Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Cardiovasc Drugs Ther ; 35(1): 87-101, 2021 02.
Article in English | MEDLINE | ID: mdl-33057968

ABSTRACT

PURPOSE: This study aimed to investigate whether inhibition of glucagon-like peptide-1 (GLP-1) on pressure overload induced cardiac hypertrophy and apoptosis is related to activation of ATP sensitive potassium (KATP) channels. METHODS: Male SD rats were randomly divided into five groups: sham, control (abdominal aortic constriction), GLP-1 analog liraglutide (0.3 mg/kg/twice day), KATP channel blocker glibenclamide (5 mg/kg/day), and liraglutide plus glibenclamide. RESULTS: Relative to the control on week 16, liraglutide upregulated protein and mRNA levels of KATP channel subunits Kir6.2/SUR2 and their expression in the myocardium, vascular smooth muscle, aortic endothelium, and cardiac microvasculature. Consistent with a reduction in aortic wall thickness (61.4 ± 7.6 vs. 75.0 ± 7.6 µm, p < 0.05), liraglutide enhanced maximal aortic endothelium-dependent relaxation in response to acetylcholine (71.9 ± 8.7 vs. 38.6 ± 4.8%, p < 0.05). Along with a reduction in heart to body weight ratio (2.6 ± 0.1 vs. 3.4 ± 0.4, mg/g, p < 0.05) by liraglutide, hypertrophied cardiomyocytes (371.0 ± 34.4 vs. 933.6 ± 156.6 µm2, p < 0.05) and apoptotic cells (17.5 ± 8.2 vs. 44.7 ± 7.9%, p < 0.05) were reduced. Expression of anti-apoptotic protein BCL-2 and contents of myocardial ATP were augmented, and expression of cleaved-caspase 3 and levels of serum Tn-I/-T were reduced. Echocardiography and hemodynamic measurement showed that cardiac systolic function was enhanced as evidenced by increased ejection fraction (88.4 ± 4.8 vs. 73.8 ± 5.1%, p < 0.05) and left ventricular systolic pressure (105.2 ± 10.8 vs. 82.7 ± 7.9 mmHg, p < 0.05), and diastolic function was preserved as shown by a reduction of ventricular end-diastolic pressure (-3.1 ± 2.9 vs. 6.7 ± 2.8 mmHg, p < 0.05). Furthermore, left ventricular internal diameter at end-diastole (5.8 ± 0.5 vs. 7.7 ± 0.6 mm, p < 0.05) and left ventricular internal diameter at end-systole (3.0 ± 0.6 vs. 4.7 ± 0.4 mm, p < 0.05) were improved. Dietary administration of glibenclamide alone did not alter all the parameters measured but significantly blocked liraglutide-exerted cardioprotection. CONCLUSION: Liraglutide ameliorates cardiac hypertrophy and apoptosis, potentially via activating KATP channel-mediated signaling pathway. These data suggest that liraglutide might be considered as an adjuvant therapy to treat patients with heart failure.


Subject(s)
Apoptosis/drug effects , Glucagon-Like Peptide 1/pharmacology , Glyburide/pharmacology , KATP Channels/drug effects , Liraglutide/pharmacology , Animals , Cardiomegaly , Drug Therapy, Combination , Male , Random Allocation , Rats , Rats, Sprague-Dawley
2.
Eur J Pharmacol ; 883: 173318, 2020 Sep 15.
Article in English | MEDLINE | ID: mdl-32621911

ABSTRACT

Mammalian target of rapamycin (mTOR) and a ribosomal protein S6 kinase (p70S6K) mediate tissue fibrosis and negatively regulate autophagy. This study aims to investigate whether glucagon-like peptide-1 (GLP-1) analog liraglutide protects the heart against aortic banding-induced cardiac fibrosis and dysfunction through inhibiting mTOR/p70S6K signaling and promoting autophagy activity. Male SD rats were randomly divided into four groups (n = 6/each group): sham operated control; abdominal aortic constriction (AAC); liraglutide treatment during AAC (0.3 mg/kg, injected subcutaneously twice daily); rapamycin treatment during AAC (0.2 mg/kg/day, administered by gastric gavage). Relative to the animals with AAC on week 16, liraglutide treatment significantly reduced heart/body weight ratio, inhibited cardiomyocyte hypertrophy, and augmented plasma GLP-1 level and tissue GLP-1 receptor expression. Phosphorylation of mTOR/p70S6K, populations of myofibroblasts and synthesis of collagen I/III in the myocardium were simultaneously inhibited. Furthermore, autophagy regulating proteins: LC3-II/LC3-I ratio and Beclin-1 were upregulated, and p62 was downregulated by liraglutide. Compared with liraglutide group, treatment with rapamycin, a specific inhibitor of mTOR, compatibly augmented GLP-1 receptor level, inhibited phosphorylation of mTOR/p70S6K and expression of p62 as well as increased level of LC3-II/LC3-I ratio and Beclin-1, suggesting that there is an interaction between GLP-1 and mTOR/p70S6K signaling in the regulation of autophagy. In line with these modifications, treatment with liraglutide and rapamycin significantly reduced perivascular/interstitial fibrosis, and preserved systolic/diastolic function. These results suggest that the inhibitory effects of liraglutide on cardiac fibrosis and dysfunction are potentially mediated by inhibiting mTOR/p70S6K signaling and enhancing autophagy activity.


Subject(s)
Autophagy/drug effects , Glucagon-Like Peptide 1/pharmacology , Hypertrophy, Left Ventricular/prevention & control , Myocytes, Cardiac/drug effects , Myofibroblasts/drug effects , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , TOR Serine-Threonine Kinases/metabolism , Ventricular Function, Left/drug effects , Ventricular Remodeling/drug effects , Animals , Aorta, Abdominal/physiopathology , Aorta, Abdominal/surgery , Autophagy-Related Proteins/metabolism , Disease Models, Animal , Fibrosis , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/metabolism , Hypertrophy, Left Ventricular/enzymology , Hypertrophy, Left Ventricular/pathology , Hypertrophy, Left Ventricular/physiopathology , Incretins/pharmacology , Ligation , Male , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Myofibroblasts/enzymology , Myofibroblasts/pathology , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Rats, Sprague-Dawley , Signal Transduction , TOR Serine-Threonine Kinases/antagonists & inhibitors
3.
Eur J Pharmacol ; 867: 172844, 2020 Jan 15.
Article in English | MEDLINE | ID: mdl-31811859

ABSTRACT

This study tested the hypothesis that the enhancement of glucagon-like peptide-1 (GLP-1) level through either exogenous supply of GLP-1 agonist, liraglutide or prevention of endogenous GLP-1 degradation with dipeptidyl peptidease-4 inhibitor, lingaliptin ameliorates angiotensin II (Ang II)-induced renal fibrosis. Sprague-Dawley rats were randomly divided into four groups: 0.9% saline or Ang II (500 ng/kg/min) was infused with osmotic minipumps for 4 weeks, defined as sham and Ang II groups. In drug treated groups, liraglutide (0.3 mg/kg) was injected subcutaneously twice daily or linagliptin (8 mg/kg) was administered daily via oral gavage during Ang II infusion. Compared with Ang II stimulation, liraglutide or linagliptin comparatively down-regulated the protein level of the AT1 receptor, and up-regulated the AT2 receptor, as identified by a reduced AT1/AT2 ratio (all p < 0.05), consistent with less locally-expressed AT1 receptor and enhanced AT2 receptor in the glomerular capillaries and proximal tubules of the renal cortex. Furthermore, both drugs significantly increased the expression of GLP-1 receptor and attenuated the protein levels of TLR4, NOX4 and IL-6. The populations of macrophages and α-SMA expressing myofibroblasts decreased with treatment of liraglutide and linagliptin, in coincidence with the reduced expression of phosphor-Smad2/3, Smad4, TGFß1, and up-regulated Smad7. Along with these modulations, renal morphology was preserved and synthesis of fibronectin/collagen I was down-regulated, as identified by small collagen-rich area in the renal cortex. These results suggest that the preservation of GLP-1 level using liraglutide or linagliptin might be considered as an add-on therapeutic option for inhibiting Ang II induced renal fibrosis and failure.


Subject(s)
Angiotensin II/metabolism , Dipeptidyl-Peptidase IV Inhibitors/administration & dosage , Glucagon-Like Peptide 1/metabolism , Incretins/administration & dosage , Kidney Failure, Chronic/prevention & control , Kidney/pathology , Angiotensin II/administration & dosage , Animals , Dipeptidyl Peptidase 4/metabolism , Disease Models, Animal , Fibrosis , Glucagon-Like Peptide 1/agonists , Humans , Kidney/drug effects , Kidney Failure, Chronic/metabolism , Kidney Failure, Chronic/pathology , Linagliptin/administration & dosage , Liraglutide/administration & dosage , Male , Proteolysis/drug effects , Rats
4.
Mol Biol Rep ; 47(2): 1207-1222, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31820314

ABSTRACT

Aldosterone produced in adrenal glands by angiotensin II (Ang II) is known to elicit myocardial fibrosis and hypertrophy. This study was designed to test the hypothesis that Ang II causes cardiac morphological changes through the steroidogenic acute regulatory protein (StAR)/aldosterone synthase (AS)-dependent aldosterone synthesis primarily initiated in the heart. Sprague-Dawley rats were randomized to following groups: Ang II infusion for a 4-week period, treatment with telmisartan, spironolactone or adrenalectomy during Ang II infusion. Sham-operated rats served as control. Relative to Sham rats, Ang II infusion significantly increased the protein levels of AT1 receptor, StAR, AS and their tissue expression in the adrenal glands and heart. In coincidence with reduced aldosterone level in the heart, telmisartan, an AT1 receptor blocker, significantly down-regulated the protein level and expression of StAR and AS. Ang II induced changes in the expression of AT1/StAR/AS were not altered by an aldosterone receptor antagonist spironolactone. Furthermore, Ang II augmented migration of macrophages, protein level of TGFß1, phosphorylation of Smad2/3 and proliferation of myofibroblasts, accompanied by enhanced perivascular/interstitial collagen deposition and cardiomyocyte hypertrophy, which all were significantly abrogated by telmisartan or spironolactone. However, adrenalectomy did not fully suppress Ang II-induced cell migration/proliferation and fibrosis/hypertrophy, indicating a role of aldosterone synthesized within the heart in pathogenesis of Ang II induced injury. These results indicate that myocardial fibrosis and hypertrophy stimulated by Ang II is associated with tissue-specific activation of aldosterone synthesis, primarily mediated by AT1/StAR/AS signaling pathways.


Subject(s)
Angiotensin II/metabolism , Cardiomegaly/etiology , Cardiomegaly/metabolism , Cardiomyopathies/etiology , Cardiomyopathies/metabolism , Cytochrome P-450 CYP11B2/metabolism , Phosphoproteins/genetics , Adrenal Glands/metabolism , Animals , Biomarkers , Biopsy , Cardiomegaly/pathology , Cardiomyopathies/pathology , Collagen/metabolism , Disease Models, Animal , Disease Susceptibility , Fibrosis , Immunohistochemistry , Macrophages/immunology , Macrophages/metabolism , Male , Models, Biological , Myocardium/metabolism , Myocardium/pathology , Myofibroblasts/metabolism , Rats , Receptor, Angiotensin, Type 1/metabolism , Signal Transduction , Smad2 Protein/metabolism , Smad3 Protein/metabolism
5.
Drug Des Devel Ther ; 13: 2745-2757, 2019.
Article in English | MEDLINE | ID: mdl-31496651

ABSTRACT

OBJECTIVE: Angiotensin II (Ang II) is known to contribute to the pathogenesis of heart failure by eliciting cardiac remodeling and dysfunction. The glucagon-like peptide-1 (GLP-1) has been shown to exert cardioprotective effects in animals and patients. This study investigates whether GLP-1 receptor agonist liraglutide inhibits abdominal aortic constriction (AAC)-induced cardiac fibrosis and dysfunction through blocking Ang II type 1 receptor (AT1R) signaling. METHODS: Sprague-Dawley rats were subjected to sham operation and abdominal aortic banding procedure for 16 weeks. In treated rats, liraglutide (0.3 mg/kg) was subcutaneously injected twice daily or telmisartan (10 mg/kg/day), the AT1R blocker, was administered by gastric gavage. RESULTS: Relative to the animals with AAC, liraglutide reduced protein level of the AT1R and upregulated the AT2R, as evidenced by reduced ratio of AT1R/AT2R (0.59±0.04 vs. 0.91±0.06, p<0.05). Furthermore, the expression of angiotensin converting enzyme 2 was upregulated, tissue levels of malondialdehyde and B-type natriuretic peptide were reduced, and superoxide dismutase activity was increased. Along with a reduction in HW/BW ratio, cardiomyocyte hypertrophy was inhibited. In coincidence with these changes, liraglutide significantly decreased the populations of macrophages and myofibroblasts in the myocardium, which were accompanied by reduced protein levels of transforming growth factor beta1, Smad2/3/4, and upregulated smad7. The synthesis of collagen I and III was inhibited and collagen-rich fibrosis was attenuated. Consistent with these findings, cardiac systolic function was preserved, as shown by increased left ventricular systolic pressure (110±5 vs. 99±2 mmHg, p<0.05), ejection fraction (83%±2% vs. 69%±4%, p<0.05) and fraction shortening (49%±2% vs. 35%±3%, p<0.05). Treatment with telmisartan provided a comparable level of protection as compared with liraglutide in all the parameters measured. CONCLUSION: Taken together, liraglutide ameliorates cardiac fibrosis and dysfunction, potentially via suppressing the AT1R-mediated events. These data indicate that liraglutide might be selected as an add-on drug to prevent the progression of heart failure.


Subject(s)
Constriction, Pathologic/drug therapy , Heart/drug effects , Liraglutide/pharmacology , Receptor, Angiotensin, Type 1/agonists , Ventricular Remodeling/drug effects , Animals , Constriction, Pathologic/metabolism , Dose-Response Relationship, Drug , Echocardiography , Injections, Subcutaneous , Liraglutide/administration & dosage , Male , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1/metabolism
6.
Shock ; 51(3): 372-380, 2019 03.
Article in English | MEDLINE | ID: mdl-29470359

ABSTRACT

This study tested the hypothesis that CD44 is involved in the development of cardiac fibrosis via angiotensin II (Ang II) AT1 receptor-stimulated TNFα/NFκB/IκB signaling pathways. Study was conducted in C57BL/6 wild type and CD44 knockout mice subjected to Ang II infusion (1,000 ng/kg/min) using osmotic minipumps up to 4 weeks or with gastric gavage administration of the AT1 receptor blocker, telmisartan at a dose of 10 mg/kg/d. Results indicated that Ang II enhances expression of the AT1 receptor, TNFα, NFκB, and CD44 as well as downregulates IκB. Further analyses revealed that Ang II increases macrophage migration, augments myofibroblast proliferation, and induces vascular/interstitial fibrosis. Relative to the Ang II group, treatment with telmisartan significantly reduced expression of the AT1 receptor and TNFα. These changes occurred in coincidence with decreased NFκB, increased IκB, and downregulated CD44 in the intracardiac vessels and intermyocardium. Furthermore, macrophage migration and myofibroblast proliferation were inhibited and fibrosis was attenuated. Knockout of CD44 did not affect Ang II-stimulated AT1 receptor and modulated TNFα/NFκB/IκB signaling, but significantly reduced macrophage/myofibroblast-mediated fibrosis as identified by less extensive collagen-rich area. These results suggest that the AT1 receptor is involved in the development of cardiac fibrosis by stimulating TNFα/NFκB/IκB-triggered CD44 signaling pathways. Knockout of CD44 blocked Ang II-induced cell migration/proliferation and cardiac fibrosis. Therefore, selective inhibition of CD44 may be considered as a potential therapeutic target for attenuating Ang II-induced deleterious cardiovascular effects.


Subject(s)
Angiotensin II/adverse effects , Heart Diseases/prevention & control , Hyaluronan Receptors/deficiency , Myocardium/metabolism , Signal Transduction/drug effects , Angiotensin II/pharmacology , Animals , Female , Fibrosis , Heart Diseases/chemically induced , Heart Diseases/genetics , Heart Diseases/metabolism , Hyaluronan Receptors/metabolism , Male , Mice , Mice, Knockout , Myocardium/pathology , NF-kappa B/genetics , NF-kappa B/metabolism , Signal Transduction/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
7.
Drug Des Devel Ther ; 11: 3019-3033, 2017.
Article in English | MEDLINE | ID: mdl-29081650

ABSTRACT

Angiotensin II (Ang II) is known to be involved in the progression of ventricular dysfunction and heart failure by eliciting cardiac fibrosis. The purpose of this study was to demonstrate whether treatment with an antioxidant compound, edaravone, reduces cardiac fibrosis and improves ventricular function by inhibiting Ang II AT1 receptor. The study was conducted in a rat model of transverse aortic constriction (TAC). In control, rats were subjected to 8 weeks of TAC. In treated rats, edaravone (10 mg/kg/day) or Ang II AT1 receptor blocker, telmisartan (10 mg/kg/day) was administered by intraperitoneal injection or gastric gavage, respectively, during TAC. Relative to the animals with TAC, edaravone reduced myocardial malonaldehyde level and increased superoxide dismutase activity. Protein level of the AT1 receptor was reduced and the AT2 receptor was upregulated, as evidenced by the reduced ratio of AT1 over AT2 receptor (0.57±0.2 vs 3.16±0.39, p<0.05) and less locally expressed AT1 receptor in the myocardium. Furthermore, the protein level of angiotensin converting enzyme 2 was upregulated. In coincidence with these changes, edaravone significantly decreased the populations of macrophages and myofibroblasts in the myocardium, which were accompanied by reduced levels of transforming growth factor beta 1 and Smad2/3. Collagen I synthesis was inhibited and collagen-rich fibrosis was attenuated. Relative to the TAC group, cardiac systolic function was preserved, as shown by increased left ventricular systolic pressure (204±51 vs 110±19 mmHg, p<0.05) and ejection fraction (82%±3% vs 60%±5%, p<0.05). Treatment with telmisartan provided a comparable level of protection as compared with edaravone in all the parameters measured. Taken together, edaravone treatment ameliorates cardiac fibrosis and improves left ventricular function in the pressure overload rat model, potentially via suppressing the AT1 receptor-mediated signaling pathways. These data indicate that edaravone might be selected in combination with other existing drugs in preventing progression of cardiac dysfunction in heart failure.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Antipyrine/analogs & derivatives , Free Radical Scavengers/pharmacology , Ventricular Function, Left/drug effects , Angiotensin II/metabolism , Angiotensin-Converting Enzyme 2 , Animals , Antipyrine/pharmacology , Aorta/pathology , Benzimidazoles/pharmacology , Benzoates/pharmacology , Disease Models, Animal , Edaravone , Fibrosis/prevention & control , Heart Failure/drug therapy , Heart Failure/physiopathology , Macrophages/drug effects , Macrophages/metabolism , Myocardium/pathology , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Peptidyl-Dipeptidase A/genetics , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1/drug effects , Receptor, Angiotensin, Type 1/genetics , Telmisartan
8.
J Renin Angiotensin Aldosterone Syst ; 18(2): 1470320317706653, 2017.
Article in English | MEDLINE | ID: mdl-28490219

ABSTRACT

INTRODUCTION: The purpose of this study was to determine whether macrophages migrated from the spleen are associated with angiotensin II-induced cardiac fibrosis and hypertension. METHODS: Sprague-Dawley rats were subjected to angiotensin II infusion in vehicle (500 ng/kg/min) for up to four weeks. In splenectomy, the spleen was removed before angiotensin II infusion. In the angiotensin II AT1 receptor blockade, telmisartan was administered by gastric gavage (10 mg/kg/day) during angiotensin II infusion. The heart and aorta were isolated for Western blot analysis and immunohistochemistry. RESULTS: Angiotensin II infusion caused a significant reduction in the number of monocytes in the spleen through the AT1 receptor-activated monocyte chemoattractant protein-1. Comparison of angiotensin II infusion, splenectomy and telmisartan comparatively reduced the recruitment of macrophages into the heart. Associated with this change, transforming growth factor ß1 expression and myofibroblast proliferation were inhibited, and Smad2/3 and collagen I/III were downregulated. Furthermore, interstitial/perivascular fibrosis was attenuated. These modifications occurred in coincidence with reduced blood pressure. At week 4, invasion of macrophages and myofibroblasts in the thoracic aorta was attenuated and expression of endothelial nitric oxide synthase was upregulated, along with a reduction in aortic fibrosis. CONCLUSIONS: These results suggest that macrophages when recruited into the heart and aorta from the spleen potentially contribute to angiotensin II-induced cardiac fibrosis and hypertension.


Subject(s)
Hypertension/pathology , Macrophages/pathology , Myocardium/pathology , Spleen/pathology , Angiotensin II , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/metabolism , Aorta, Thoracic/pathology , Blood Pressure/drug effects , Cell Proliferation/drug effects , Chemokine CCL2/metabolism , Collagen/metabolism , Fibrosis , Macrophages/drug effects , Male , Monocytes/metabolism , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Myofibroblasts/pathology , Nitric Oxide Synthase Type III/metabolism , Phosphorylation/drug effects , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1/metabolism , Smad Proteins/metabolism , Splenectomy , Transforming Growth Factor beta1/pharmacology
9.
Zhongguo Ying Yong Sheng Li Xue Za Zhi ; 26(3): 257-60, 2010 Aug.
Article in Chinese | MEDLINE | ID: mdl-21038664

ABSTRACT

OBJECTIVE: To investigate the distribution characteristics of autoantibody against beta1 adrenergic receptor (beta1 AR) in the sera of arrhythmia patients and whether the autoantibody could induce arrhythmia. METHODS: Healthy subjects and patients with arrhythmia or coronary artery disease were chosen. The autoantibody against beta1 AR in the sera was screened by enzyme-linked immunosorbent assay (ELISA). IgG in the positive autoantibody sera from arrhythmia patients were purified and administrated to normal rats; then the ECGs were dynamic monitored. RESULTS: The positive rate of autoantibody against beta1 AR in arrhythmia patients was 52.8%, which was significantly higher than that in coronary heart disease group (24%, P < 0.01) and healthy people group (5%, P < 0.01), respectively. Moreover, the autoantibody against beta1 AR could lead to the occurring of arrhythmia in normal rats, most of which were ventricular arrhythmia. CONCLUSION: In the sera of arrhythmia patients, the autoantibody against beta1 AR has a high titer and it could lead to the arrhythmia of rats in vivo.


Subject(s)
Arrhythmias, Cardiac/immunology , Autoantibodies/immunology , Receptors, Adrenergic, beta-1/immunology , Animals , Arrhythmias, Cardiac/etiology , Autoantibodies/blood , Case-Control Studies , Female , Humans , Immunoglobulin G/blood , Male , Middle Aged , Rats
10.
Acta Pharmacol Sin ; 31(10): 1381-8, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20835263

ABSTRACT

AIM: To investigate the association between autoantibodies against angiotensin AT1 receptor (AT1-AAs) and endothelial dysfunction in vivo. METHODS: Rat models with AT1 receptor antibodies (AT1-Abs) were established by active immunization for nine months. Lactate dehydrogenase (LDH) activity was regarded as an indicator of cell necrotic death. Endothelin-1 (ET-1) in the sera of rats was determined and endothelium-dependent vasodilatation was detected in isolated thoracic aorta. Endothelial intercellular adhesion molecule-1 (ICAM-1) expression in aorta endothelium was assessed using confocal microscopy. Coronary artery endothelial ultrastructure was observed. RESULTS: IgGs in the immunized group significantly increased the LDH activity (0.84±0.17 vs 0.39±0.12, P<0.01 vs vehicle group IgGs)in incubated human umbilical vein endothelial cells through AT1 receptor. Higher content of ET-1 occurred in the immunized rats than that of the vehicle group, and reached two peaks at month 3 (27±4 ng/L, P<0.01) and month 7 (35±5 ng/L, P<0.01), respectively. In addition, aortic endothelium-dependent vasodilatation was attenuated; endothelial ICAM-1 level was markedly increased and cardiac capillary endothelium was damaged following immunization. CONCLUSION: Our study demonstrated that AT1-Abs contributed to endothelial dysfunction in vivo, which was a potential mechanism through which the antibodies play vital roles in related diseases.


Subject(s)
Autoantibodies/metabolism , Endothelial Cells/pathology , Endothelium, Vascular/pathology , Immunoglobulin G/metabolism , Receptor, Angiotensin, Type 1/immunology , Animals , Aorta, Thoracic/pathology , Aorta, Thoracic/physiopathology , Capillaries/pathology , Coronary Vessels/pathology , Endothelial Cells/metabolism , Endothelin-1/metabolism , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Enzyme Activation , Humans , Immunization , In Vitro Techniques , Intercellular Adhesion Molecule-1/metabolism , L-Lactate Dehydrogenase/metabolism , Male , Rats , Rats, Wistar , Receptor, Angiotensin, Type 1/metabolism , Umbilical Veins/metabolism , Vasodilation
SELECTION OF CITATIONS
SEARCH DETAIL
...