Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.273
Filtrar
1.
Biochem Biophys Res Commun ; 661: 34-41, 2023 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-37086572

RESUMEN

Physiological activities of the body exhibit an obvious biological rhythm. At the core of the circadian rhythm, BMAL1 is the only clock gene whose deletion leads to abnormal physiological functions. However, whether intermittent heat stress influences cardiovascular function by altering the circadian rhythm of clock genes has not been reported. This study aimed to investigate whether intermittent heat stress induces autophagy and apoptosis, and the effects of BMAL1 on thoracic aortic autophagy and apoptosis. An intermittent heat stress model was established in vitro, and western blotting and immunofluorescence were used to detect the expression of autophagy, apoptosis, the AMPK/mTOR/ULK1 pathway, and BMAL1. After BMAL1 silencing, RT-qPCR was performed to detect the expression levels of autophagy and apoptosis-related genes. Our results suggest that heat stress induces autophagy and apoptosis in RTAECs. In addition, intermittent heat stress increased the phosphorylation of AMPK and ULK1, but reduced the phosphorylation of mTOR, AMPK inhibitor Compound C reversed the phosphorylation of AMPK, mTOR, and ULK1, and Beclin1 and LC3-II/LC3-I were downregulated. Furthermore, BMAL1 expression was elevated in vitro and shBMAL1 decreased autophagy and apoptosis. We revealed that intermittent heat stress induces autophagy and apoptosis, and that BMAL1 may be involved in the occurrence of autophagy and apoptosis.


Asunto(s)
Factores de Transcripción ARNTL , Autofagia , Células Endoteliales , Respuesta al Choque Térmico , Animales , Ratas , Aorta Torácica/citología , Células Endoteliales/citología , Factores de Transcripción ARNTL/metabolismo , Quinasas de la Proteína-Quinasa Activada por el AMP/metabolismo , Transducción de Señal , Fosforilación , Apoptosis , Células Cultivadas
2.
Chem Biol Interact ; 348: 109620, 2021 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-34411564

RESUMEN

Naringenin is a naturally occurring flavanone (flavonoid) known to have bioactive effects on human health. It has been reported to show cardiovascular effects. This study aimed to investigate the possible vasorelaxant effect of naringenin and the mechanism behind it by using a Sprague Dawley rat aortic ring assay model. Naringenin caused significant vasorelaxation of endothelium-intact aortic rings precontracted with phenylephrine (pD2 = 4.27 ± 0.05; Rmax = 121.70 ± 4.04%) or potassium chloride (pD2 = 4.00 ± 0.04; Rmax = 103.40 ± 3.82%). The vasorelaxant effect decreased in the absence of an endothelium (pD2 = 3.34 ± 0.10; Rmax = 62.29 ± 2.73%). The mechanisms of the vasorelaxant effect of naringenin in the presence of antagonists were also investigated. Indomethacin, 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one, atropine, 4-aminopyridine, Nω-nitro-l-arginine methyl ester, glibenclamide and propranolol significantly reduced the relaxation stimulated by naringenin in the presence of endothelium. Besides that, the effect of naringenin on the voltage-operated calcium channel (VOCC) in the endothelium-intact aortic ring was studied, as was intracellular Ca2+ release from the sarcoplasmic reticulum (SR) in the endothelium-denuded aortic ring. The results showed that naringenin also significantly blocked the entry of Ca2+ via the VOCC, SERCA/SOCC and suppressed the release of Ca2+ from the SR. Thus, the vasorelaxant effect shown by naringenin mostly involve the COX pathway, the endothelium-dependent pathway via NO/sGC/prostaglandin, calcium and potassium channels.


Asunto(s)
Aorta Torácica/efectos de los fármacos , Canales de Calcio/metabolismo , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Flavanonas/farmacología , Canales de Potasio/metabolismo , Vasodilatadores/farmacología , Animales , Aorta Torácica/citología , Aorta Torácica/metabolismo , Aorta Torácica/fisiología , Relación Dosis-Respuesta a Droga , Ratas , Ratas Sprague-Dawley
3.
J Steroid Biochem Mol Biol ; 212: 105948, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34224859

RESUMEN

Oxidative stress injury is an important link in the pathogenesis of diabetes, and reducing oxidative stress damage caused by long-term hyperglycemia is an important diabetic treatment strategy. Melatonin has been proved to be a free radical scavenger with strong antioxidant activity, and its protective effect on diabetes and the complications has been confirmed. However, the role and potential mechanism of melatonin in oxidative stress injury of diabetic aorta have not been reported. Besides, Notch signaling pathway plays an important role in vascular growth, differentiation, and apoptosis. We speculated that melatonin could improve oxidative stress injury of diabetic aorta through Notch1/Hes1 signaling pathway. STZ-induced diabetic rats and vascular smooth muscle cells (VSMCs) cultured with high glucose were treated with or without melatonin, melatonin receptor antagonist Luzindole, γ-secretase inhibitor DAPT respectively. We found that melatonin could improve the oxidative stress injury of diabetic aorta and reduce the apoptosis of VSMCs. Interestingly, melatonin could activate Notch1 signaling pathway, play an antioxidant role, and reduce the expression of apoptosis-related proteins. However, these protective effects could be largely eliminated by Luzindole or DAPT. We concluded that the repression of Notch1 signaling pathway would inhibit the repair of oxidative stress injury in diabetes. Melatonin could ameliorate oxidative stress injury and apoptosis of diabetic aorta by activating Notch1/Hes1 signaling pathway.


Asunto(s)
Antioxidantes/uso terapéutico , Diabetes Mellitus Experimental/tratamiento farmacológico , Hipoglucemiantes/uso terapéutico , Melatonina/uso terapéutico , Animales , Antioxidantes/farmacología , Aorta Torácica/citología , Apoptosis/efectos de los fármacos , Glucemia/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/metabolismo , Hipoglucemiantes/farmacología , Masculino , Melatonina/farmacología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Estrés Oxidativo/efectos de los fármacos , Ratas Sprague-Dawley , Receptor Notch1/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Transcripción HES-1/metabolismo
4.
J Am Heart Assoc ; 10(10): e020641, 2021 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-33938228

RESUMEN

Background Adropin is a peptide hormone that promotes nitric oxide (NO) production via activation of endothelial NO synthase (eNOS) in endothelial cells. Its circulating levels are reduced with aging and increased with aerobic exercise training (AT). Using a mouse model, we hypothesized that AT restores aging-associated reductions in arterial and circulating adropin and improves adropin-induced NO-dependent vasorelaxation. Further, we hypothesized these findings would be consistent with data obtained in elderly humans. Methods and Results In the animal study, 50-week-old SAMP1 male mice that underwent 12 weeks of voluntary wheel running, or kept sedentary, were studied. A separate cohort of 25-week-old SAMP1 male mice were used as a mature adult sedentary group. In the human study, 14 healthy elderly subjects completed an 8-week AT program consisting of 45 minutes of cycling 3 days/week. In mice, we show that advanced age is associated with a decline in arterial and circulating levels of adropin along with deterioration of endothelial function, arterial NO production, and adropin-induced vasodilation. All these defects were restored by AT. Moreover, AT-induced increases in arterial adropin were correlated with increases in arterial eNOS phosphorylation and NO production. Consistently with these findings in mice, AT in elderly subjects enhanced circulating adropin levels and these effects were correlated with increases in circulating nitrite/nitrate (NOx) and endothelial function. Conclusions Changes in arterial adropin that occur with age or AT relate to alterations in endothelial function and NO production, supporting the notion that adropin should be considered a therapeutic target for vascular aging. Registration URL: https://www.umin.ac.jp; Unique identifier: UMIN000035520.


Asunto(s)
Envejecimiento/genética , Aorta Torácica/metabolismo , Células Endoteliales/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Óxido Nítrico/farmacología , Resistencia Física/fisiología , Vasodilatación/efectos de los fármacos , Envejecimiento/metabolismo , Animales , Aorta Torácica/citología , Aorta Torácica/efectos de los fármacos , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Regulación de la Expresión Génica , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Masculino , Ratones , Condicionamiento Físico Animal/métodos , ARN/genética , Rigidez Vascular/fisiología
5.
Proc Natl Acad Sci U S A ; 118(21)2021 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-34001623

RESUMEN

Vascular endothelial cells (ECs) sense and respond to hemodynamic forces such as pulsatile shear stress (PS) and oscillatory shear stress (OS). Among the metabolic pathways, glycolysis is differentially regulated by atheroprone OS and atheroprotective PS. Studying the molecular mechanisms by which PS suppresses glycolytic flux at the epigenetic, transcriptomic, and kinomic levels, we have demonstrated that glucokinase regulatory protein (GCKR) was markedly induced by PS in vitro and in vivo, although PS down-regulates other glycolysis enzymes such as hexokinase (HK1). Using next-generation sequencing data, we identified the binding of PS-induced Krüppel-like factor 4 (KLF4), which functions as a pioneer transcription factor, binding to the GCKR promoter to change the chromatin structure for transactivation of GCKR. At the posttranslational level, PS-activated AMP-activated protein kinase (AMPK) phosphorylates GCKR at Ser-481, thereby enhancing the interaction between GCKR and HK1 in ECs. In vivo, the level of phosphorylated GCKR Ser-481 and the interaction between GCKR and HK1 were increased in the thoracic aorta of wild-type AMPKα2+/+ mice in comparison with littermates with EC ablation of AMPKα2 (AMPKα2-/-). In addition, the level of GCKR was elevated in the aortas of mice with a high level of voluntary wheel running. The underlying mechanisms for the PS induction of GCKR involve regulation at the epigenetic level by KLF4 and at the posttranslational level by AMPK.


Asunto(s)
Proteínas Quinasas Activadas por AMP/genética , Aorta Torácica/metabolismo , Epigénesis Genética , Glucólisis/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Aorta Torácica/citología , Fenómenos Biomecánicos , Hexoquinasa/genética , Hexoquinasa/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Factor 4 Similar a Kruppel/genética , Factor 4 Similar a Kruppel/metabolismo , Masculino , Ratones , Ratones Transgénicos , Regiones Promotoras Genéticas , Unión Proteica , Reología , Transcriptoma
6.
Mitochondrion ; 57: 97-107, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33253916

RESUMEN

BACKGROUND: Vascular remodeling plays a pivotal role in regulation of hypoxia-mediated pulmonary and systemic hypertension via the phenotypic modulation of smooth muscle cells (SMCs) of pulmonary and systemic arteries, respectively. Mitochondria serve as putative oxygen (O2) sensors, and consequently, adaptations to hypoxia are mediated via HIF (hypoxia-inducible factors) activation, which impinges on mitochondrial function by suppressing the mitochondrial activity. Therefore, we explored the implication of hypoxia-mediated mitochondrial stress in pulmonary and systemic arterial remodeling. METHODS: The hypoxic (10% O2) effect on human pulmonary artery and aortic SMCs was examined in vitro by cell viability assay, proliferation index, autophagy, and comet assays. Mitochondrial ROS (mtROS), membrane potential (MMP), and mitochondrial morphology were assessed using mitochondrial-selective fluorescent probes. Further, the cell cycle distribution was analyzed by flow cytometry using propidium iodide staining. RESULTS: Our data indicate no significant alterations in cell viability and active proliferation of hypoxic PASMCs; however, an excessive rise in mtROS production and disrupted MMP, accompanied by enhanced DNA damage and reduced autophagy was observed, highlighting the 'apoptosis resistance' phenotype in these cells. Conversely, in hypoxia-treated hASMCs, a modest rise in mtROS levels was associated with reduced DNA damage; followed by upregulated autophagy; increased S-phase DNA content and cell viability, depicting the cytoprotective effect of hypoxia-induced autophagy against mitochondrial damage in hASMCs. CONCLUSION: Our findings suggest that differential impact of mtROS on proliferative capacity may contribute to the variable hypoxic responses in pulmonary and systemic vasculature. Therefore, targeting mtROS may serve as an effective therapeutic strategy to prevent hypoxia-induced hypertension.


Asunto(s)
Aorta Torácica/citología , Mitocondrias/metabolismo , Arteria Pulmonar/citología , Especies Reactivas de Oxígeno/metabolismo , Aorta Torácica/metabolismo , Diferenciación Celular , Hipoxia de la Célula , Línea Celular , Proliferación Celular , Supervivencia Celular , Daño del ADN , Humanos , Potencial de la Membrana Mitocondrial , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Arteria Pulmonar/metabolismo , Remodelación Vascular
7.
J Cell Mol Med ; 24(18): 10542-10550, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32783377

RESUMEN

Calcium deposition in vascular smooth muscle cells (VSMCs) is a form of ectopic ossification in blood vessels. It can result in rigidity of the vasculature and an increase in cardiac events. Here, we report that the microRNA miR-134-5p potentiates inorganic phosphate (Pi)-induced calcium deposition in VSMCs by inhibiting histone deacetylase 5 (HDAC5). Using miRNA microarray analysis of Pi-treated rat VSMCs, we first selected miR-134-5p for further evaluation. Quantitative RT-PCR confirmed that miR-134-5p was increased in Pi-treated A10 cells, a rat VSMC line. Transfection of miR-134-5p mimic potentiated the Pi-induced increase in calcium contents. miR-134-5p increased the amounts of bone runt-related transcription factor 2 (RUNX2) protein and bone morphogenic protein 2 (BMP2) mRNA in the presence of Pi but decreased the expression of osteoprotegerin (OPG). Bioinformatic analysis showed that the HDAC5 3'untranslated region (3'UTR) was one of the targets of miR-134-5p. The luciferase construct containing the 3'UTR of HDAC5 was down-regulated by miR-134-5p mimic in a dose-dependent manner in VSMCs. Overexpression of HDAC5 mitigated the calcium deposition induced by miR-134-5p. Our results suggest that a Pi-induced increase of miR-134-5p may cause vascular calcification through repression of HDAC5.


Asunto(s)
Calcio/metabolismo , Histona Desacetilasas/efectos de los fármacos , MicroARNs/fisiología , Miocitos del Músculo Liso/metabolismo , Calcificación Vascular/etiología , Regiones no Traducidas 3' , Animales , Aorta Torácica/citología , Línea Celular , Simulación por Computador , Subunidad alfa 1 del Factor de Unión al Sitio Principal/biosíntesis , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/inmunología , Regulación hacia Abajo , Regulación de la Expresión Génica , Genes Reporteros , Histona Desacetilasas/biosíntesis , Histona Desacetilasas/genética , MicroARNs/genética , Análisis por Micromatrices , Músculo Liso Vascular/citología , Osteoprotegerina/biosíntesis , Osteoprotegerina/genética , Fosfatos/toxicidad , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes de Fusión/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Transfección , Calcificación Vascular/genética , Calcificación Vascular/metabolismo , Calcificación Vascular/prevención & control
8.
J Surg Res ; 255: 602-611, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32653692

RESUMEN

BACKGROUND: Thoracic aortic aneurysm (TAA) is a severe threat that is characterized by the increased aortic diameter. The dysfunction of vascular smooth muscle cells (VSMCs) contributes to the formation of TAA. Previous research indicated that long noncoding RNAs (lncRNAs) were involved in the development of TAA. This article aimed to explore the role of lncRNA hypoxia-inducible factor-1 alpha-antisense RNA 1 (HIF1A-AS1) and potential action mechanisms in VSMCs. METHODS: The expression of HIF1A-AS1, collagen I, collagen III, microRNA let-7g (let-7g) and apoptotic protease-activating factor 1 (APAF1) was detected by quantitative real-time polymerase chain reaction. Cell proliferation and cell apoptosis were assessed by Cell Counting Kit-8 and flow cytometry assays, respectively. The protein levels of proliferating cell nuclear antigen, Cleaved caspase-3 (Cleaved-cas3), B cell lymphoma/leukemia-2 (Bcl-2), Collagen I, Collagen III, and APAF1 were quantified by Western blot. The relationship between let-7g and HIF1A-AS1 or APAF1 was predicted by the online bioinformatics tool and verified by dual-luciferase reporter assay and RNA pull-down assay. RESULTS: HIF1A-AS1 was upregulated in TAA tissues and was a valuable diagnostic marker of TAA. HIF1A-AS1 overexpression suppressed proliferation, induced apoptosis, and reduced the expression of extracellular matrix proteins in VSMCs. let-7 g was a target of HIF1A-AS1, and its inhibition functioned the same role as HIF1A-AS1 overexpression. APAF1 was a target of let-7g, and its knockdown played the opposite role with HIF1A-AS1 overexpression. The reintroduction of let-7g or APAF1 knockdown reversed the effects of HIF1A-AS1 overexpression in VSMCs. CONCLUSIONS: HIF1A-AS1 regulated the proliferation, apoptosis ,and the activity of extracellular matrix proteins in VSMCs through modulating APAF1 by targeting let-7g, leading to the development of TAA.


Asunto(s)
Aneurisma de la Aorta Torácica/genética , Factor Apoptótico 1 Activador de Proteasas/genética , MicroARNs/metabolismo , Músculo Liso Vascular/patología , ARN Largo no Codificante/metabolismo , Aorta Torácica/citología , Aorta Torácica/patología , Aneurisma de la Aorta Torácica/patología , Apoptosis/efectos de los fármacos , Apoptosis/genética , Factor Apoptótico 1 Activador de Proteasas/metabolismo , Línea Celular , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Biología Computacional , Proteínas de la Matriz Extracelular/genética , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Masculino , MicroARNs/agonistas , MicroARNs/antagonistas & inhibidores , Persona de Mediana Edad , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/patología
9.
Biomed Res Int ; 2020: 4370832, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32461988

RESUMEN

Platelet-derived growth factor-BB (PDGF-BB) can induce the proliferation, migration, and phenotypic modulation of vascular smooth muscle cells (VSMCs). We used patch clamp methods to study the effects of PDGF-BB on inward rectifier K+ channel 2.1 (Kir2.1) channels in rat thoracic aorta VSMCs (RASMCs). PDGF-BB (25 ng/mL) increased Kir2.x currents (-11.81 ± 2.47 pA/pF, P < 0.05 vs. CON, n = 10). Ba2+(50 µM) decreased Kir2.x currents (-2.13 ± 0.23 pA/pF, P < 0.05 vs. CON, n = 10), which were promoted by PDGF-BB (-6.98 ± 1.03 pA/pF). PDGF-BB specifically activates Kir2.1 but not Kir2.2 and Kir2.3 channels in HEK-293 cells. The PDGF-BB-induced stimulation of Kir2.1 currents was blocked by the PDGF-BB receptor ß (PDGF-BBRß) inhibitor AG1295 and was not affected by the PDGF-BBRα inhibitor AG1296. The PDGF-BB-induced stimulation of Kir2.1 currents was blocked by the protein kinase A inhibitor Rp-8-CPT-cAMPs; however, the antagonist of protein kinase B (GSK690693) had marginal effects on current activity. The PDGF-BB-induced stimulation of Kir2.1 currents was enhanced by forskolin, an adenylyl cyclase (AC) activator, and was blocked by the AC inhibitor SQ22536. We conclude that PDGF-BB increases Kir2.1 currents via PDGF-BBRß through activation of cAMP-PKA signaling in RASMCs.


Asunto(s)
Becaplermina/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Músculo Liso Vascular/citología , Canales de Potasio de Rectificación Interna , Animales , Aorta Torácica/citología , Células Cultivadas , Colforsina/farmacología , Células HEK293 , Humanos , Masculino , Canales de Potasio de Rectificación Interna/efectos de los fármacos , Canales de Potasio de Rectificación Interna/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos
10.
Eur J Pharmacol ; 880: 173165, 2020 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-32423869

RESUMEN

Vascular calcification is a highly regulated process similar to osteogenesis involving phenotypic change of vascular smooth muscle cells (VSMCs). 25-Hydroxycholesterol (25-HC), one of oxysterols synthesized by the enzyme cholesterol 25-hydroxylase, has been shown to promote bovine calcifying vascular cells (CVC) calcification. However, whether and how 25-HC regulates vascular calcification are not completely understood. In this study, in vitro and ex vivo models of vascular calcification were used to determine whether 25-HC regulates vascular calcification. Alizarin red staining and calcium content assay showed that 25-HC treatment promoted calcification of rat and human VSMCs in a dose-dependent manner. Similarly, ex vivo study further confirmed that 25-HC accelerated calcification of rat aortic rings. In addition, western blot analysis showed that 25-HC significantly up-regulated the expression of endoplasmic reticulum stress (ERS) signaling molecules including ATF4 and CHOP in VSMCs and flow cytometry analysis revealed that 25-HC increased apoptosis of VSMCs. Moreover, knockdown of CHOP by siRNA blocked 25-HC-induced mineral deposition in VSMCs. Collectively, this study for the first time demonstrates that 25-HC promotes vascular calcification via ATF4/CHOP signaling using in vitro and ex vivo models, suggesting that ERS is involved in the regulation of 25-HC-induced vascular calcification.


Asunto(s)
Estrés del Retículo Endoplásmico/efectos de los fármacos , Hidroxicolesteroles/farmacología , Miocitos del Músculo Liso/efectos de los fármacos , Calcificación Vascular/inducido químicamente , Factor de Transcripción Activador 4/metabolismo , Animales , Aorta Torácica/citología , Apoptosis/efectos de los fármacos , Calcio/metabolismo , Células Cultivadas , Humanos , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , ARN Interferente Pequeño/genética , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Factor de Transcripción CHOP/genética , Calcificación Vascular/metabolismo
11.
Eur Rev Med Pharmacol Sci ; 24(4): 1609-1615, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32141527

RESUMEN

OBJECTIVE: The aim of this study was to explore the influence of micro ribonucleic acid (miR)-26b on gestational diabetes mellitus in rats via the phosphatidylinositol 3-hydroxy kinase/protein kinase B (PI3K/Akt) signaling pathway. MATERIALS AND METHODS: A total of 60 healthy pregnant female rats were randomly divided into three groups, including group A (normal group), group B (model group), and group C (model + miR-26b group). The differences in fasting blood glucose (FBG), C-reactive protein (CRP), and phosphatidylinositol 3-hydroxy kinase/protein kinase B (PI3K/AKT) among the three groups were analyzed via serum CRP test, morphological observation, quantitative Reverse Transcription-Polymerase Chain Reaction (qRT-PCR), and Western blotting, respectively. RESULTS: The levels of FBG ad CRP were significantly up-regulated in group B when compared with group A (p<0.01). Meanwhile, they increased significantly in group C when compared with group B (p<0.01). Rats in group A exhibited smooth and flat thoracic aortic intimas, as well as neatly arranged smooth muscle cells at the media layer. However, rats in group B showed fractured intimas with enlarged junction gaps, as well as necrotic and detached endothelial cells. Compared with group B, group C exhibited extremely poorly arranged cells at all the layers, rough and rugged intimas, larger areas of necrotic and detached endothelial cells, and markedly worsened lesions. QRT-PCR results indicated that the expression of phosphorylated-PI3K (p-PI3K) was significantly lower in group B than that of group A (p=0.04). Meanwhile, it was markedly lower in group C than that in group B (p=0.04). The expression of p-Akt was remarkably lower in group B than group A (p=0.04), which was also significantly lower in group C than group B (p=0.04). Compared with group A, the expressions of p-PI3K and p-Akt in the thoracic aorta of group B were evidently down-regulated (p<0.01). Furthermore, they decreased markedly in group C when compared with group B (p<0.01). CONCLUSIONS: MiR-26b accelerates the progression of gestational diabetes by inhibiting the PI3K/Akt signaling pathway.


Asunto(s)
Diabetes Gestacional/genética , MicroARNs , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Aorta Torácica/citología , Glucemia , Diabetes Gestacional/metabolismo , Diabetes Gestacional/patología , Células Endoteliales/patología , Femenino , Fosfatidilinositol 3-Quinasas/genética , Embarazo , Proteínas Proto-Oncogénicas c-akt/genética , Ratas , Transducción de Señal
12.
Eur J Vasc Endovasc Surg ; 59(6): 1000-1010, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31879145

RESUMEN

OBJECTIVE: Thoracic aortic dissection (TAD) has a high mortality rate. Intermittent hypoxia (IH) triggers both harmful and beneficial effects in numerous physiological systems. The effects of IH on TAD development were explored in a mouse model. METHODS: ß-Aminopropionitrile monofumarate (BAPN) was used to induce TAD in C57BL/6 mice. Three week old male mice were treated with 1 g/kg/day BAPN in drinking water for four weeks and simultaneously subjected to IH (n = 30) (21%-5% O2, 90 s/cycle, 10 h/day, IH + BAPN group) or normoxia (n = 30) (21% O2, 24 h/day, BAPN group). Human VSMCs (HUASMCs) exposed to IH (30 min, 5% O2)/re-oxygenation (30 min, 21% O2) cycles with a maximum of 60 min/cycle to detect the effect of IH on HIF-1α and LOX via HIF-1α-siRNA. RESULTS: It was found that BAPN administration significantly increased the lumen size and wall thickness of aortas compared with the normal group, but was significantly reversed by IH exposure. Additionally, IH exposure significantly increased the survival rate of BAPN induced TAD (70% vs. 40%). Furthermore, IH exposure reduced BAPN induced elastin breaks and apoptosis of vascular smooth muscle cells. IH exposure also reversed BAPN induced upregulation of inflammation and extracellular matrix (ECM) degradation. Real time polymerase chain reaction (RT-PCR) confirmed that IH inhibited inflammation and ECM degradation related genes interleukin (IL)-1ß, IL-6, cathepsin S (Cat S), and matrix metalloproteinase 9 (MMP-9), but upregulated the ECM synthesis related genes lysyl oxidase (LOX) and collagen type I alpha2 (Col1a2) compared with the BAPN group. In vitro results suggest that IH promotes the expression of LOX via HIF-1α. CONCLUSION: The results suggest that IH alleviates BAPN induced TAD in C57BL/6 mice.


Asunto(s)
Aorta Torácica/fisiopatología , Aneurisma de la Aorta Torácica/terapia , Disección Aórtica/terapia , Hipoxia/fisiopatología , Poscondicionamiento Isquémico/métodos , Aminopropionitrilo/análogos & derivados , Aminopropionitrilo/toxicidad , Disección Aórtica/etiología , Animales , Aorta Torácica/citología , Aorta Torácica/efectos de los fármacos , Aneurisma de la Aorta Torácica/inducido químicamente , Aneurisma de la Aorta Torácica/complicaciones , Modelos Animales de Enfermedad , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/patología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL
13.
Life Sci ; 241: 117098, 2020 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-31794773

RESUMEN

INTRODUCTION: Sepsis survivors are at higher risk for cardiovascular events. Lipopolysaccharide (LPS) activates Toll-like receptor 4 (TLR4) in sepsis. Activation of TLR4 modulates vascular smooth muscle cells (VSMCs) phenotype and contributes to cardiovascular changes after sepsis. AIM: Investigate changes in VSMCs phenotype caused by LPS-induced TLR4 activation. METHODS: Rat VSMCs were incubated with LPS. Two incubation conditions were used in cell contraction and migration assays: acute stimulation - LPS stimulus was initiated at the beginning of the assay and maintained throughout; and preconditioning - LPS stimulation was applied prior to the assay then discontinued. Nitric oxide (NO) production, mRNA expression of cytokines and phenotype markers, and interleukin (IL)-6 production were evaluated. KEY FINDINGS: LPS increased gene expression of IL-1ß, IL-6, TNFα and MCP-1 (p < .001), of secretory phenotype markers collagen and vimentin (p < .0479) and of the contractile marker smooth muscle 22α (SM22α) (p = .0067). LPS exposure increased IL-6 secretion after 24 and 48 h (p < .0001), and NO at 8 and 24 h (p < .0249) via inducible nitric oxide synthase (iNOS), as demonstrated by a decrease in NO after incubation with aminoguanidine. Acute stimulation with LPS reduced migration and contraction in a NO-dependent manner, while preconditioning with LPS increased both in an IL-6-dependent manner. SIGNIFICANCE: LPS affects VSMCs by modulating their secretory, contractile and migratory phenotypes. LPS acute stimulation of VSMCs promoted a NO-dependent reduction in migration and contraction, while preconditioning with LPS promoted IL-6-dependent increases in migration and contraction, evidencing that VSMCs can present phenotype modifications that persist after sepsis, thereby contributing to postsepsis cardiovascular events.


Asunto(s)
Lipopolisacáridos/toxicidad , Contracción Muscular/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Sepsis/fisiopatología , Animales , Aorta Torácica/citología , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Regulación de la Expresión Génica/efectos de los fármacos , Interleucina-6/genética , Interleucina-6/metabolismo , Masculino , Contracción Muscular/fisiología , Músculo Liso Vascular/patología , Músculo Liso Vascular/fisiología , Óxido Nítrico , Fenotipo , Ratas Wistar
14.
Eur J Pharmacol ; 865: 172772, 2019 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-31697934

RESUMEN

Vascular calcification increases the risk of morbidity and mortality in patients with cardiovascular diseases, chronic kidney diseases, and diabetes. However, viable therapeutic methods to target vascular calcification are limited. Aloe-emodin (AE), an anthraquinone is a natural compound found in the leaves of Aloe-vera. In this study, we investigated the underlying mechanism of AE in the calcification of vascular smooth muscle cells (VSMCs) and murine thoracic aorta. We demonstrate that AE repressed not only the phenotypes of Ca2+ induced calcification but also level of calcium in VSMCs. AE has no effect on cell viability in VSMC cells. Alizarin red, von Kossa stainings and calcium quantification showed that Ca2+ induced vascular calcification is significantly decreased by AE in a concentration-dependent manner. In contrast, AE attenuated Ca2+ induced calcification through inhibiting osteoblast differentiation genes such as SMAD4, collagen 1α, osteopontin (OPN), Runt-related transcription factor (RUNX-2) and Osterix. AE also suppressed Ca2+ induced osteoblast-related protein expression including collagen 1α, bone morphogenic protein 2 (BMP-2), RUNX-2 and smooth muscle actin (SMA). Furthermore, Alizarin red, von Kossa stainings and calcium quantification showed that AE significantly inhibited the calcification of ex vivo ring formation in murine thoracic aorta, and markedly inhibited vitamin D3 induced medial aorta calcification in vivo. Taken together, our findings suggest that AE may have therapeutic potential for the prevention of vascular calcification program.


Asunto(s)
Antraquinonas/farmacología , Calcificación Fisiológica/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Animales , Aorta Torácica/citología , Proteína Morfogenética Ósea 2/metabolismo , Calcio/metabolismo , Masculino , Ratones Endogámicos ICR , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/fisiología , Osteogénesis/efectos de los fármacos , Proteína Smad4/genética
15.
Biochem Biophys Res Commun ; 517(3): 470-476, 2019 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-31376939

RESUMEN

Osteogenic differentiation of VSMC is one of the main causes of diabetic vascular calcification, and AGEs accumulation accelerates the calcification of VSMCs in diabetic patients. Autophagy has also been found to play an important role in the process of vascular calcification. However, the potential link between AGEs, autophagy and vascular calcification is still unclear and was investigated in this study. Primary VSMCs were isolated from the thoracic aorta of Sprague Dawley rats and cultured with AGEs-BSA to induce osteogenic differentiation. VSMCs calcification was evaluated by measuring the calcium content, RUNX2 protein levels, and by Alizarin red S staining. We demonstrated that treatment of VSMCs with AGE-BSA increased the expression of HIF-1α and PDK4. AGE-BSA treatment increased LC3-II and decreased p62 protein levels. AGE-BSA exposure enhanced autophagic flux determined by mRFP-GFP-LC3 adenovirus, induced co-localization of LC3-II and LAMP-1, and increased the number of autophagasome under TEM. HIF-1α/PDK4 pathway was activated during AGEs-induced autophagy of VSMCs. In addition, autophagy played a protective role during AGE-induced calcification of VSMCs. In conclusion, AGEs enhance autophagy via the HIF-1α/PDK4 signaling pathway, and autophagy helps attenuate AGE-induced calcification of VSMCs.


Asunto(s)
Productos Finales de Glicación Avanzada/farmacología , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Miocitos del Músculo Liso/efectos de los fármacos , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora/genética , Albúmina Sérica Bovina/farmacología , Calcificación Vascular/genética , Animales , Aorta Torácica/citología , Aorta Torácica/efectos de los fármacos , Aorta Torácica/metabolismo , Autofagia/efectos de los fármacos , Autofagia/genética , Diferenciación Celular/efectos de los fármacos , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Regulación de la Expresión Génica , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Proteínas de Membrana de los Lisosomas/genética , Proteínas de Membrana de los Lisosomas/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , 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 , Osteogénesis/efectos de los fármacos , Osteogénesis/genética , Cultivo Primario de Células , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora/metabolismo , Ratas , Ratas Sprague-Dawley , Proteína Sequestosoma-1/genética , Proteína Sequestosoma-1/metabolismo , Transducción de Señal , Calcificación Vascular/inducido químicamente , Calcificación Vascular/metabolismo , Calcificación Vascular/patología
16.
Arterioscler Thromb Vasc Biol ; 39(8): 1629-1644, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31189430

RESUMEN

OBJECTIVE: Periaortic arch adipose tissue (PAAT) plays critical roles in regulating vascular homeostasis; however, its anatomic features, developmental processes, and origins remain unclear. Approach and Results: Anatomic analysis and genetic lineage tracing of Wnt1 (wingless-type MMTV [mouse mammary tumor virus] integration site family member 1)-Cre+;Rosa26RFP/+ mice, Myf5 (myogenic factor 5)-Cre+;Rosa26RFP/+ mice, and SM22α-Cre+;Rosa26RFP/+ mice are performed, and the results show that PAAT has unique anatomic features, and the developmental processes of PAAT are independent of the others periaortic adipose tissues. PAAT adipocytes are mainly derived from neural crest cells (NCCs) rather than from Myf5+ progenitors. Most PAAT adipocyte progenitors expressed SM22α+ (smooth muscle protein 22-alpha) during development. Using Wnt1-Cre+;PPARγflox/flox mice, we found that knockout of PPAR (peroxisome proliferator-activated receptor)-γ in NCCs results in PAAT developmental delay and dysplasia, further confirming that NCCs contribute to PAAT formation. And we further indicated PAAT dysplasia aggravates Ang II (angiotensin II)-induced inflammation and remodeling of the common carotid artery close to aorta arch. We also found that NCCs can be differentiated into both brown and white adipocytes in vivo and in vitro. RNA sequencing results suggested NCC-derived adipose tissue displays a distinct transcriptional profile compared with the non-NCC-derived adipose tissue in PAAT. CONCLUSIONS: PAAT has distinctive anatomic features and developmental processes. Most PAAT adipocytes are originated from NCCs which derive from ectoderm. NCCs are progenitors not only of white adipocytes but also of brown adipocytes. This study indicates that the PAAT is derived from multiple cell lineages, the adipocytes derived from different origins have distinct transcriptional profiles, and PAAT plays a critical role in Ang II-induced common carotid artery inflammation and remodeling.Visual OvervieW: An online visual overview is available for this article.


Asunto(s)
Adipocitos Marrones/citología , Adipogénesis , Tejido Adiposo/fisiología , Cresta Neural/citología , Tejido Adiposo/citología , Angiotensina II/farmacología , Animales , Aorta Torácica/citología , Arteria Carótida Común/citología , Diferenciación Celular , Linaje de la Célula , Células Cultivadas , Perfilación de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , PPAR gamma/fisiología , Proteína Wnt1/fisiología
17.
Biomolecules ; 9(6)2019 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-31212721

RESUMEN

Despite pharmacotherapeutic advances, cardiovascular disease (CVD) remains the primary cause of global mortality. Alternative approaches, such as herbal medicine, continue to be sought to reduce this burden. Origanum majorana is recognized for many medicinal values, yet its vasculoprotective effects remain poorly investigated. Here, we subjected rat thoracic aortae to increasing doses of an ethanolic extract of Origanummajorana (OME). OME induced relaxation in a dose-dependent manner in endothelium-intact rings. This relaxation was significantly blunted in denuded rings. N(ω)-nitro-l-arginine methyl ester (L-NAME) or 1H-[1,2,4]oxadiazolo[4,3,-a]quinoxalin-1-one (ODQ) significantly reduced the OME-induced vasorelaxation. Cyclic guanosine monophosphate (cGMP) levels were also increased by OME. Moreover, wortmannin or LY294002 significantly reduced OME-induced vasorelaxation. Blockers of ATP-sensitive or Ca2+-activated potassium channels such as glibenclamide or tetraethylamonium (TEA), respectively, did not significantly affect OME-induced relaxation. Similarly, verapamil, a Ca2+ channel blocker, indomethacin, a non-selective cyclooxygenase inhibitor, and pyrilamine, a H1 histamine receptor blocker, did not significantly modulate the observed relaxation. Taken together, our results show that OME induces vasorelaxation via an endothelium-dependent mechanism involving the phosphoinositide 3-kinase (PI3-K)/ endothelial nitric oxide (NO) synthase (eNOS)/cGMP pathway. Our findings further support the medicinal value of marjoram and provide a basis for its beneficial intake. Although consuming marjoram may have an antihypertensive effect, further studies are needed to better determine its effects in different vascular beds.


Asunto(s)
Aorta Torácica/efectos de los fármacos , Aorta Torácica/fisiología , GMP Cíclico/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Origanum/química , Fosfatidilinositol 3-Quinasa/metabolismo , Extractos Vegetales/farmacología , Animales , Aorta Torácica/citología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Masculino , Norepinefrina/farmacología , Bloqueadores de los Canales de Potasio/farmacología , Prostaglandina-Endoperóxido Sintasas/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Vasoconstricción/efectos de los fármacos , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología
18.
Mol Pharmacol ; 95(6): 629-637, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30988014

RESUMEN

Nitric oxide (NO) stimulates soluble guanylyl cyclase (sGC) activity, leading to elevated intracellular cyclic guanosine 3',5'-monophosphate (cGMP) and subsequent vascular smooth muscle relaxation. It is known that downregulation of sGC expression attenuates vascular dilation and contributes to the pathogenesis of cardiovascular disease. However, it is not well understood how sGC transcription is regulated. Here, we demonstrate that pharmacological inhibition of Forkhead box subclass O (FoxO) transcription factors using the small-molecule inhibitor AS1842856 significantly blunts sGC α and ß mRNA expression by more than 90%. These effects are concentration-dependent and concomitant with greater than 90% reduced expression of the known FoxO transcriptional targets, glucose-6-phosphatase and growth arrest and DNA damage protein 45 α (Gadd45α). Similarly, sGC α and sGC ß protein expression showed a concentration-dependent downregulation. Consistent with the loss of sGC α and ß mRNA and protein expression, pretreatment of vascular smooth muscle cells with the FoxO inhibitor decreased sGC activity measured by cGMP production following stimulation with an NO donor. To determine if FoxO inhibition resulted in a functional impairment in vascular relaxation, we cultured mouse thoracic aortas with the FoxO inhibitor and conducted ex vivo two-pin myography studies. Results showed that aortas have significantly blunted sodium nitroprusside-induced (NO-dependent) vasorelaxation and a 42% decrease in sGC expression after 48-hour FoxO inhibitor treatment. Taken together, these data are the first to identify that FoxO transcription factor activity is necessary for sGC expression and NO-dependent relaxation.


Asunto(s)
Proteínas de Ciclo Celular/genética , Músculo Liso Vascular/citología , Quinolonas/farmacología , Guanilil Ciclasa Soluble/genética , Animales , Aorta Torácica/citología , Aorta Torácica/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Factores de Transcripción Forkhead/antagonistas & inhibidores , Regulación de la Expresión Génica/efectos de los fármacos , Ratones , Relajación Muscular/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Óxido Nítrico/metabolismo , Ratas , Guanilil Ciclasa Soluble/deficiencia , Guanilil Ciclasa Soluble/metabolismo
19.
Circ Res ; 123(6): 686-699, 2018 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-30355234

RESUMEN

RATIONALE: Regeneration of lost cardiomyocytes is a fundamental unresolved problem leading to heart failure. Despite several strategies developed from intensive studies performed in the past decades, endogenous regeneration of heart tissue is still limited and presents a big challenge that needs to be overcome to serve as a successful therapeutic option for myocardial infarction. OBJECTIVE: One of the essential prerequisites for cardiac regeneration is the identification of endogenous cardiomyocyte progenitors and their niche that can be targeted by new therapeutic approaches. In this context, we hypothesized that the vascular wall, which was shown to harbor different types of stem and progenitor cells, might serve as a source for cardiac progenitors. METHODS AND RESULTS: We describe generation of spontaneously beating mouse aortic wall-derived cardiomyocytes without any genetic manipulation. Using aortic wall-derived cells (AoCs) of WT (wild type), αMHC (α-myosin heavy chain), and Flk1 (fetal liver kinase 1)-reporter mice and magnetic bead-associated cell sorting sorting of Flk1+ AoCs from GFP (green fluorescent protein) mice, we identified Flk1+CD (cluster of differentiation) 34+Sca-1 (stem cell antigen-1)-CD44- AoCs as the population that gives rise to aortic wall-derived cardiomyocytes. This AoC subpopulation delivered also endothelial cells and macrophages with a particular accumulation within the aortic wall-derived cardiomyocyte containing colonies. In vivo, cardiomyocyte differentiation capacity was studied by implantation of fluorescently labeled AoCs into chick embryonic heart. These cells acquired cardiomyocyte-like phenotype as shown by αSRA (α-sarcomeric actinin) expression. Furthermore, coronary adventitial Flk1+ and CD34+ cells proliferated, migrated into the myocardium after mouse myocardial infarction, and expressed Isl-1+ (insulin gene enhancer protein-1) indicative of cardiovascular progenitor potential. CONCLUSIONS: Our data suggest Flk1+CD34+ vascular adventitia-resident stem cells, including those of coronary adventitia, as a novel endogenous source for generating cardiomyocytes. This process is essentially supported by endothelial cells and macrophages. In summary, the therapeutic manipulation of coronary adventitia-resident cardiac stem and their supportive cells may open new avenues for promoting cardiac regeneration and repair after myocardial infarction and for preventing heart failure.


Asunto(s)
Adventicia/citología , Aorta Torácica/citología , Diferenciación Celular , Proliferación Celular , Miocitos Cardíacos/fisiología , Células Madre/fisiología , Animales , Antígenos CD34/metabolismo , Antígenos Ly/metabolismo , Células Cultivadas , Embrión de Pollo , Modelos Animales de Enfermedad , Femenino , Genes Reporteros , Separación Inmunomagnética , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Infarto del Miocardio/cirugía , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/trasplante , Cadenas Pesadas de Miosina/genética , Fenotipo , Regeneración , Trasplante de Células Madre , Células Madre/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Miosinas Ventriculares/genética
20.
Mol Med Rep ; 18(5): 4709-4715, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30221709

RESUMEN

To detect the structure­activity associations of farrerol derivatives, the relaxation activity of farrerol derivatives was observed in isolated aortic rings pre­contracted using phenylephrine in Sprague­Dawley rats. All compounds tested in the present study produced a relaxation effect, which was significantly affected by the molecular structure. Using a collagen gel contraction assay, the present study further evaluated the inhibitiory effect of farrerol derivatives in a decreased collagen gel area, induced by Angiotensin II. The results indicated that farrerol derivatives could inhibit collagen contraction, and that the inhibitory effect was associated with the molecular structure of the compounds. Furthermore, the inhibitory strength of the different compounds was consistent with the results of vascular tension detection. The activity of the farrerol derivatives was closely associated with the molecular structure. The analysis indicated that an electron­withdrawing substituent in the ortho position of the phenyl group (ring B) was crucial in order to observe improved vasorelaxation activity, whereas a hydroxyl or methoxy group was unfavorable. A para electron­donating group was oberved to increase compound activity. In addition, when the B ring was heterocycle rather than a phenyl ring, the vasorelaxation ability was weakened.


Asunto(s)
Cromonas/farmacología , Relación Estructura-Actividad , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología , Angiotensina II/farmacología , Animales , Aorta Torácica/citología , Aorta Torácica/efectos de los fármacos , Cromonas/química , Técnicas de Cultivo de Órganos , Fenilefrina/farmacología , Ratas , Vasodilatadores/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA