when individuals have a higher probability of adjusting their relation signs, but conformity may mediate the effect of network adaptation. From a population-wide view, network adaptation and conformity are capable of forming the structures of attractors or repellers.

5.
J Cell Physiol ; 237(8): 3317-3327, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35621078

RESUMEN

Hypertension is an important risk factor in the pathogenesis of diastolic dysfunction. Growing evidence indicates that glucose metabolism plays an essential role in diastolic dysfunction. TP53-induced glycolysis and apoptosis regulator (TIGAR) has been shown to regulate glucose metabolism and heart failure (HF). In the present study, we investigated the role of TIGAR in diastolic function and cardiac fibrosis during pressure overload (PO)-induced HF. WT mice subjected to transverse aortic constriction (TAC), a commonly used method to induce diastolic dysfunction, exhibited diastolic dysfunction as evidenced by increased E/A ratio and E/E' ratio when compared to its sham controls. This was accompanied by increased cardiac interstitial fibrosis. In contrast, the knockout of TIGAR attenuated PO-induced diastolic dysfunction and interstitial fibrosis. Mechanistically, the levels of glucose transporter Glut-1, Glut-4, and key glycolytic enzyme phosphofructokinase 1 (PFK-1) were significantly elevated in TIGAR KO subjected to TAC as compared to that of WT mice. Knockout of TIGAR significantly increased fructose 2,6-bisphosphate levels and phosphofructokinase activity in mouse hearts. In addition, PO resulted in a significant increase in perivascular fibrosis and endothelial activation in the WT mice, but not in the TIGAR KO mice. Our present study suggests a necessary role of TIGAR-mediated glucose metabolism in PO-induced cardiac fibrosis and diastolic dysfunction.


Asunto(s)
Proteínas Reguladoras de la Apoptosis , Insuficiencia Cardíaca , Fosfofructoquinasas , Monoéster Fosfórico Hidrolasas , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Diástole , Modelos Animales de Enfermedad , Fibrosis , Glucosa/metabolismo , Glucólisis , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/enzimología , Fosfofructoquinasas/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo
6.
J Cell Physiol ; 236(11): 7578-7590, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33928637

RESUMEN

Endothelial glycolytic metabolism plays an important role in the process of angiogenesis. TP53-induced glycolysis and apoptosis regulator (TIGAR) is a significant mediator of cellular energy homeostasis. However, the role of TIGAR in endothelial metabolism, angiogenesis, and coronary flow reserve (CFR) has not been studied. The present study investigated whether knockout (KO) of TIGAR improves endothelial glycolytic function and angiogenesis. In vitro, aortic endothelial cells (ECs) from TIGAR KO mice exhibited increased expression of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase isoform-3 (PFKFB3) and increased glycolytic function. These were accompanied by increased mitochondrial basal/maximal respiration and ATP production. Furthermore, knockout of TIGAR in ECs enhanced endothelial proliferation, migration, and tube formation. Knockout of TIGAR also significantly increased aortic sprouting ex vivo. In vivo, knockout of TIGAR increased the expression of proangiogenic factor, angiopoietin-1 (Ang-1) in mouse hearts. Knockout of TIGAR also significantly increased coronary capillary density with enhanced CFR in these hearts. Furthermore, TIGAR KO mice subjected to pressure overload (PO), a common model to study angiogenesis and cardiac hypertrophy, exhibited elevated expression of Ang-1, VEGF, and PFKFB3 than that of the wild-type (WT) mice. WT mice subjected to PO exhibited a significant reduction of coronary capillary density and impaired CFR, but TIGAR KO mice did not. In addition, knockout of TIGAR blunted TAC-induced cardiac hypertrophy and dysfunction seen in the WT mice. In conclusion, knockout of TIGAR improves endothelial angiogenetic capabilities by enhancing the endothelial glycolytic function, mitochondrial respiration, and proangiogenic signaling, which leads to increased coronary capillary density and vascular function and protects against chronic stress.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Cardiomegalia/metabolismo , Vasos Coronarios/metabolismo , Células Endoteliales/metabolismo , Glucólisis , Neovascularización Fisiológica , Monoéster Fosfórico Hidrolasas/metabolismo , Angiopoyetina 1/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/genética , Cardiomegalia/genética , Cardiomegalia/patología , Cardiomegalia/fisiopatología , Movimiento Celular , Proliferación Celular , Células Cultivadas , Circulación Coronaria , Vasos Coronarios/patología , Modelos Animales de Enfermedad , Células Endoteliales/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Densidad Microvascular , Mitocondrias/genética , Mitocondrias/metabolismo , Fosfofructoquinasa-2/metabolismo , Monoéster Fosfórico Hidrolasas/genética , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/metabolismo , Función Ventricular Izquierda
7.
J Am Soc Nephrol ; 31(4): 687-700, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32029431

RESUMEN

BACKGROUND: The genes and mechanisms involved in the association between diabetes or hypertension and CKD risk are unclear. Previous studies have implicated a role for γ-adducin (ADD3), a cytoskeletal protein encoded by Add3. METHODS: We investigated renal vascular function in vitro and in vivo and the susceptibility to CKD in rats with wild-type or mutated Add3 and in genetically modified rats with overexpression or knockout of ADD3. We also studied glomeruli and primary renal vascular smooth muscle cells isolated from these rats. RESULTS: This study identified a K572Q mutation in ADD3 in fawn-hooded hypertensive (FHH) rats-a mutation previously reported in Milan normotensive (MNS) rats that also develop kidney disease. Using molecular dynamic simulations, we found that this mutation destabilizes a critical ADD3-ACTIN binding site. A reduction of ADD3 expression in membrane fractions prepared from the kidney and renal vascular smooth muscle cells of FHH rats was associated with the disruption of the F-actin cytoskeleton. Compared with renal vascular smooth muscle cells from Add3 transgenic rats, those from FHH rats had elevated membrane expression of BKα and BK channel current. FHH and Add3 knockout rats exhibited impairments in the myogenic response of afferent arterioles and in renal blood flow autoregulation, which were rescued in Add3 transgenic rats. We confirmed these findings in a genetic complementation study that involved crossing FHH and MNS rats that share the ADD3 mutation. Add3 transgenic rats showed attenuation of proteinuria, glomerular injury, and kidney fibrosis with aging and mineralocorticoid-induced hypertension. CONCLUSIONS: This is the first report that a mutation in ADD3 that alters ACTIN binding causes renal vascular dysfunction and promotes the susceptibility to kidney disease.


Asunto(s)
Proteínas de Unión a Calmodulina/genética , Hipertensión/complicaciones , Enfermedades Renales/etiología , Mutación/efectos de los fármacos , Circulación Renal/genética , Animales , Modelos Animales de Enfermedad , Homeostasis , Hipertensión/genética , Glomérulos Renales/metabolismo , Glomérulos Renales/patología , Masculino , Músculo Liso Vascular/fisiología , Miocitos del Músculo Liso/metabolismo , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas
8.
Am J Physiol Renal Physiol ; 319(2): F345-F357, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32715763

RESUMEN

Angiotensin II (ANG II) is the key contributor to renal fibrosis and injury. The present study investigated the role of endothelium prolyl hydroxylase 2 (PHD2) in ANG II-mediated renal fibrosis and injury. In vitro, endothelial cells (ECs) were isolated from PHD2f/f control [wild-type (WT)] mice or PHD2 EC knockout (PHD2ECKO) mice. In vivo, WT and PHD2ECKO mice were infused with ANG II (1,000 ng·kg-1·min-1) for 28 days. Renal fibrosis, reactive oxygen species (ROS), and iron contents were measured. Knockout of PHD2 resulted in a significant increase in the expression of hypoxia-inducible factor (HIF)-1α and HIF-2α in ECs. Intriguingly, knockout of PHD2 significantly reduced expression of the ANG II type 1 receptor (AT1R) in ECs. WT mice infused with ANG II caused increases in renal fibrosis, ROS formation, and iron contents. ANG II treatment led to a downregulation of PHD1 expression and upregulation of HIF-1α and HIF-2α in the renal cortex and medulla. Knockout of PHD2 in EC blunted ANG II-induced downregulation of PHD1 expression. Furthermore, knockout of PHD2 in ECs attenuated ANG II-induced expression of HIF-1α, HIF-2α, transforming growth factor-ß1, p47phox, gp91phox, heme oxygenase-1, and ferroportin. This was accompanied by a significant suppression of renal fibrosis, ROS formation, and iron accumulation. In summary, knockout of endothelial PHD2 suppressed the expression of AT1R in ECs and blunted ANG II-induced downregulation of PHD1 and upregulation of HIF-α in the kidney. Our study, for the first time, demonstrates a necessary role of endothelial PHD2 in ANG II-mediated renal fibrosis and injury.


Asunto(s)
Angiotensina II/metabolismo , Células Endoteliales/metabolismo , Fibrosis/metabolismo , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Enfermedades Renales/enzimología , Riñón/lesiones , Angiotensina II/farmacología , Animales , Células Endoteliales/efectos de los fármacos , Endotelio/metabolismo , Prolina Dioxigenasas del Factor Inducible por Hipoxia/genética , Riñón/enzimología , Ratones , Ratones Noqueados , Procolágeno-Prolina Dioxigenasa/genética , Procolágeno-Prolina Dioxigenasa/metabolismo , ARN Interferente Pequeño/metabolismo
9.
Int J Mol Sci ; 21(24)2020 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-33371209

RESUMEN

BACKGROUND: Heart failure with preserved ejection fraction (HFpEF) is characterized by a diastolic dysfunction and is highly prevalent in aged women. Our study showed that ablation of endothelial Sirtuin 3 (SIRT3) led to diastolic dysfunction in male mice. However, the sex-specific role of endothelial SIRT3 deficiency on blood pressure and diastolic function in female mice remains to be investigated. METHODS AND RESULTS: In this study, we demonstrate that the ablation of endothelial SIRT3 in females elevated blood pressure as compared with control female mice. Diastolic function measurement also showed that the isovolumic relaxation time (IVRT) and myocardial performance index (MPI) were significantly increased, whereas the E' velocity/A' velocity (E'/A') ratio was reduced in the endothelial-specific SIRT3 knockout (SIRT3 ECKO) female mice. To further investigate the regulatory role of endothelial SIRT3 on blood pressure and diastolic dysfunction in metabolic stress, SIRT3 ECKO female mice were fed a normal diet and high-fat diet (HFD) for 20 weeks. The knockout of endothelial SIRT3 resulted in an increased blood pressure in female mice fed with an HFD. Intriguingly, SIRT3 ECKO female mice + HFD exhibited impaired coronary flow reserve (CFR) and more severe diastolic dysfunction as evidenced by an elevated IVRT as compared with control female mice + HFD. In addition, female SIRT3 ECKO mice had higher blood pressure and diastolic dysfunction as compared to male SIRT3 ECKO mice. Moreover, female SIRT3 ECKO mice + HFD had an impaired CFR and diastolic dysfunction as compared to male SIRT3 ECKO mice + HFD. CONCLUSIONS: These results implicate a sex-specific role of endothelial SIRT3 in regulating blood pressure and diastolic function in mice. Deficiency of endothelial SIRT3 may be responsible for a diastolic dysfunction in aged female.


Asunto(s)
Endotelio Vascular/patología , Insuficiencia Cardíaca/patología , Sirtuina 3/fisiología , Volumen Sistólico , Animales , Presión Sanguínea , Endotelio Vascular/metabolismo , Femenino , Insuficiencia Cardíaca/etiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Caracteres Sexuales
10.
J Cell Physiol ; 234(3): 2252-2265, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30132870

RESUMEN

Sirtuin 3 (SIRT3) a mitochondrial enzyme that plays an important role in energy homeostasis, cardiac remodeling, and heart failure (HF). The expression of SIRT3 declines with advanced age, cardiovascular, and metabolic diseases. Accumulating evidence suggests that SIRT3 plays a critical role in protecting the heart from cardiac hypertrophy, cardiac dysfunction associated with HF, and in the protection of cardiac cells from stress-mediated cell death. Clinical studies have demonstrated that HF with preserved ejection fraction (HFpEF) in patients present with abnormalities in coronary microcirculation related to endothelial dysfunction and coronary microvascular rarefaction. Although SIRT3-mediated regulation of mitochondrial homeostasis and heart function has been intensively investigated, the effect of SIRT3 on endothelial cell (EC) glycolytic metabolism and microvascular function has not been well studied. ECs utilize glycolysis for generating ATP rather than oxidative phosphorylation to maintain their normal functions and promote angiogenesis and EC-cardiomyocyte interactions. Emerging evidence indicates that SIRT3 is involved in the regulation of endothelial metabolism and angiogenesis and thus affects the development of cardiovascular diseases associated with aging. This review will discuss the current knowledge of SIRT3 and its functional role on endothelial metabolism, cardiac function, and cardiovascular diseases.


Asunto(s)
Enfermedades Cardiovasculares/genética , Insuficiencia Cardíaca/genética , Neovascularización Patológica/genética , Sirtuina 3/genética , Enfermedades Cardiovasculares/patología , Células Endoteliales/metabolismo , Células Endoteliales/patología , Glucólisis/genética , Insuficiencia Cardíaca/patología , Humanos , Miocitos Cardíacos/metabolismo , Neovascularización Patológica/patología , Volumen Sistólico/genética
11.
J Pharmacol Exp Ther ; 370(2): 206-217, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31118214

RESUMEN

Dual-specificity protein phosphatase 5 (DUSP5) is a member of the tyrosine-threonine phosphatase family with the ability to dephosphorylate and inactivate extracellular signal-related kinase (ERK). The present study investigates whether knockout (KO) of Dusp5 improves renal hemodynamics and protects against hypertension-induced renal injury. The renal expression of DUSP5 was reduced, and the levels of phosphorylated (p) ERK1/2 and p-protein kinase C (PKC) α were elevated in the KO rats. KO of Dusp5 enhanced the myogenic tone of the renal afferent arteriole and interlobular artery in vitro with or without induction of deoxycorticosterone acetate-salt hypertension. Inhibition of ERK1/2 and PKC diminished the myogenic response to a greater extent in Dusp5 KO rats. Autoregulation of renal blood flow was significantly impaired in hypertensive wild-type (WT) rats but remained intact in Dusp5 KO animals. Proteinuria was markedly decreased in hypertensive KO versus WT rats. The degree of glomerular injury was reduced, and the expression of nephrin in the glomerulus was higher in hypertensive Dusp5 KO rats. Renal fibrosis and medullary protein cast formation were attenuated in hypertensive Dusp5 KO rats in association with decreased expression of monocyte chemoattractant protein 1, transforming growth factor-ß1, matrix metalloproteinase (MMP) 2, and MMP9. These results indicate that KO of Dusp5 protects against hypertension-induced renal injury, at least in part, by maintaining the myogenic tone of the renal vasculature and extending the range of renal blood flow autoregulation to higher pressures, which diminish glomerular injury, protein cast formation, macrophage infiltration, and epithelial-mesenchymal transformation in the kidney. SIGNIFICANCE STATEMENT: Dual-specificity protein phosphatase 5 (DUSP5) is a tyrosine-threonine phosphatase that inactivates extracellular signal-related kinase (ERK). We previously reported that knockout (KO) of Dusp5 enhanced the myogenic response and autoregulation in the cerebral circulation. The present study investigates whether KO of DUSP5 improves renal hemodynamics and protects against hypertension-induced renal injury. Downregulation of DUSP5 enhanced the myogenic tone of renal arteriole and artery and autoregulation of renal blood flow in association with reduced proteinuria, glomerular injury, and interstitial fibrosis after the induction of hypertension. Inhibition of ERK1/2 and protein kinase C diminished the myogenic response to a greater extent in Dusp5 KO rats. These results suggest that DUSP5 might be a viable drug target for the treatment of hypertension nephropathy.


Asunto(s)
Fosfatasas de Especificidad Dual/deficiencia , Fosfatasas de Especificidad Dual/genética , Técnicas de Inactivación de Genes , Hipertensión Renal/genética , Nefritis/genética , Animales , Quimiocina CCL2/metabolismo , Regulación hacia Abajo/genética , Transición Epitelial-Mesenquimal/genética , Fibrosis , Regulación Enzimológica de la Expresión Génica/genética , Hemodinámica/genética , Hipertensión Renal/metabolismo , Hipertensión Renal/patología , Hipertensión Renal/fisiopatología , Glomérulos Renales/irrigación sanguínea , Glomérulos Renales/metabolismo , Glomérulos Renales/patología , Masculino , Metaloproteinasas de la Matriz/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Desarrollo de Músculos/genética , Nefritis/metabolismo , Nefritis/patología , Nefritis/fisiopatología , Proteína Quinasa C/metabolismo , Ratas , Flujo Sanguíneo Regional/genética , Factor de Crecimiento Transformador beta/metabolismo
12.
Am J Physiol Renal Physiol ; 315(6): F1843-F1854, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30207168

RESUMEN

The incidence and severity of acute kidney injury is increased in patients with diabetes and with aging. However, the mechanisms involved have not been clearly established. The present study examined the effects of aging and diabetes on the severity of renal ischemia-reperfusion (IR) injury in Sprague-Dawley (SD) and type 2 diabetic (T2DN) rats. T2DN rats develop diabetes at 3 mo of age and progressive proteinuria and diabetic nephropathy as they age from 6 to 18 mo. Plasma creatinine levels after bilateral IR were significantly higher (3.4 ± 0.1 mg/dl) in 18-mo-old elderly T2DN rats than in middle-aged (12 mo) T2DN rats with less severe diabetic nephropathy or young (3 mo) and elderly (18 mo) control SD rats (1.5 ± 0.2, 1.8 ± 0.1, and 1.7 ± 0.1 mg/dl, respectively). Elderly T2DN rats exhibited a greater fall in medullary blood flow 2 h following renal IR and a more severe and prolonged decline in glomerular filtration rate than middle-aged T2DN and young or elderly SD rats. The basal expression of the adhesion molecules ICAM-1 and E-selectin and the number of infiltrating immune cells was higher in the kidney of elderly T2DN than age-matched SD rats or young and middle-aged T2DN rats before renal IR. These results indicate that elderly T2DN rats with diabetic nephropathy are more susceptible to renal IR injury than diabetic animals with mild injury or age-matched control animals. This is associated with increased expression of ICAM-1, E-selectin and immune cell infiltration, renal medullary vasocongestion, and more prolonged renal medullary ischemia.


Asunto(s)
Lesión Renal Aguda/etiología , Envejecimiento , Diabetes Mellitus Tipo 2/complicaciones , Nefropatías Diabéticas/etiología , Riñón/irrigación sanguínea , Circulación Renal , Daño por Reperfusión/etiología , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Lesión Renal Aguda/fisiopatología , Factores de Edad , Envejecimiento/metabolismo , Envejecimiento/patología , Animales , Glucemia/metabolismo , Presión Sanguínea , Creatinina/sangre , Diabetes Mellitus Tipo 2/metabolismo , Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/patología , Nefropatías Diabéticas/fisiopatología , Modelos Animales de Enfermedad , Tasa de Filtración Glomerular , Mediadores de Inflamación/metabolismo , Riñón/metabolismo , Riñón/patología , Masculino , Ratas Sprague-Dawley , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Daño por Reperfusión/fisiopatología , Índice de Severidad de la Enfermedad
13.
J Mol Cell Cardiol ; 112: 104-113, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28935506

RESUMEN

Endothelial glycolysis plays a critical role in the regulation of angiogenesis. We investigated the role of Sirtuin 3 (SIRT3) on endothelial cell (EC) glycolytic metabolism, angiogenesis, and diastolic function. Our aim was to test the hypothesis that loss of SIRT3 in ECs impairs endothelial glycolytic metabolism and angiogenesis and contributes to myocardial capillary rarefaction and the development of diastolic dysfunction. Using SIRT3 deficient ECs, SIRT3 was found to regulate a metabolic switch between mitochondrial respiration and glycolysis. SIRT3 knockout (KO)-ECs exhibited higher mitochondrial respiration and reactive oxygen species (ROS) formation. SIRT3 knockout (KO)-ECs exhibited a reduction in the expression of glycolytic enzyme, PFKFB3, and a fall in glycolysis and angiogenesis. Blockade of PFKFB3 reduced glycolysis and downregulated expression of VEGF and Angiopoietin-1 (Ang-1) in ECs. Deletion of SIRT3 in ECs also impaired hypoxia-induced expression of HIF-2α, VEGF, and Ang-1, as well as reduced angiogenesis. In vivo, endothelial-specific SIRT3 KO (ECKO) mice exhibited a myocardial capillary rarefaction together with a reduced coronary flow reserve (CFR) and diastolic dysfunction. Histologic study further demonstrated that knockout of SIRT3 in ECs significantly increased perivascular fibrosis in the coronary artery. These results implicate a role of SIRT3 in modulating endothelial function and cardiac function. Ablation of SIRT3 leads to impairment of EC glycolytic metabolism and angiogenic signaling, which may contribute to coronary microvascular rarefaction and diastolic dysfunction in SIRT3 ECKO mice.


Asunto(s)
Diástole , Eliminación de Gen , Glucólisis , Corazón/fisiopatología , Neovascularización Fisiológica , Sirtuina 3/metabolismo , Animales , Capilares/metabolismo , Hipoxia de la Célula , Circulación Coronaria , Células Endoteliales/metabolismo , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Ratones Noqueados , Modelos Biológicos , Especificidad de Órganos , Consumo de Oxígeno , Fenotipo , Fosfofructoquinasa-2/metabolismo , Transducción de Señal , Sirtuina 3/deficiencia , Volumen Sistólico
14.
J Cell Mol Med ; 21(9): 1967-1978, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28266128

RESUMEN

Accumulating evidence demonstrates that hypoxia-inducible factor (HIF-α) hydroxylase system has a critical role in vascular remodelling. Using an endothelial-specific prolyl hydroxylase domain protein-2 (PHD2) knockout (PHD2EC KO) mouse model, this study investigates the regulatory role of endothelial HIF-α hydroxylase system in the development of renal fibrosis. Knockout of PHD2 in EC up-regulated the expression of HIF-1α and HIF-2α, resulting in a significant decline of renal function as evidenced by elevated levels of serum creatinine. Deletion of PHD2 increased the expression of Notch3 and transforming growth factor (TGF-ß1) in EC, thus further causing glomerular arteriolar remodelling with an increased pericyte and pericyte coverage. This was accompanied by a significant elevation of renal resistive index (RI). Moreover, knockout of PHD2 in EC up-regulated the expression of fibroblast-specific protein-1 (FSP-1) and increased interstitial fibrosis in the kidney. These alterations were strongly associated with up-regulation of Notch3 and TGF-ß1. We concluded that the expression of PHD2 in endothelial cells plays a critical role in renal fibrosis and vascular remodelling in adult mice. Furthermore, these changes were strongly associated with up-regulation of Notch3/TGF-ß1 signalling and excessive pericyte coverage.


Asunto(s)
Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Riñón/irrigación sanguínea , Riñón/patología , Eliminación de Secuencia , Remodelación Vascular , Animales , Arterias/patología , Arteriolas/patología , Presión Sanguínea , Fibrosis , Regulación de la Expresión Génica , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Riñón/fisiopatología , Glomérulos Renales/patología , Glomérulos Renales/fisiopatología , Ratones Noqueados , Pericitos/metabolismo , Pericitos/patología , Fenotipo
15.
J Cell Mol Med ; 19(1): 53-61, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25311234

RESUMEN

Heart failure following myocardial infarction (MI) is the leading cause of death in diabetic patients. Angiogenesis contributes to cardiac repair and functional recovery in post-MI. Our previous study shows that apelin (APLN) increases Sirtuin 3 (Sirt3) expression and ameliorates diabetic cardiomyopathy. In this study, we further investigated the direct role of Sirt3 in APLN-induced angiogenesis in post-MI model of diabetes. Wild-type (WT) and Sirt3 knockout (Sirt3KO) mice were induced into diabetes by i.p. streptozotocin (STZ). STZ mice were then subjected to MI followed by immediate intramyocardial injection with adenovirus-apelin (Ad-APLN). Our studies showed that Sirt3 expression was significantly reduced in the hearts of STZ mice. Ad-APLN treatment resulted in up-regulation of Sirt3, angiopoietins/Tie-2 and VEGF/VEGFR2 expression together with increased myocardial vascular densities in WT-STZ+MI mice, but these alterations were not observed in Sirt3KO-STZ+MI mice. In vitro, overexpression of APLN increased Sirt3 expression and angiogenesis in endothelial progenitor cells (EPC) from WT mice, but not in EPC from Sirt3KO mice. APLN gene therapy increases angiogenesis and improves cardiac functional recovery in diabetic hearts via up-regulation of Sirt3 pathway.


Asunto(s)
Inductores de la Angiogénesis/metabolismo , Diabetes Mellitus Experimental/complicaciones , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/uso terapéutico , Infarto del Miocardio/terapia , Sirtuina 3/metabolismo , Adenoviridae/metabolismo , Adipoquinas , Animales , Apelina , Apoptosis , Proliferación Celular , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Experimental/fisiopatología , Células Endoteliales/metabolismo , Terapia Genética , Pruebas de Función Cardíaca , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Ratones Noqueados , Infarto del Miocardio/diagnóstico por imagen , Infarto del Miocardio/etiología , Infarto del Miocardio/fisiopatología , Miocardio/patología , Neovascularización Fisiológica , Sirtuina 3/deficiencia , Ultrasonografía
16.
J Cell Mol Med ; 19(8): 1847-56, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25782072

RESUMEN

Mitochondrial dysfunction plays an important role in obesity-induced cardiac impairment. SIRT3 is a mitochondrial protein associated with increased human life span and metabolism. This study investigated the functional role of SIRT3 in obesity-induced cardiac dysfunction. Wild-type (WT) and SIRT3 knockout (KO) mice were fed a normal diet (ND) or high-fat diet (HFD) for 16 weeks. Body weight, fasting glucose levels, reactive oxygen species (ROS) levels, myocardial capillary density, cardiac function and expression of hypoxia-inducible factor (HIF)-1α/-2α were assessed. HFD resulted in a significant reduction in SIRT3 expression in the heart. Both HFD and SIRT3 KO mice showed increased ROS formation, impaired HIF signalling and reduced capillary density in the heart. HFD induced cardiac hypertrophy and impaired cardiac function. SIRT3 KO mice fed HFD showed greater ROS production and a further reduction in cardiac function compared to SIRT3 KO mice on ND. Thus, the adverse effects of HFD on cardiac function were not attributable to SIRT3 loss alone. However, HFD did not further reduce capillary density in SIRT3 KO hearts, implicating SIRT3 loss in HFD-induced capillary rarefaction. Our study demonstrates the importance of SIRT3 in preserving heart function and capillary density in the setting of obesity. Thus, SIRT3 may be a potential therapeutic target for obesity-induced heart failure.


Asunto(s)
Corazón/fisiopatología , Sirtuina 3/metabolismo , Remodelación Ventricular , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Glucemia/metabolismo , Capilares/metabolismo , Cardiomegalia/complicaciones , Cardiomegalia/diagnóstico por imagen , Cardiomegalia/patología , Cardiomegalia/fisiopatología , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Experimental/fisiopatología , Dieta Alta en Grasa , Ayuno/sangre , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Masculino , Ratones Noqueados , Obesidad/complicaciones , Obesidad/patología , Obesidad/fisiopatología , Pericitos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Sirtuina 3/deficiencia , Ultrasonografía
17.
Am J Physiol Heart Circ Physiol ; 306(4): H585-97, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24363305

RESUMEN

Microvascular insufficiency contributes to cardiac hypertrophy and worsens heart dysfunction in diabetic cardiomyopathy. Our recent study shows that apelin may protect ischemic heart failure via upregulation of sirtuin 3 (Sirt3) and angiogenesis. This study investigated whether apelin promotes angiogenesis and ameliorates diabetic cardiomyopathy via activation of Sirt3. Wild-type (WT) and diabetic db/db mice were administrated with adenovirus-apelin to overexpressing apelin. In WT mice, overexpression of apelin increased Sirt3, VEGF/VEGFR2, and angiopoietin-1 (Ang-1)/Tie-2 expression in the heart. In vitro, treatment of endothelial cells (EC) with apelin increased VEGF and Ang-1 expression. In EC isolated from Sirt3KO mice, however, apelin treatment did not upregulate VEGF and Ang-1 expression. Moreover, apelin-induced angiogenesis was diminished in Sirt3KO mice. In db/db mice, the basal levels of apelin and Sirt3 expression were significantly reduced in the heart. This was accompanied by a significant reduction of capillary and arteriole densities in the heart. Overexpression of apelin increased Sirt3, VEGF/VEGFR2, and Ang-1/Tie-2 expression together with improved vascular density in db/db mice. Overexpression of apelin further improved cardiac function in db/db mice. Treatment with apelin significantly attenuated high glucose (HG)-induced reactive oxygen species (ROS) formation and EC apoptosis. The protection of apelin against HG-induced ROS formation and EC apoptosis was diminished in Sirt3KO-EC. We conclude that apelin gene therapy increases vascular density and alleviates diabetic cardiomyopathy by a mechanism involving activation of Sirt3 and upregulation of VEGF/VEGFR2 and Ang-1/Tie-2 expression.


Asunto(s)
Cardiomiopatías Diabéticas/terapia , Péptidos y Proteínas de Señalización Intercelular/genética , Miocardio/metabolismo , Neovascularización Patológica/terapia , Sirtuina 3/genética , Regulación hacia Arriba/genética , Adipoquinas , Angiopoyetina 1/genética , Angiopoyetina 1/metabolismo , Angiopoyetina 2/metabolismo , Animales , Apelina , Apoptosis/fisiología , Cardiomiopatías Diabéticas/genética , Cardiomiopatías Diabéticas/metabolismo , Cardiomiopatías Diabéticas/fisiopatología , Endotelio Vascular/metabolismo , Endotelio Vascular/fisiopatología , Corazón/fisiopatología , Masculino , Ratones , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Neovascularización Patológica/fisiopatología , Especies Reactivas de Oxígeno/metabolismo , Receptor TIE-2/genética , Receptor TIE-2/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular/genética , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Sirtuina 3/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
18.
Geroscience ; 46(4): 3945-3956, 2024 08.
Artículo en Inglés | MEDLINE | ID: mdl-38462569

RESUMEN

Obesity and exercise intolerance greatly reduce the life quality of older people. Prolyl hydroxylase domain-containing protein 2 (PHD2) is an important enzyme in modulating hypoxia-inducible factor-alpha (HIF) protein. Using vascular endothelial cell-specific PHD2 gene knockout (PHD2 ECKO) mice, we investigated the role of endothelial PHD2 in aging-related obesity and exercise capacity. Briefly, PHD2 ECKO mice were obtained by crossing PHD2-floxed mice with VE-Cadherin (Cdh5)-Cre transgenic mice. The effect of PHD2 ECKO on obesity and exercise capacity in PHD2 ECKO mice and control PHD2f/f mice were determined in young mice (6 to 7 months) and aged mice (16-18 months). We found that aged PHD2 ECKO mice, but not young mice, exhibited a lean phenotype, characterized by lower fat mass, and its ratio to lean weight, body weight, or tibial length, while their food uptake was not reduced compared with controls. Moreover, as compared with aged control mice, aged PHD2 ECKO mice exhibited increased oxygen consumption at rest and during exercise, and the maximum rate of oxygen consumption (VO2 max) during exercise. Furthermore, as compared with corresponding control mice, both young and aged PHD2 ECKO mice demonstrated improved glucose tolerance and lower insulin resistance. Together, these data demonstrate that inhibition of vascular endothelial PHD2 signaling significantly attenuates aging-related obesity, exercise intolerance, and glucose intolerance.


Asunto(s)
Envejecimiento , Tolerancia al Ejercicio , Prolina Dioxigenasas del Factor Inducible por Hipoxia , Ratones Noqueados , Obesidad , Animales , Obesidad/genética , Envejecimiento/fisiología , Envejecimiento/genética , Envejecimiento/metabolismo , Prolina Dioxigenasas del Factor Inducible por Hipoxia/genética , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Ratones , Tolerancia al Ejercicio/fisiología , Condicionamiento Físico Animal/fisiología , Masculino , Consumo de Oxígeno/fisiología , Resistencia a la Insulina/fisiología , Resistencia a la Insulina/genética , Modelos Animales de Enfermedad
19.
Redox Biol ; 76: 103325, 2024 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-39197316

RESUMEN

Selenium (Se) deficiency is associated with the development of Keshan disease, a cardiomyopathy associated with massive cardiac immune cell infiltration that can lead to heart failure (HF). The purpose of this study was to determine whether high Se diet can attenuate systolic overload-induced cardiopulmonary inflammation and HF. Briefly, transverse aortic constriction (TAC)-induced cardiopulmonary oxidative stress, inflammation, left ventricular (LV) dysfunction, and pulmonary remodeling were determined in male mice fed with either high Se diet or normal Se diet. High Se diet had no detectable effect on LV structure and function in mice under control conditions, but high Se diet significantly protected mice from TAC-induced LV hypertrophy, dysfunction, increase of lung weight, and right ventricular hypertrophy. As compared with mice treated with normal Se diet, high Se diet also reduced TAC-induced LV cardiomyocyte hypertrophy, fibrosis, leukocyte infiltration, pulmonary inflammation, pulmonary fibrosis, and pulmonary micro-vessel muscularization. In addition, high Se diet significantly ameliorated TAC-induced accumulation and activation of pulmonary F4/80+ macrophages, and activation of dendritic cells. Interestingly, high Se diet also significantly attenuated TAC-induced activation of pulmonary CD4+ and CD8+ T cells. Moreover, we found that TAC caused a significant increase in cardiac and pulmonary ROS production, increases of 4-hydroxynonenal (4-HNE) and 3-nitrotyrosine (3-NT), as well as a compensatory increases of LV glutathione peroxidase 1 (GPX1) and 4 (GPX4) in mice fed with normal Se diet. Above changes were diminished in mice fed with high Se diet. Collectively, these data demonstrated that high Se diet significantly attenuated systolic pressure overload-induced cardiac oxidative stress, inflammation, HF development, and consequent pulmonary inflammation and remodeling.

20.
PLoS One ; 18(9): e0291778, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37725633

RESUMEN

The topological characterization of complex systems has significantly contributed to our understanding of the principles of collective dynamics. However, the representation of general complex networks is not enough for explaining certain problems, such as collective actions. Considering the effectiveness of hypernetworks on modeling real-world complex networks, in this paper, we proposed a hypernetwork-based Pólya urn model that considers the effect of group identity. The mathematical deduction and simulation experiments show that social influence provides a strong imitation environment for individuals, which can prevent the dynamics from being self-correcting. Additionally, the unpredictability of the social system increases with growing social influence, and the effect of group identity can moderate market inequality caused by individual preference and social influence. The present work provides a modeling basis for a better understanding of the logic of collective dynamics.


Asunto(s)
Poli A , Humanos , Simulación por Computador
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA