Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 776
Filter
1.
Cardiovasc Res ; 118(1): 254-266, 2022 01 07.
Article in English | MEDLINE | ID: mdl-33483748

ABSTRACT

AIMS: Rheumatoid arthritis (RA) is a chronic inflammatory disease affecting joints and blood vessels. Despite low levels of low-density lipoprotein cholesterol (LDL-C), RA patients exhibit endothelial dysfunction and are at increased risk of death from cardiovascular complications, but the molecular mechanism of action is unknown. We aimed in the present study to identify the molecular mechanism of endothelial dysfunction in a mouse model of RA and in patients with RA. METHODS AND RESULTS: Endothelium-dependent relaxations to acetylcholine were reduced in aortae of two tumour necrosis factor alpha (TNFα) transgenic mouse lines with either mild (Tg3647) or severe (Tg197) forms of RA in a time- and severity-dependent fashion as assessed by organ chamber myograph. In Tg197, TNFα plasma levels were associated with severe endothelial dysfunction. LOX-1 receptor was markedly up-regulated leading to increased vascular oxLDL uptake and NFκB-mediated enhanced Arg2 expression via direct binding to its promoter resulting in reduced NO bioavailability and vascular cGMP levels as shown by ELISA and chromatin immunoprecipitation. Anti-TNFα treatment with infliximab normalized endothelial function together with LOX-1 and Arg2 serum levels in mice. In RA patients, soluble LOX-1 serum levels were also markedly increased and closely related to serum levels of C-reactive protein. Similarly, ARG2 serum levels were increased. Similarly, anti-TNFα treatment restored LOX-1 and ARG2 serum levels in RA patients. CONCLUSIONS: Increased TNFα levels not only contribute to RA, but also to endothelial dysfunction by increasing vascular oxLDL content and activation of the LOX-1/NFκB/Arg2 pathway leading to reduced NO bioavailability and decreased cGMP levels. Anti-TNFα treatment improved both articular symptoms and endothelial function by reducing LOX-1, vascular oxLDL, and Arg2 levels.


Subject(s)
Aorta, Thoracic/drug effects , Arginase/metabolism , Arthritis, Rheumatoid/drug therapy , Endothelial Cells/drug effects , Endothelium, Vascular/drug effects , Scavenger Receptors, Class E/metabolism , Tumor Necrosis Factor Inhibitors/therapeutic use , Tumor Necrosis Factor-alpha/metabolism , Vasodilation/drug effects , Adult , Animals , Animals, Genetically Modified , Aorta, Thoracic/enzymology , Aorta, Thoracic/immunology , Aorta, Thoracic/physiopathology , Arginase/genetics , Arthritis, Rheumatoid/enzymology , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/physiopathology , Case-Control Studies , Disease Models, Animal , Endothelial Cells/enzymology , Endothelial Cells/immunology , Endothelium, Vascular/enzymology , Endothelium, Vascular/immunology , Endothelium, Vascular/physiopathology , Female , Humans , Lipoproteins, LDL/metabolism , Male , Mice, Inbred C57BL , Mice, Inbred CBA , Middle Aged , NF-kappa B/metabolism , Scavenger Receptors, Class E/genetics , Signal Transduction , Tumor Necrosis Factor-alpha/genetics
3.
J Am Heart Assoc ; 10(16): e020554, 2021 08 17.
Article in English | MEDLINE | ID: mdl-34350769

ABSTRACT

Background Adventitial remodeling is a pathological hallmark of hypertension that results in target organ damage. Activated adventitial fibroblasts have emerged as critical regulators in this process, but the precise mechanism remains unclear. Methods and Results Interleukin 11 (IL-11) knockout and wild-type mice were subjected to angiotensin II (Ang II) infusion to establish models of hypertension-associated vascular remodeling. IL-11 mRNA and protein were increased especially in the adventitia in response to Ang II. Compared with wild-type mice, Ang II-treated IL-11 knockout mice showed amelioration of vascular hypertrophy, adventitial fibrosis, macrophage infiltration, and inflammatory factor expression. Recombination mouse IL-11 exacerbated adventitial fibrosis in Ang II-infused wild-type mice. Interestingly, IL-11 neutralizing antibody attenuated adventitial fibrosis, macrophage infiltration, and inflammatory factor expression after Ang II infusion for 7 days. Mechanistically, in primary cultured adventitial fibroblasts, Krüppel-like factor 15 negatively regulated Ang II-induced IL-11 expression. Ang II increased extracellular signal-regulated kinases 1 and 2 activation, especially in adventitia, and caused biphasic extracellular signal-regulated kinases 1 and 2 activation in adventitial fibroblasts. A rapid and early activation increased IL-11 production through decreasing Krüppel-like factor 15 expression, which, in turn, induced the second extracellular signal-regulated kinases 1 and 2 activation, resulting in posttranscriptional profibrotic gene expression. Conclusions These results demonstrate that extracellular signal-regulated kinases 1 and 2 activation is important for Krüppel-like factor 15-mediated IL-11 expression in adventitial fibroblasts to promote adventitial remodeling in Ang II-induced hypertension. Therefore, targeting the Krüppel-like factor 15/IL-11 axis might serve as a new therapeutic strategy for vascular diseases.


Subject(s)
Adventitia/enzymology , Aorta, Thoracic/enzymology , Fibroblasts/enzymology , Hypertension/enzymology , Interleukin-11/metabolism , Kruppel-Like Transcription Factors/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Vascular Remodeling , Adventitia/pathology , Angiotensin II , Animals , Aorta, Thoracic/pathology , Disease Models, Animal , Fibroblasts/pathology , Fibrosis , HEK293 Cells , Humans , Hypertension/chemically induced , Hypertension/genetics , Hypertension/pathology , Inflammation Mediators/metabolism , Interleukin-11/genetics , Kruppel-Like Transcription Factors/genetics , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Rats, Sprague-Dawley , Signal Transduction
4.
J Pharmacol Exp Ther ; 379(2): 117-124, 2021 11.
Article in English | MEDLINE | ID: mdl-34389653

ABSTRACT

Streptolysin O (SLO) is produced by common hemolytic streptococci that cause a wide range of diseases from pharyngitis to life-threatening necrotizing fasciitis and toxic shock syndrome. Although the importance of SLO in invasive hemolytic streptococcus infection has been well demonstrated, the role of circulating SLO in noninvasive infection remains unclear. The aim of this study was to characterize the pharmacological effect of SLO on vascular functions, focusing on cellular signaling pathways. In control Wistar rats, SLO treatment (1-1000 ng/ml) impaired acetylcholine-induced endothelial-dependent relaxation in the aorta and second-order mesenteric artery in a dose-dependent manner without any effects on sodium nitroprusside-induced endothelium-independent relaxation or agonist-induced contractions. SLO also increased phosphorylation of the endothelial NO synthase (eNOS) inhibitory site at Thr495 in the aorta. Pharmacological analysis indicated that either endothelial dysfunction or eNOS phosphorylation was mediated by protein kinase Cß (PKCß), but not by the p38 mitogen-activated protein kinase pathway. Consistent with this, SLO increased phosphorylation levels of protein kinase C substrates in the aorta. In vivo study of control Wistar rats indicated that intravenous administration of SLO did not change basal blood pressure but significantly counteracted the acetylcholine-induced decrease in blood pressure. Interestingly, plasma anti-SLO IgG levels were significantly higher in 10- to 15-week-old spontaneously hypertensive rats compared with age-matched control rats (P < 0.05). These findings demonstrated that SLO causes vascular endothelial dysfunction, which is mediated by PKCß-induced phosphorylation of the eNOS inhibitory site. SIGNIFICANCE STATEMENT: This study showed for the first time that in vitro exposure of vascular tissues to SLO impairs endothelial function, an effect that is mediated by protein kinase C ß-induced phosphorylation of the endothelial NO synthase inhibitory site. Intravenous administration of SLO in control and hypertensive rats blunted the acetylcholine-induced decrease in blood pressure, providing evidence for a possible role of SLO in dysregulation of blood pressure.


Subject(s)
Endothelium, Vascular/drug effects , Endothelium, Vascular/enzymology , Protein Kinase C beta/metabolism , Streptolysins/toxicity , Vasoconstriction/drug effects , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/enzymology , Bacterial Proteins/toxicity , Dose-Response Relationship, Drug , Male , Mesenteric Arteries/drug effects , Mesenteric Arteries/enzymology , Organ Culture Techniques , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Rats, Wistar , Vasoconstriction/physiology
5.
Arterioscler Thromb Vasc Biol ; 41(9): 2483-2493, 2021 09.
Article in English | MEDLINE | ID: mdl-34320838

ABSTRACT

Objective: Despite considerable research, the goal of finding nonsurgical remedies against thoracic aortic aneurysm and acute aortic dissection remains elusive. We sought to identify a novel aortic PK (protein kinase) that can be pharmacologically targeted to mitigate aneurysmal disease in a well-established mouse model of early-onset progressively severe Marfan syndrome (MFS). Approach and Results: Computational analyses of transcriptomic data derived from the ascending aorta of MFS mice predicted a probable association between thoracic aortic aneurysm and acute aortic dissection development and the multifunctional, stress-activated HIPK2 (homeodomain-interacting protein kinase 2). Consistent with this prediction, Hipk2 gene inactivation significantly extended the survival of MFS mice by slowing aneurysm growth and delaying transmural rupture. HIPK2 also ranked among the top predicted PKs in computational analyses of DEGs (differentially expressed genes) in the dilated aorta of 3 MFS patients, which strengthened the clinical relevance of the experimental finding. Additional in silico analyses of the human and mouse data sets identified the TGF (transforming growth factor)-ß/Smad3 signaling pathway as a potential target of HIPK2 in the MFS aorta. Chronic treatment of MFS mice with an allosteric inhibitor of HIPK2-mediated stimulation of Smad3 signaling validated this prediction by mitigating thoracic aortic aneurysm and acute aortic dissection pathology and partially improving aortic material stiffness. Conclusions: HIPK2 is a previously unrecognized determinant of aneurysmal disease and an attractive new target for antithoracic aortic aneurysm and acute aortic dissection multidrug therapy.


Subject(s)
Aorta, Thoracic/drug effects , Aortic Aneurysm, Thoracic/prevention & control , Aortic Dissection/prevention & control , Fibrillin-1/genetics , Marfan Syndrome/genetics , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Vascular Remodeling/drug effects , Adult , Aortic Dissection/enzymology , Aortic Dissection/genetics , Aortic Dissection/pathology , Animals , Aorta, Thoracic/enzymology , Aorta, Thoracic/pathology , Aortic Aneurysm, Thoracic/enzymology , Aortic Aneurysm, Thoracic/genetics , Aortic Aneurysm, Thoracic/pathology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Dilatation, Pathologic , Disease Models, Animal , Disease Progression , Humans , Male , Marfan Syndrome/complications , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Severity of Illness Index , Signal Transduction , Smad3 Protein/metabolism
6.
Am J Physiol Regul Integr Comp Physiol ; 321(2): R228-R237, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34189960

ABSTRACT

The mechanistic target of rapamycin complex 1 (mTORC1) signaling complex is emerging as a critical regulator of cardiovascular function with alterations in this pathway implicated in cardiovascular diseases. In this study, we used animal models and human tissues to examine the role of vascular mTORC1 signaling in the endothelial dysfunction associated with obesity. In mice, obesity induced by high-fat/high-sucrose diet feeding for ∼2 mo resulted in aortic endothelial dysfunction without appreciable changes in vascular mTORC1 signaling. On the other hand, chronic high-fat diet feeding (45% or 60% kcal: ∼9 mo) in mice resulted in endothelial dysfunction associated with elevated vascular mTORC1 signaling. Endothelial cells and visceral adipose vessels isolated from obese humans display a trend toward elevated mTORC1 signaling. Surprisingly, genetic disruption of endothelial mTORC1 signaling through constitutive or tamoxifen inducible deletion of endothelial Raptor (critical subunit of mTORC1) did not prevent or rescue the endothelial dysfunction associated with high-fat diet feeding in mice. Endothelial mTORC1 deficiency also failed to reverse the endothelial dysfunction evoked by a high-fat/high-sucrose diet in mice. Taken together, these data show increased vascular mTORC1 signaling in obesity, but this vascular mTORC1 activation appears not to be required for the development of endothelial impairment in obesity.


Subject(s)
Endothelium, Vascular/enzymology , Mechanistic Target of Rapamycin Complex 1/deficiency , Obesity/prevention & control , Subcutaneous Fat/blood supply , Vasodilation , Animals , Aorta, Thoracic/enzymology , Aorta, Thoracic/physiopathology , Case-Control Studies , Diet, High-Fat , Dietary Sucrose , Disease Models, Animal , Endothelium, Vascular/physiopathology , Humans , Male , Mechanistic Target of Rapamycin Complex 1/genetics , Mesenteric Arteries/enzymology , Mesenteric Arteries/physiopathology , Mice, Inbred C57BL , Mice, Knockout , Obesity/enzymology , Obesity/genetics , Obesity/physiopathology , Regulatory-Associated Protein of mTOR/deficiency , Regulatory-Associated Protein of mTOR/genetics , Signal Transduction
8.
Eur J Pharmacol ; 899: 173978, 2021 May 15.
Article in English | MEDLINE | ID: mdl-33691164

ABSTRACT

Metabolic syndrome is linked to an increased risk of cardiovascular complications by a mechanism involving mainly decreased nitric oxide (NO) bioavailability and impaired NO-soluble guanylate cyclase (sGC)- cyclic guanosine monophosphate (cGMP) signalling (NO-sGC-cGMP). To further develop this scientific point, this study aimed to investigate the effects of long-term treatment with BAY 41-2272 (a sGC stimulator) on cardiovascular reactivity of spontaneously hypertensive rats (SHR) as a model of metabolic syndrome. SHR were randomly divided into 3 groups: control group, cafeteria diet (CD)-fed group and CD-fed group treated daily with BAY 41-2272 (5 mg/kg) by gastric gavage for 12 weeks. In vivo measurements of body weight, abdominal circumference, blood pressure and glucose tolerance test were performed. At the end of the feeding period, ex vivo cumulative concentration-response curves were performed on isolated perfused heart (isoproterenol (0.1 nM - 1 µM)) and thoracic aorta (phenylephrine (1 nM-10 µM), acetylcholine (1 nM-10 µM), and sodium nitroprusside (SNP) (0.1 nM-0.1 µM)). We showed that chronic CD feeding induced abdominal obesity, hypertriglyceridemia, glucose intolerance and exacerbated arterial hypertension in SHR. Compared to control group, CD-fed group showed a decrease in ß-adrenoceptor-induced cardiac inotropy, in coronary perfusion pressure and in aortic contraction to phenylephrine. While relaxing effects of acetylcholine and SNP were unchanged. BAY 41-2272 long-term treatment markedly prevented arterial hypertension development and glucose intolerance, enhanced the α1-adrenoceptor-induced vasoconstriction, and restored cardiac inotropy and coronary vasodilation. These findings suggest that BAY 41-2272 may be a potential novel drug for preventing metabolic and cardiovascular complications of metabolic syndrome.


Subject(s)
Cardiovascular Diseases/prevention & control , Enzyme Activators/pharmacology , Metabolic Syndrome/prevention & control , Pyrazoles/pharmacology , Pyridines/pharmacology , Soluble Guanylyl Cyclase/metabolism , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/enzymology , Aorta, Thoracic/physiopathology , Cardiovascular Diseases/enzymology , Cardiovascular Diseases/etiology , Cardiovascular Diseases/physiopathology , Coronary Circulation/drug effects , Cyclic GMP/metabolism , Disease Models, Animal , Enzyme Activation , Glucose Intolerance/enzymology , Glucose Intolerance/etiology , Glucose Intolerance/physiopathology , Glucose Intolerance/prevention & control , Hypertension/enzymology , Hypertension/etiology , Hypertension/physiopathology , Hypertension/prevention & control , Hypertriglyceridemia/enzymology , Hypertriglyceridemia/etiology , Hypertriglyceridemia/physiopathology , Hypertriglyceridemia/prevention & control , Isolated Heart Preparation , Male , Metabolic Syndrome/enzymology , Metabolic Syndrome/etiology , Metabolic Syndrome/physiopathology , Nitric Oxide Synthase Type II/metabolism , Obesity, Abdominal/enzymology , Obesity, Abdominal/etiology , Obesity, Abdominal/physiopathology , Obesity, Abdominal/prevention & control , Rats, Inbred SHR , Vasoconstriction/drug effects , Vasodilation/drug effects , Ventricular Function, Left/drug effects , Ventricular Pressure/drug effects
9.
Eur J Pharmacol ; 898: 173991, 2021 May 05.
Article in English | MEDLINE | ID: mdl-33684451

ABSTRACT

In the present study, we investigated the vasorelaxant effects of alogliptin, an oral antidiabetic drug in the dipeptidyl peptidase-4 (DPP-4) inhibitor class, using phenylephrine (Phe)-induced pre-contracted aortic rings. Alogliptin induced vasorelaxation in a dose-dependent manner. Pre-treatment with the voltage-dependent K+ (Kv) channel inhibitor 4-aminopyridine (4-AP) significantly decreased the vasorelaxant effect of alogliptin, whereas pre-treatment with the inwardly rectifying K+ (Kir) channel inhibitor Ba2+, ATP-sensitive K+ (KATP) channel inhibitor glibenclamide, and large-conductance Ca2+-activated K+ (BKCa) channel inhibitor paxilline did not alter the effects of alogliptin. Although pre-treatment with the Ca2+ channel inhibitor nifedipine did not affect the vasorelaxant effect of alogliptin, pre-treatment with the sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) pump inhibitors thapsigargin and cyclopiazonic acid effectively attenuated the vasorelaxant response of alogliptin. Neither cGMP/protein kinase G (PKG)-related signaling pathway inhibitors (guanylyl cyclase inhibitor ODQ and PKG inhibitor KT 5823) nor cAMP/protein kinase A (PKA)-related signaling pathway inhibitors (adenylyl cyclase inhibitor SQ 22536 and PKA inhibitor KT 5720) reduced the vasorelaxant effect of alogliptin. Similarly, the vasorelaxant effect of alogliptin was not changed by endothelium removal or pre-treatment with the nitric oxide (NO) synthase inhibitor L-NAME or the small- and intermediate-conductance Ca2+-activated K+ (SKCa and IKCa) channel inhibitors apamin and TRAM-34. Based on these results, we suggest that alogliptin induced vasorelaxation in rabbit aortic smooth muscle by activating Kv channels and the SERCA pump independent of other K+ channels, cGMP/PKG-related or cAMP/PKA-related signaling pathways, and the endothelium.


Subject(s)
Muscle, Smooth, Vascular/drug effects , Piperidines/pharmacology , Potassium Channels, Voltage-Gated/agonists , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Uracil/analogs & derivatives , Vasodilation/drug effects , Vasodilator Agents/pharmacology , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/enzymology , Enzyme Activation , Male , Muscle, Smooth, Vascular/enzymology , Potassium Channels, Voltage-Gated/metabolism , Rabbits , Signal Transduction , Uracil/pharmacology
10.
Cell Death Dis ; 12(2): 176, 2021 02 12.
Article in English | MEDLINE | ID: mdl-33579894

ABSTRACT

It is well-established that long-term exposure of the vasculature to metabolic disturbances leads to abnormal vascular tone, while the physiological regulation of vascular tone upon acute metabolic challenge remains unknown. Here, we found that acute glucose challenge induced transient increases in blood pressure and vascular constriction in humans and mice. Ex vivo study in isolated thoracic aortas from mice showed that glucose-induced vascular constriction is dependent on glucose oxidation in vascular smooth muscle cells. Specifically, mitochondrial membrane potential (ΔΨm), an essential component in glucose oxidation, was increased along with glucose influx and positively regulated vascular smooth muscle tone. Mechanistically, mitochondrial hyperpolarization inhibited the activity of myosin light chain phosphatase (MLCP) in a Ca2+-independent manner through activation of Rho-associated kinase, leading to cell contraction. However, ΔΨm regulated smooth muscle tone independently of the small G protein RhoA, a major regulator of Rho-associated kinase signaling. Furthermore, myosin phosphatase target subunit 1 (MYPT1) was found to be a key molecule in mediating MLCP activity regulated by ΔΨm. ΔΨm positively phosphorylated MYPT1, and either knockdown or knockout of MYPT1 abolished the effects of glucose in stimulating smooth muscle contraction. In addition, smooth muscle-specific Mypt1 knockout mice displayed blunted response to glucose challenge in blood pressure and vascular constriction and impaired clearance rate of circulating metabolites. These results suggested that glucose influx stimulates vascular smooth muscle contraction via mitochondrial hyperpolarization-inactivated myosin phosphatase, which represents a novel mechanism underlying vascular constriction and circulating metabolite clearance.


Subject(s)
Glucose/administration & dosage , Membrane Potential, Mitochondrial/drug effects , Mitochondria, Muscle/drug effects , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Myosin-Light-Chain Phosphatase/metabolism , Vasoconstriction/drug effects , Adult , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/enzymology , Blood Glucose/metabolism , Blood Pressure/drug effects , Cells, Cultured , Glucose/metabolism , Humans , Male , Mannitol/administration & dosage , Mannitol/blood , Mice, Inbred C57BL , Mice, Knockout , Mitochondria, Muscle/metabolism , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Myosin-Light-Chain Phosphatase/genetics , Oxidation-Reduction , Random Allocation , Signal Transduction , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
11.
Microvasc Res ; 133: 104078, 2021 01.
Article in English | MEDLINE | ID: mdl-32980388

ABSTRACT

The present study assessed the impact of impaired tetrahydrobiopterin (BH4) production on vasoreactivity from conduit and small arteries along the vascular tree as seen during aging. For this purpose, the mutant hyperphenylalaninemic mouse (hph-1) was used. This model is reported to be deficient in GTP cyclohydrolase I, a rate limiting enzyme in BH4 biosynthesis. BH4 is a key regulator of vascular homeostasis by regulating the nitric oxide synthase 3 (NOS3) activity. In GTP-CH deficient mice, the aortic BH4 levels were decreased, by -77% in 12 week-middle-aged mice (young) and by -83% in 35-45 week-middle-aged mice (middle-aged). In young hph-1, the mesenteric artery ability to respond to flow was slightly reduced by 9%. Aging induced huge modification in many vascular functions. In middle-aged hph-1, we observed a decrease in aortic cGMP levels, biomarker of NO availability (-46%), in flow-mediated vasodilation of mesenteric artery (-31%), in coronary hyperemia response measured in isolated heart following transient ischemia (-27%) and in cutaneous microcirculation dilation in response to acetylcholine assessed in vivo by laser-doppler technic (-69%). In parallel, the endothelium-dependent relaxation in response to acetylcholine in conduit blood vessel, measured on isolated aorta rings, was unchanged in hph-1 mice whatever the age. Our findings demonstrate that in middle-aged GTP-CH depleted mice, the reduction of BH4 was characterized by an alteration of microcirculation dilatory properties observed in various parts of the vascular tree. Large conduit blood vessels vasoreactivity, ie aorta, was unaltered even in middle-aged mice emphasizing the main BH4-deletion impact on the microcirculation.


Subject(s)
GTP Cyclohydrolase/deficiency , Microcirculation , Microvessels/enzymology , Phenylketonurias/enzymology , Skin/blood supply , Vasodilation , Age Factors , Animals , Aorta, Thoracic/enzymology , Aorta, Thoracic/physiopathology , Biopterins/analogs & derivatives , Biopterins/metabolism , Coronary Vessels/enzymology , Coronary Vessels/physiopathology , Disease Models, Animal , GTP Cyclohydrolase/genetics , Male , Mesenteric Arteries/enzymology , Mesenteric Arteries/physiopathology , Mice, Inbred C57BL , Mice, Knockout , Microvessels/physiopathology , Phenylketonurias/genetics , Phenylketonurias/physiopathology
12.
Microvasc Res ; 133: 104075, 2021 01.
Article in English | MEDLINE | ID: mdl-32950484

ABSTRACT

OBJECTIVE: Phosphorylation plays an essential role in the regulation of endothelial nitric oxide synthase (eNOS) activity. However, the phosphorylation of eNOS under hypoglycemia and whether hypoglycemia changes eNOS activity is unknown. This paper aims to clarify the regulation of eNOS phosphorylation and its activity change under hypoglycemia. METHODS: Bovine aortic endothelial cells (BAECs) and Sprague-Dawley rats were treated with hypoglycemia, and the phosphorylation of eNOS was subjected to western blot. Blood nitric oxide (NO) concentration was determined by NO kit and endothelial-dependent vasodilation was detected by multi-wire myograph. RESULTS: In both BAECs and rats' thoracic aorta, hypoglycemia induced eNOS phosphorylation decrease specifically on Threonine (Thr) 497. Inhibition of ubiquitination of protein kinase C α subunit (PKCα) reverses the decrease of eNOS phosphorylation in hypoglycemia. Ubiquitinated PKCα can be reversed by AMPK knockdown. In rats, insulin induced hypoglycemia increased the concentration of NO in arterial blood, and progressively enhanced the endothelium-dependent vasodilation of the thoracic and mesenteric aorta. CONCLUSIONS: In vitro, the activation of AMPK may lead to the expression of PKCα by regulating ubiquitination, resulting in a decrease in the level of P-eNOS Thr497 phosphorylation under hypoglycemia. In vivo, insulin-induced hypoglycemia produces a beneficial cardiovascular effect on rats.


Subject(s)
Aorta, Thoracic/enzymology , Endothelial Cells/enzymology , Hypoglycemia/enzymology , Mesenteric Arteries/enzymology , Nitric Oxide Synthase Type III/metabolism , Vasodilation , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Animals , Aorta, Thoracic/physiopathology , Blood Glucose/metabolism , Cattle , Cells, Cultured , Disease Models, Animal , Female , Hypoglycemia/physiopathology , Male , Mesenteric Arteries/physiopathology , Nitric Oxide/blood , Phosphorylation , Protein Kinase C-alpha/genetics , Protein Kinase C-alpha/metabolism , Rats, Sprague-Dawley , Signal Transduction
13.
Life Sci ; 264: 118606, 2021 Jan 01.
Article in English | MEDLINE | ID: mdl-33091444

ABSTRACT

AIMS: Sepsis is a severe endothelial dysfunction syndrome. The role of endothelial nitric oxide synthase (eNOS) in endothelial dysfunction induced by sepsis is controversial. To explore the role of eNOS in vascular dysfunction. MAIN METHODS: The effect of sepsis on vasodilation and eNOS levels was examined in septic mouse arteries and in cell models. KEY FINDINGS: In early sepsis mouse arteries, endothelium-dependent relaxation decreased and phosphorylation of the inhibitory Thr495 site in endothelial nitric oxide synthase increased. Mechanically, the phosphorylation of endothelial nitric oxide synthase at Thr497 in bovine aortic endothelial cells occurred in a protein kinase C-α dependent manner. In late sepsis, both nitric oxide-dependent relaxation responses and endothelial nitric oxide synthase levels were decreased in septic mice arteries. Endothelial nitric oxide synthase levels expression levels decreased in tumor necrosis factor-α-treated human umbilical vein endothelial cells and this could be prevented by the ubiquitin proteasome inhibitor (MG-132). MG-132 could reverse the decrease in endothelial nitric oxide synthase expression and improve nitric oxide-dependent vasodilator dysfunction in septic mice arteries. SIGNIFICANCE: These data indicate that vasodilator dysfunction is induced by the increased phosphorylation of endothelial nitric oxide synthase in early sepsis and its degradation in late sepsis.


Subject(s)
Gene Expression Regulation, Enzymologic , Nitric Oxide Synthase Type III/biosynthesis , Sepsis/enzymology , Sepsis/physiopathology , Vasodilation/physiology , Acetylcholine/pharmacology , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/enzymology , Aorta, Thoracic/physiopathology , Cattle , Dose-Response Relationship, Drug , Human Umbilical Vein Endothelial Cells , Humans , Lipopolysaccharides/toxicity , Male , Mesenteric Arteries/drug effects , Mesenteric Arteries/enzymology , Mesenteric Arteries/physiopathology , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type III/genetics , Organ Culture Techniques , Sepsis/chemically induced , Vasodilation/drug effects , Vasodilator Agents/pharmacology
14.
Biomed Pharmacother ; 134: 111162, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33360932

ABSTRACT

Non-specific histone deacetylase (HDAC) inhibition reduces high blood pressure in essential hypertensive animal models. However, the exact HDAC isoforms that play a critical role in controlling hypertension are not known. Here, we investigated the role of HDAC5 in vascular contraction, hypertrophy, and oxidative stress in the context of angiotensin II (Ang II)-induced hypertension. Genetic deletion of HDAC5 and treatment with class IIa HDAC inhibitors (TMP269 and TMP195) prevented Ang II-induced increases in blood pressure and arterial wall thickness. Hdac5-knockout mice were also resistant to the thromboxane A2 agonist (U46619)-induced vascular contractile response. Furthermore, the expression of Rho-associated protein kinase (ROCK) 2 was downregulated in the aortas of Ang II-treated Hdac5-knockout mice. Knockdown of HDAC5, RhoA, or ROCK2 reduced collagen gel contraction, whereas silencing of ROCK1 increased it. VSMC hypertrophy reduced on knocking down HDAC5, ROCK1, and ROCK2. Here we showed that genetic deletion of HDAC5 and pharmacological inhibition of class IIa HDACs ameliorated Ang II-induced ROS generation. Moreover, ROCK1 and ROCK2, the downstream targets of HDAC5, influenced ROS generation. The relative protein levels of HDAC5, ROCK1, and ROCK2 were increased both in the cytoplasm and nuclear fraction in response to Ang II stimulation in vascular smooth muscle cells. Inhibition of HDAC5 expression or activity reduced vascular hypertrophy, vasoconstriction, and oxidative stress in the Ang II-induced hypertension model. These findings indicate that HDAC5 may serve as a potential target in the treatment of hypertension.


Subject(s)
Antihypertensive Agents/pharmacology , Arterial Pressure/drug effects , Benzamides/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Hypertension/prevention & control , Muscle, Smooth, Vascular/drug effects , Oxadiazoles/pharmacology , Oxidative Stress/drug effects , Vascular Remodeling/drug effects , Vasoconstriction/drug effects , Angiotensin II , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/enzymology , Aorta, Thoracic/physiopathology , Cells, Cultured , Disease Models, Animal , Histone Deacetylases/deficiency , Histone Deacetylases/genetics , Hypertension/chemically induced , Hypertension/enzymology , Hypertension/physiopathology , Male , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/physiopathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/enzymology , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
15.
Life Sci ; 266: 118885, 2021 Feb 01.
Article in English | MEDLINE | ID: mdl-33316265

ABSTRACT

AIM: We determined the role played by O-linked N-acetylglucosamine (O-GlcNAc) of proteins in systemic arteries during late pregnancy in normotensive and hypertensive rats. MAIN METHODS: O-GlcNAc levels and O-GlcNAc modification of endothelial nitric oxide synthase (eNOS) were determined in aorta (conductance vessel) and mesenteric arteries (resistance vessels) of non-pregnant (NP) and pregnant (P) Wistar rats and spontaneously hypertensive rats (SHR). Vascular O-GlcNAc-modified proteins, O-GlcNAcase (OGA) and O-GlcNAc transferase (OGT) expression, and OGA activity were analyzed. Concentration-response to phenylephrine (PE) curves were constructed for arteries with and without endothelium. Arteries were treated with vehicle or PugNAc (OGA inhibitor, 100 µmol/L) in the presence of L-NAME (NOS inhibitor, 100 µmol/L). KEY FINDINGS: The content of vascular O-GlcNAc-modified proteins was lower, OGT and OGA expression did not change, and OGA activity was higher in arteries of P-Wistar rats and P-SHR compared to arteries of NP-groups. Reactivity to PE increased in arteries of P-Wistar rats treated with PugNAc compared to vehicle. O-GlcNAcylation of eNOS decreased in P-SHR compared to NP-SHR. PugNAc partially inhibited the effects of endothelium removal and L-NAME on reactivity to PE in arteries of P-Wistar rats. However, PugNAc did not alter reactivity to PE in arteries of P-SHR. Our data showed that pregnancy decreased the content of vascular O-GlcNAc-modified proteins. SIGNIFICANCE: Increased OGA activity and decreased O-GlcNAc modification of eNOS boosts eNOS activity in arteries of P-Wistar rats. In P-SHR, altered OGA activity may lower the content of O-GlcNAc-modified proteins, but decreased OGT activity seems a potential mechanism to reduce glycosylation.


Subject(s)
Acetylglucosamine/chemistry , Aorta, Thoracic/physiopathology , Hypertension/physiopathology , Mesenteric Arteries/physiopathology , Protein Processing, Post-Translational , beta-N-Acetylhexosaminidases/metabolism , Animals , Aorta, Thoracic/enzymology , Female , Glycosylation , Hypertension/enzymology , Mesenteric Arteries/enzymology , N-Acetylglucosaminyltransferases , Pregnancy , Rats , Rats, Inbred SHR , Rats, Wistar , beta-N-Acetylhexosaminidases/chemistry
16.
Aging (Albany NY) ; 12(21): 21423-21445, 2020 11 03.
Article in English | MEDLINE | ID: mdl-33144524

ABSTRACT

Vascular endothelial cell senescence and endoplasmic reticulum (ER) stress induced unfolded protein response (UPR) are two critical contributors to individual aging. However, whether these two biological events have crosstalk and are controlled by shared upstream regulators are largely unknown. Here, we found PARP16, a member of the Poly (ADP-ribose) polymerases family that tail-anchored ER transmembrane, was upregulated in angiotensin II (Ang II)-induced vascular aging and promoted UPR. Further, PARP16 was epigenetically upregulated by Smyd3, a histone H3 lysine 4 methyltransferase that bound to the promotor region of Parp16 gene and increased H3K4me3 level to activate its host gene's transcription. Intervention of either Smyd3 or PARP16 ameliorated vascular aging associated phenotypes in both cell and mice models. This study identified Smyd3-PARP16 as a novel signal axis in regulating UPR and endothelial senescence, and targeting this axis has implications in preventing vascular aging and related diseases.


Subject(s)
Aorta, Thoracic/enzymology , Cellular Senescence , Endothelial Cells/enzymology , Poly(ADP-ribose) Polymerases/metabolism , Unfolded Protein Response , Angiotensin II/toxicity , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/pathology , Binding Sites , Cells, Cultured , Cellular Senescence/drug effects , DNA Methylation , Endothelial Cells/drug effects , Endothelial Cells/pathology , Epigenesis, Genetic , Gene Expression Regulation , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Male , Mice, Inbred C57BL , Poly(ADP-ribose) Polymerases/genetics , Promoter Regions, Genetic , Rats , Signal Transduction , Unfolded Protein Response/drug effects
17.
Am J Physiol Heart Circ Physiol ; 319(2): H341-H348, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32618512

ABSTRACT

Progesterone exerts antihypertensive actions partially by modulating endothelial nitric oxide synthase (eNOS) activity. Here, we aimed to investigate the effects and mechanisms of progesterone on eNOS expression. First, human umbilical vein endothelial cells (HUVECs) were exposed to progesterone and then the eNOS transcription factor specificity protein-1 (SP-1) and progesterone receptor (PRA/B) expression were assessed by Western blotting and qRT-PCR. The interaction between SP-1 and PRA/B was next determined through coimmunoprecipitation assay. The chromatin immunoprecipitation assay and luciferase assay were used to investigate the relationship of PRA/B, SP-1, and eNOS promoter. At last, rats were intraperitoneally injected with progesterone receptor antagonist RU-486, and then the expression of eNOS and vasodilation function in thoracic aorta and mesenteric artery were measured. The results showed that progesterone could increase eNOS expression in HUVECs. Further study showed that progesterone increased PRA-SP-1 complex formation and facilitated PRA/B and SP-1 binding to eNOS promoter. Mutating SP-1 or PR-binding motif on eNOS promoter abolished the effect of progesterone on eNOS gene transcription. We also observed that progesterone receptor antagonist RU-486 reduced eNOS expression and impaired vasodilation in rats. Those results suggest that progesterone modulates eNOS expression through promoting PRA-SP-1 complex formation, and progesterone antagonist attenuates eNOS expression, leading to the loss of vascular relaxation.NEW & NOTEWORTHY Progesterone directly upregulated endothelial nitric oxide synthase (eNOS) expression in human endothelial cells. Progesterone augmented eNOS promoter activity through a progesterone receptor A- and specificity protein-1-dependent manner. Antagonism of the progesterone receptor reduced eNOS expression and impaired vasodilation in rats.


Subject(s)
Cell Nucleus/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Nitric Oxide Synthase Type III/biosynthesis , Progesterone/pharmacology , Receptors, Progesterone/agonists , Sp1 Transcription Factor/metabolism , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/enzymology , Binding Sites , Cell Nucleus/metabolism , Cells, Cultured , Enzyme Induction , Female , Hormone Antagonists/pharmacology , Human Umbilical Vein Endothelial Cells/enzymology , Humans , Mesenteric Arteries/drug effects , Mesenteric Arteries/enzymology , Nitric Oxide Synthase Type III/genetics , Promoter Regions, Genetic , Rats, Sprague-Dawley , Receptors, Progesterone/antagonists & inhibitors , Receptors, Progesterone/metabolism , Signal Transduction , Vasodilation/drug effects
18.
Eur J Pharmacol ; 883: 173374, 2020 Sep 15.
Article in English | MEDLINE | ID: mdl-32682786

ABSTRACT

Abnormal aortic adventitial fibroblasts (AFs) play essential roles in the development of vascular remodeling and disorders. Previous studies revealed that microRNA-122 (miR-122) levels were elevated in the aortic adventitia of hypertensive rats with vascular injury. Here, we aim to evaluate the biological effects and underlying mechanisms of miR-122 in rat AFs. Exposure to angiotensin II (ATII) in rat AFs resulted in decreased levels of sirtuin 6 (SIRT6), elabela (ELA), and angiotensin-converting enzyme 2 (ACE2). Additionally, stimulation with ATII contributed to a decline in autophagic flux and obvious increases in cellular migration, oxidative stress, and apoptosis, which were exacerbated by the transfection of miR-122-5p mimic but were rescued by miR-122-5p inhibitor, exogenous replenishment of ELA, and recombinant adeno-associated virus expressing SIRT6 (rAAV-SIRT6), respectively. Moreover, stimulation with miR-122-5p mimic led to a marked reduction in the levels of SIRT6 and ELA in rat AFs, which were elevated by stimulation with rAAV-SIRT6. Furthermore, miR-122-5p inhibitor-mediated pro-autophagic, anti-oxidant and anti-apoptotic effects in rat AFs were partially suppressed by 3-methyladenine, SIRT6 small interfering RNA (siRNA) and ELA siRNA, which were linked with the downregulation in the protein levels of LC3-II, beclin-1, and ACE2 and the upregulation of p62 expression and bax/bcl-2 ratio. Our findings indicated that miR-122-5p inhibition prevented ATII-mediated loss of autophagy, and the promotion of apoptosis and oxidative stress via activating the SIRT6-ELA-ACE2 signaling. MiR-122-5p may be a novel predictive biomarker of adventitial injury, and targeting the SIRT6-ELA-ACE2 signaling may have the potential therapeutic importance of controlling vascular remodeling and disorders.


Subject(s)
Adventitia/drug effects , Angiotensin II/pharmacology , Angiotensin-Converting Enzyme 2/metabolism , Aorta, Thoracic/drug effects , Apoptosis/drug effects , Autophagy/drug effects , Fibroblasts/drug effects , MicroRNAs/metabolism , Peptide Hormones/metabolism , Sirtuins/metabolism , Adventitia/enzymology , Adventitia/pathology , Angiotensin-Converting Enzyme 2/genetics , Animals , Aorta, Thoracic/enzymology , Aorta, Thoracic/pathology , Apoptosis Regulatory Proteins/metabolism , Autophagy-Related Proteins/metabolism , Cells, Cultured , Fibroblasts/enzymology , Fibroblasts/pathology , Male , MicroRNAs/genetics , Oxidative Stress/drug effects , Peptide Hormones/genetics , Rats, Sprague-Dawley , Signal Transduction , Sirtuins/genetics
19.
Toxins (Basel) ; 12(6)2020 06 25.
Article in English | MEDLINE | ID: mdl-32630586

ABSTRACT

Zearalenone (ZEN) is a non-steroidal mycotoxin that has various toxicological impacts on mammalian health. Here, we found that ZEN significantly affected the production of nitric oxide (NO) and the expression of endothelial NO synthase (eNOS) of bovine aortic endothelial cells (BAECs). A promoter analysis using 5'-serially deleted human eNOS promoter revealed that the proximal region (-135 to +22) was responsible for ZEN-mediated reduction of the human eNOS promoter activity. This effect was reversed by mutation of two specificity protein 1 (Sp1) binding elements in the human eNOS promoter. A chromatin immunoprecipitation assay revealed that ZEN increased Sp1 binding to the bovine eNOS promoter region (-113 to -12), which is homologous to -135 to +22 of the human eNOS promoter region. We also found that ZEN promoted the binding of the pregnane X receptor (PXR) to Sp1 of the bovine eNOS, consequently decreasing eNOS expression. This reduction of eNOS could have contributed to the decreased acetylcholine-induced vessel relaxation upon ZEN treatment in our ex vivo study using mouse aortas. In conclusion, our data demonstrate that ZEN decreases eNOS expression by enhancing the binding of PXR-Sp1 to the eNOS promoter, thereby decreasing NO production and potentially causing vessel dysfunction.


Subject(s)
Endothelial Cells/drug effects , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide/metabolism , Pregnane X Receptor/metabolism , Sp1 Transcription Factor/metabolism , Transcription, Genetic , Zearalenone/toxicity , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/enzymology , Binding Sites , Cattle , Cells, Cultured , Down-Regulation , Endothelial Cells/enzymology , Male , Mice, Inbred C57BL , Nitric Oxide Synthase Type III/genetics , Promoter Regions, Genetic , Protein Binding , Vasodilation/drug effects
20.
Eur J Pharmacol ; 883: 173384, 2020 Sep 15.
Article in English | MEDLINE | ID: mdl-32707188

ABSTRACT

Alamandine attenuates hypertension and cardiac remodeling in spontaneously hypertensive rats (SHRs). We examined whether alamandine attenuates vascular remodeling in mice, and regulates angiotensin II (Ang II)-induced fibrosis in rat vascular smooth muscle cells (VSMCs). Alamandine attenuated hypertension in mice induced by Ang II. Ang II increased the fibrosis of thoracic aorta in mice, which was attenuated by alamandine treatment. Increased levels of collagen I, transforming growth factor-ß (TGF-ß), and connective tissue growth factor (CTGF) levels in thoracic aortas after Ang II treatment in mice were inhibited by alamandine. Ang II-stimulated collagen I, TGF-ß, and CTGF level increases were inhibited by alamandine in rat VSMCs. This could be reversed by Mas-related G protein-coupled receptor, member D (MrgD) antagonist D-Pro7-Ang-(1-7) but not Mas receptor antagonist A779. MrgD expression was increased in the thoracic aortas of mice or VSMCs treatment with Ang II. Ang II increased p-p38 and cAMP levels in rat VSMCs, and alamandine blocked Ang II-induced these increases. Cyclic adenosine monophosphate (cAMP) reversed the inhibitory effects of alamandine on the Ang II-induced increases in collagen I, TGF-ß, and CTGF levels. These results demonstrate alamandine attenuates vascular fibrosis by stimulating MrgD expression and decreases arterial fibrosis by blocking p-p38 expression. Alamandine/MrgD axis is a potential target for the treatment of vascular remodeling.


Subject(s)
Angiotensin II , Antihypertensive Agents/pharmacology , Hypertension/prevention & control , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Oligopeptides/pharmacology , Vascular Remodeling/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/enzymology , Aorta, Thoracic/physiopathology , Arterial Pressure/drug effects , Cells, Cultured , Disease Models, Animal , Fibrosis , Hypertension/chemically induced , Hypertension/enzymology , Hypertension/physiopathology , Male , Mice, Inbred C57BL , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/physiopathology , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/pathology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Phosphorylation , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...