Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 98
Filter
1.
Cardiovasc Res ; 116(3): 708-720, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31241138

ABSTRACT

AIMS: Adventitial vasa vasorum provides oxygen and nourishment to the vascular wall, but whether it regulates vascular disease remains unclear. We have previously shown that an increased expression of VEGF (vascular endothelial growth factor) is associated with macrophage infiltration. This study aims to determine whether adventitial fibroblast (AF)-derived VEGF increases the number of vasa vasorum contributing to neointima formation through macrophage recruitment. METHODS AND RESULTS: In rat balloon injury model, vasa vasorum count was increased particularly in the adventitia accompanied by cell proliferation and VEGF expression. Both endogenous and PKH26-labelled exogenous macrophages were mainly distributed in adventitia around vasa vasorum. Interestingly, perivascular delivery of Ranibizumab preferentially concentrated in adventitia resulted in a decrease of neointima formation with concurrent reduction of vasa vasorum count and macrophage infiltration. AFs with adenovirus-mediated VEGF over-expression delivered to the adventitia significantly enhanced these pathological changes after injury. In Tie2-cre/Rosa-LoxP-RFP mice, endothelial cells were increased in the adventitia after wire injury. By using multiphoton laser scanning microscopy, macrophage rolling, adhesion and transmigration were observed in vasa vasorum. Moreover, adoptive transfer of macrophages accelerated injury-induced neointima formation. VEGF-neutralizing antibody administration also attenuated wire injury-induced neointima formation and macrophage infiltration. In primary cultured AFs, exogenous VEGF increased VEGF expression and secretion in a time- and dose-dependent manner. AF-conditioned medium promoted endothelial cell angiogenesis, vascular cell adhesion molecule-1 expression and macrophage adhesion was blocked by VEGF-neutralizing antibody and VEGFR2 inhibitor ZM323881, which also inhibited activation of VEGFR2/ERK1/2 pathway. CONCLUSION: These results demonstrate that AF-derived VEGF plays a significant role in the increase of vasa vasorum count which is involved in macrophage recruitment and neointima formation.


Subject(s)
Adventitia/metabolism , Carotid Arteries/metabolism , Carotid Artery Injuries/metabolism , Femoral Artery/metabolism , Fibroblasts/metabolism , Macrophages/metabolism , Neointima , Vasa Vasorum/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular System Injuries/metabolism , Adoptive Transfer , Adventitia/drug effects , Adventitia/pathology , Angiogenesis Inhibitors/pharmacology , Animals , Carotid Arteries/drug effects , Carotid Arteries/pathology , Carotid Artery Injuries/genetics , Carotid Artery Injuries/pathology , Carotid Artery Injuries/prevention & control , Cells, Cultured , Disease Models, Animal , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Femoral Artery/drug effects , Femoral Artery/pathology , Fibroblasts/drug effects , Fibroblasts/pathology , Macrophages/drug effects , Macrophages/pathology , Macrophages/transplantation , Male , Mice, Inbred C57BL , Paracrine Communication , Rats, Sprague-Dawley , Signal Transduction , Tissue Culture Techniques , Vasa Vasorum/drug effects , Vasa Vasorum/pathology , Vascular Endothelial Growth Factor A/genetics , Vascular System Injuries/genetics , Vascular System Injuries/pathology , Vascular System Injuries/prevention & control
2.
Aging Cell ; 18(4): e12969, 2019 08.
Article in English | MEDLINE | ID: mdl-31087498

ABSTRACT

Aging is an independent risk factor for vascular diseases. Perivascular adipose tissue (PVAT), an active component of the vasculature, contributes to vascular dysfunction during aging. Identification of underlying cell types and their changes during aging may provide meaningful insights regarding the clinical relevance of aging-related vascular diseases. Here, we take advantage of single-cell RNA sequence to characterize the resident stromal cells in the PVAT (PVASCs) and identified different clusters between young and aged PVASCs. Bioinformatics analysis revealed decreased endothelial and brown adipogenic differentiation capacities of PVASCs during aging, which contributed to neointimal hyperplasia after perivascular delivery to ligated carotid arteries. Mechanistically, in vitro and in vivo studies both suggested that aging-induced loss of peroxisome proliferator-activated receptor-γ coactivator-1 α (PGC1α) was a key regulator of decreased brown adipogenic differentiation in senescent PVASCs. We further demonstrated the existence of human PVASCs (hPVASCs) and overexpression of PGC1α improved hPVASC delivery-induced vascular remodeling. Our finding emphasizes that differentiation capacities of PVASCs alter during aging and loss of PGC1α in aged PVASCs contributes to vascular remodeling via decreased brown adipogenic differentiation.


Subject(s)
Adipose Tissue, Brown/cytology , Aging/physiology , Mesenchymal Stem Cells/metabolism , Vascular Remodeling/physiology , Adipogenesis/genetics , Adult , Aged , Animals , Coronary Artery Bypass , Female , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Mice, Transgenic , Middle Aged , Neointima/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Transcriptome
3.
FASEB J ; 33(5): 6254-6268, 2019 05.
Article in English | MEDLINE | ID: mdl-30776250

ABSTRACT

Krüppel-like factor (KLF) 15 has emerged as a critical regulator of fibrosis in cardiovascular diseases. However, the precise role that KLF15 and its functional domain played in adventitial inflammation and fibrosis remains unclear. This study aims to investigate the role of the transactivation domain (TAD) of KLF15 in angiotensin II (Ang II)-induced adventitial pathologic changes. KLF15 expression was decreased in the vascular adventitia of Ang II-infused mice (1000 ng/kg/min, 14 d) and in adventitial fibroblasts (AFs) stimulated by Ang II (10-7 M). Adenovirus-mediated KLF15 overexpression normalized Ang II-induced vascular hypertrophy, increased collagen deposition, macrophage infiltration, and CCL2 and VCAM-1 expression. Interestingly, KLF15-ΔTAD (KLF15 with deletion of TAD at amino acids 132-152) overexpression showed no effect on the above pathologic changes. Similarly, perivascularly overexpression of KLF15 but not KLF15-ΔTAD in carotid arteries also attenuated Ang II-induced vascular inflammation and fibrosis. Furthermore, KLF15 overexpression after Ang II infusion rescued Ang II-induced vascular remodeling. CCL2 or VCAM-1-mediated monocyte and macrophage migration or adhesion to AFs in response to Ang II was negatively regulated by KLF15 through TAD. Ang II-enhanced Smad2/3 activation and adventitial migration, proliferation, and differentiation of AFs were suppressed by KLF15 but not KLF15-ΔTAD overexpression. Conversely, small interfering RNA knockdown of KLF15 aggravated Ang II-induced Smad2/3 activation and dysfunction of AFs. Luciferase, coimmunoprecipitation, and chromatin immunoprecipitation assay were used to demonstrate that interaction of KLF15 with Smad2/3 suppressed CCL2 expression through TAD. Mechanistically, activation of Ang II type 1 receptor/phospholipase Cγ 1/ERK1/2 signaling resulted in a decrease of KLF15 expression. In conclusion, these results demonstrate that KLF15 negatively regulates activation of AFs through TAD, which plays an important role in Ang II-induced adventitial inflammation and fibrosis.-Lu, Y.-Y., Li, X.-D., Zhou, H.-D., Shao, S., He, S., Hong, M.-N., Liu, J.-C., Xu, Y.-L., Wu, Y.-J., Zhu, D.-L., Wang, J.-G., Gao, P.-J. Transactivation domain of Krüppel-like factor 15 negatively regulates angiotensin II-induced adventitial inflammation and fibrosis.


Subject(s)
Adventitia/metabolism , Angiotensin II/metabolism , Fibroblasts/metabolism , Kruppel-Like Transcription Factors/metabolism , Adventitia/pathology , Animals , Cell Movement , Cells, Cultured , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Collagen/metabolism , Fibroblasts/pathology , Fibrosis/metabolism , HEK293 Cells , Humans , Inflammation/metabolism , Kruppel-Like Transcription Factors/chemistry , Kruppel-Like Transcription Factors/genetics , MAP Kinase Signaling System , Macrophages/physiology , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Monocytes/physiology , Protein Domains , RAW 264.7 Cells , Rats , Rats, Sprague-Dawley , Smad Proteins/metabolism , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism
4.
Hypertension ; 72(2): 350-360, 2018 08.
Article in English | MEDLINE | ID: mdl-29915018

ABSTRACT

Endothelial-to-mesenchymal transition (EndoMT) has recently emerged as a potentially important contributor in promoting fibrosis in chronic kidney disease. However, little is known about the role and molecular basis of its involvement in hypertensive renal injury. Here, we aim to determine the role of SIRT (sirtuin) 3 on EndoMT in hypertensive renal injury and to explore its underlying mechanisms. We found that SIRT3 expression was significantly reduced in Ang II (angiotensin II)-induced hypertensive model, accompanied with induction of EndoMT and increased reactive oxygen species and renal fibrosis. In SIRT3-/- (SIRT3 knockout) mice subjected to Ang II infusion, renal dysfunction was aggravated with an increased EndoMT and reactive oxygen species level, whereas in SIRT3-TgEC (SIRT3 endothelial cell-specific transgenic) mice, the Ang II-induced renal fibrosis and EndoMT and oxidative stress were ameliorated. With primary mouse glomerular endothelial cells, we confirmed that Ang II treatment initiated EndoMT and decreased catalase expression, which were suppressed by SIRT3 overexpression. Using immunoprecipitation, luciferase, and chromatin immunoprecipitation assay, we demonstrated that SIRT3-mediated deacetylation and nuclear localization of Foxo3a (forkhead box O3a) resulted in activated Foxo3a-dependent catalase expression. Moreover, Foxo3a knockdown abolished SIRT3-mediated suppression of EndoMT. In conclusion, these results established the SIRT3-Foxo3a-catalase pathway as a critical factor in the maintenance of endothelial homeostasis and point to an important role of EndoMT in the vascular pathology of renal fibrosis, which may provide a new therapeutic target to impede the progression of hypertensive renal injury.


Subject(s)
Endothelial Cells/metabolism , Gene Expression Regulation , Hypertension/complications , Kidney Diseases/etiology , Oxidative Stress , RNA/genetics , Sirtuin 3/genetics , Animals , Disease Models, Animal , Endothelial Cells/pathology , Hypertension/genetics , Hypertension/metabolism , Kidney Diseases/genetics , Kidney Diseases/metabolism , Male , Mice , Mice, Transgenic , Reactive Oxygen Species/metabolism , Signal Transduction , Sirtuin 3/biosynthesis
5.
Diabetes ; 67(8): 1549-1560, 2018 08.
Article in English | MEDLINE | ID: mdl-29794241

ABSTRACT

Obesity increases the risk of vascular diseases, including aortic aneurysm (AA). Perivascular adipose tissue (PVAT) surrounding arteries are altered during obesity. However, the underlying mechanism of adipose tissue, especially PVAT, in the pathogenesis of AA is still unclear. Here we showed that angiotensin II (AngII) infusion increases the incidence of AA in leptin-deficient obese mice (ob/ob) and high-fat diet-induced obese mice with adventitial inflammation. Furthermore, transcriptome analysis revealed that platelet-derived growth factor-D (PDGF-D) was highly expressed in the PVAT of ob/ob mice. Therefore, we hypothesized that PDGF-D mediates adventitial inflammation, which provides a direct link between PVAT dysfunction and AA formation in AngII-infused obese mice. We found that PDGF-D promotes the proliferation, migration, and inflammatory factors expression in cultured adventitial fibroblasts. In addition, the inhibition of PDGF-D function significantly reduced the incidence of AA in AngII-infused obese mice. More importantly, adipocyte-specific PDGF-D transgenic mice are more susceptible to AA formation after AngII infusion accompanied by exaggerated adventitial inflammatory and fibrotic responses. Collectively, our findings reveal a notable role of PDGF-D in the AA formation during obesity, and modulation of this cytokine might be an exploitable treatment strategy for the condition.


Subject(s)
Aorta, Abdominal/metabolism , Aortic Aneurysm, Abdominal/etiology , Intra-Abdominal Fat/metabolism , Lymphokines/metabolism , Obesity/physiopathology , Platelet-Derived Growth Factor/metabolism , Adventitia/drug effects , Adventitia/immunology , Adventitia/metabolism , Adventitia/pathology , Angiotensin II/administration & dosage , Angiotensin II/adverse effects , Animals , Aorta, Abdominal/diagnostic imaging , Aorta, Abdominal/drug effects , Aorta, Abdominal/pathology , Aortic Aneurysm, Abdominal/diagnostic imaging , Aortic Aneurysm, Abdominal/metabolism , Aortic Aneurysm, Abdominal/pathology , Benzimidazoles/pharmacology , Cells, Cultured , Diet, High-Fat/adverse effects , Drug Implants , Gene Expression Regulation/drug effects , Inflammation Mediators/metabolism , Intra-Abdominal Fat/drug effects , Intra-Abdominal Fat/immunology , Intra-Abdominal Fat/pathology , Lymphokines/agonists , Lymphokines/antagonists & inhibitors , Lymphokines/genetics , Male , Mice , Mice, Mutant Strains , Mice, Transgenic , Obesity/etiology , Obesity/metabolism , Obesity/pathology , Organ Specificity , Platelet-Derived Growth Factor/agonists , Platelet-Derived Growth Factor/antagonists & inhibitors , Platelet-Derived Growth Factor/genetics , Quinolines/pharmacology , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Subcutaneous Fat, Abdominal/drug effects , Subcutaneous Fat, Abdominal/immunology , Subcutaneous Fat, Abdominal/metabolism , Subcutaneous Fat, Abdominal/pathology , Survival Analysis
6.
Circ Res ; 122(7): 970-983, 2018 03 30.
Article in English | MEDLINE | ID: mdl-29437833

ABSTRACT

RATIONALE: Inflammation and immunity play crucial roles in the development of hypertension. Complement activation-mediated innate immune response is involved in the regulation of hypertension and target-organ damage. However, whether complement-mediated T-cell functions could regulate blood pressure elevation in hypertension is still unclear. OBJECTIVE: We aim to determine whether C3aR (complement component 3a receptor) and C5aR (complement component 5a receptor) could regulate blood pressure via modulating regulatory T cells (Tregs). METHODS AND RESULTS: We showed that angiotensin II (Ang II)-induced hypertension resulted in an elevated expression of C3aR and C5aR in Foxp3 (forkhead box P3)+ Tregs. By using C3aR and C5aR DKO (double knockout) mice, we showed that C3aR and C5aR deficiency together strikingly decreased both systolic and diastolic blood pressure in response to Ang II compared with WT (wild type), single C3aR-deficient (C3aR-/-), or C5aR-deficient (C5aR-/-) mice. Flow cytometric analysis showed that Ang II-induced Treg reduction in the kidney and blood was also blocked in DKO mice. Histological analysis indicated that renal and vascular structure remodeling and damage after Ang II treatment were attenuated in DKO mice compared with WT mice. In vitro, Ang II was able to stimulate C3aR and C5aR expression in cultured CD4+CD25+ natural Tregs. CD3 and CD28 antibody stimuli downregulated Foxp3 expression in WT but not DKO Tregs. More important, depletion of Tregs with CD25 antibody abolished the protective effects against Ang II-induced hypertension and target-organ damage in DKO mice. Adoptive transfer of DKO Tregs showed much more profound protective effects against Ang II-induced hypertension than WT Treg transfer. Furthermore, we demonstrated that C5aR expression in Foxp3+ Tregs was higher in hypertensive patients compared with normotensive individuals. CONCLUSIONS: C3aR and C5aR DKO-mediated Treg function prevents Ang II-induced hypertension and target-organ damage. Targeting C3aR and C5aR in Tregs specifically may be an alternative novel approach for hypertension treatment.


Subject(s)
Hypertension/immunology , Receptor, Anaphylatoxin C5a/deficiency , Receptors, Complement 3b/deficiency , T-Lymphocytes, Regulatory/immunology , Angiotensin II/toxicity , Animals , Cells, Cultured , Hypertension/etiology , Hypertension/genetics , Male , Mice , Mice, Inbred BALB C
7.
Sci Rep ; 7: 40253, 2017 01 16.
Article in English | MEDLINE | ID: mdl-28091516

ABSTRACT

Vascular calcification (VC) is a highly regulated ectopic mineral deposition process involving immune cell infiltration in the vasculatures, which has been recognized to be promoted by hypertension. The matricellular glycoprotein osteopontin (OPN) is strongly induced in myeloid cells as a potential inflammatory mediator of vascular injury. This study aims to examine whether OPN is involved in the regulation of macrophage activation and osteoclast formation in hypertensive subjects with VC. We firstly found an increased proportion of CD11c+CD163- pro-inflammatory peripheral monocytes in hypertensive subjects with VC compared to those without VC by flow cytometric analysis. Primary cultured macrophages from hypertensive subjects with VC also showed altered expression profile of inflammatory factors and higher serum OPN level. Exogenous OPN promoted the differentiation of peripheral monocytes into an alternative, anti-inflammatory phenotype, and inhibited macrophage-to-osteoclast differentiation from these VC patients. In addition, calcified vessels showed increased osteoclasts accumulation accompanied with decreased macrophages infiltration in the of hypertensive subjects. Taken together, these demonstrated that OPN exerts an important role in the monocytes/macrophage phenotypic differentiation from hypertensive patients with VC, which includes reducing inflammatory factor expression and attenuating osteoclast formation.


Subject(s)
Hypertension/immunology , Inflammation Mediators/immunology , Macrophage Activation , Osteoclasts/immunology , Osteopontin/immunology , Vascular Calcification/immunology , Cell Differentiation , Cells, Cultured , Female , Humans , Hypertension/complications , Hypertension/metabolism , Inflammation Mediators/metabolism , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , Osteoclasts/metabolism , Osteopontin/metabolism , RNA, Messenger/metabolism , Vascular Calcification/complications , Vascular Calcification/metabolism
8.
FASEB J ; 31(3): 1120-1129, 2017 03.
Article in English | MEDLINE | ID: mdl-27974594

ABSTRACT

Perivascular adipose tissue (PVAT)-derived adiponectin (APN) is a secreted adipokine that protects against hypertension-related cardiovascular injury. However, the regulation of APN expression in hypertension remains to be explored. In this study, we demonstrated that down-regulation of APN was associated with complement activation in the PVAT of desoxycorticosterone acetate (DOCA)-salt hypertensive mice. Complement 3-deficient hypertensive mice were protected from ANP decrease in the PVAT. APN deficiency blockaded the protective effects of complement inhibition against hypertensive vascular injury. Mechanistically, complement 5a (C5a)-induced TNF-α secretion from macrophages is required for inhibiting APN expression in adipocytes. Macrophage depletion reversed C5a agonist peptide-induced TNF-α up-regulation and APN down-regulation in the PVAT of DOCA mice. Moreover, we detected increased macrophage infiltration and C5a expression associated with decreased APN expression in adipose tissue from patients with aldosterone-producing adenoma. These results identify a novel interaction between macrophages and adipocytes in the PVAT, where complement-mediated inhibition of APN acts as a potential risk factor for hypertensive vascular inflammation.-Ruan, C.-C., Ma, Y., Ge, Q., Li, Y., Zhu, L.-M., Zhang, Y., Kong, L.-R., Wu, Q-H., Li, F., Cheng, L., Zhao, A. Z., Zhu, D.-L., Gao, P.-J. Complement-mediated inhibition of adiponectin regulates perivascular inflammation and vascular injury in hypertension.


Subject(s)
Adipocytes/metabolism , Adiponectin/metabolism , Complement C3/metabolism , Complement C5a/metabolism , Hypertension/metabolism , Vascular Remodeling , Adiponectin/genetics , Animals , Down-Regulation , Humans , Hypertension/pathology , Inflammation/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Tumor Necrosis Factor-alpha/metabolism
9.
Peptides ; 86: 85-94, 2016 12.
Article in English | MEDLINE | ID: mdl-27773659

ABSTRACT

Apelin has been proved to be a critical mediator of vascular function and homeostasis. Here, we investigated roles of Apelin in aortic remodeling and fibrosis in rats with transverse aortic constriction (TAC). Male Sprague-Dawley rats were subjected to TAC and then randomized to daily deliver Apelin-13 (50µg/kg) or angiotensin type 1 receptor (AT1) blocker Irbesartan (50mg/kg) for 4 weeks. Pressure overload resulted in myocardial hypertrophy, systolic dysfunction, aortic remodeling and adventitial fibrosis with reduced levels of Apelin in ascending aortas of rat after TAC compared with sham-operated group. These changes were associated with marked increases in levels of miRNA-122, TGFß1, CTGF, NFAT5, LGR4, and ß-catenin. More importantly, Apelin and Irbesartan treatment strikingly prevented TAC-mediated aortic remodeling and adventitial fibrosis in pressure overloaded rats by blocking AT1 receptor and miRNA-122 levels and repressing activation of the CTGF-NFAT5 and LGR4-ß-catenin signaling. In cultured primary rat adventitial fibroblasts, exposure to angiotensin II (100nmolL-1) led to significant increases in cellular migration and levels of TGFß1, CTGF, NFAT5, LGR4 and ß-catenin, which were effectively reversed by pre-treatment with Apelin (100nmolL-1) and miRNA-122 inhibitor (50nmolL-1). In conclusion, Apelin counterregulated against TAC-mediated ascending aortic remodeling and angiotensin II-induced promotion of cellular migration by blocking AT1 receptor and miRNA-122 levels and preventing activation of the TGFß1-CTGF-NFAT5 and LGR4-ß-catenin signaling, ultimately contributing to attenuation of aortic adventitial fibrosis. Our data point to Apelin as an important regulator of aortic remodeling and adventitial fibrosis and a promising target for vasoprotective therapies.


Subject(s)
Adventitia/pathology , Aorta/pathology , Aortic Aneurysm, Thoracic/pathology , Intercellular Signaling Peptides and Proteins/physiology , Animals , Aortic Aneurysm, Thoracic/metabolism , Cardiomegaly/metabolism , Cells, Cultured , Fibrosis , Intercellular Signaling Peptides and Proteins/pharmacology , Male , MicroRNAs/metabolism , Myocardium/metabolism , Myocardium/pathology , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/metabolism , Transcription Factors/metabolism , Vascular Remodeling , Ventricular Remodeling , Wnt Signaling Pathway , beta Catenin/metabolism
10.
Oncotarget ; 7(42): 67828-67840, 2016 Oct 18.
Article in English | MEDLINE | ID: mdl-27661131

ABSTRACT

The sympathetic nervous system interacts with the renin-angiotensin-aldosterone system (RAAS) contributing to cardiovascular diseases. In this study, we sought to determine if renal denervation (RDN) inhibits aldosterone expression and associated cardiovascular pathophysiological changes in angiotensin II (Ang II)-induced hypertension. Bilateral RDN or SHAM operation was performed before chronic 14-day Ang II subcutaneous infusion (200ng/kg/min) in male Sprague-Dawley rats. Bilateral RDN blunted Ang II-induced hypertension and ameliorated the mesenteric vascular dysfunction. Cardiovascular hypertrophy in response to Ang II was significantly attenuated by RDN as shown by histopathology and transthoracic echocardiography. Moreover, Ang II-induced vascular and myocardial inflammation and fibrosis were suppressed by RDN with concurrent decrease in fibronectin and collagen deposition, macrophage infiltration, and MCP-1 expression. Interestingly, RDN also inhibited Ang II-induced aldosterone expression in the plasma, kidney and heart. This was associated with the reduction of calcitonin gene-related peptide (CGRP) in the adrenal gland. Ang II promoted aldosterone secretion which was partly attenuated by CGRP in the adrenocortical cell line, suggesting a protective role of CGRP in this model. Activation of transforming growth factor-ß (TGF-ß)/Smad and mitogen-activated protein kinases (MAPKs) signaling pathway was both inhibited by RDN especially in the heart. These results suggest that the regulation of the renal sympathetic nerve in Ang II-induced hypertension and associated cardiovascular pathophysiological changes is likely mediated by aldosterone, with CGRP involvement.


Subject(s)
Aldosterone/metabolism , Heart/physiopathology , Hypertension/physiopathology , Kidney/metabolism , Aldosterone/blood , Angiotensin II , Animals , Calcitonin Gene-Related Peptide/metabolism , Cell Line, Tumor , Denervation , Humans , Hypertension/chemically induced , Hypertension/metabolism , Kidney/innervation , Male , Rats, Sprague-Dawley , Renin-Angiotensin System/physiology , Signal Transduction/physiology
11.
Peptides ; 79: 49-57, 2016 05.
Article in English | MEDLINE | ID: mdl-27018342

ABSTRACT

Angiotensin-converting enzyme 2 (ACE2) has been shown to prevent atherosclerotic lesions and renal inflammation. However, little was elucidated upon the effects and mechanisms of ACE2 in atherosclerotic kidney fibrosis progression. Here, we examined regulatory roles of ACE2 in renal fibrosis in the apolipoprotein E (ApoE) knockout (KO) mice. The ApoEKO mice were randomized to daily deliver either angiotensin (Ang) II (1.5mg/kg) and/or human recombinant ACE2 (rhACE2; 2mg/kg) for 2 weeks. Downregulation of ACE2 and upregulation of phosphorylated Akt, mTOR and ERK1/2 levels were observed in ApoEKO kidneys. Ang II infusion led to increased tubulointerstitial fibrosis in the ApoEKO mice with greater activation of the mTOR/ERK1/2 signaling. The Ang II-mediated renal fibrosis and structural injury were strikingly rescued by rhACE2 supplementation, associated with reduced mRNA expression of TGF-ß1 and collagen I and elevated renal Ang-(1-7) levels. In cultured mouse kidney fibroblasts, exposure with Ang II (100nmolL(-1)) resulted in obvious elevations in superoxide generation, phosphorylated levels of mTOR and ERK1/2 as well as mRNA levels of TGF-ß1, collagen I and fibronectin 1, which were dramatically prevented by rhACE2 (1mgmL(-1)) or mTOR inhibitor rapamycin (10µmolL(-1)). These protective effects of rhACE2 were eradicated by the Ang-(1-7)/Mas receptor antagonist A779 (1µmolL(-1)). Our results demonstrate the importance of ACE2 in amelioration of kidney fibrosis and renal injury in the ApoE-mutant mice via modulation of the mTOR/ERK signaling and renal Ang-(1-7)/Ang II balance, thus indicating potential therapeutic strategies by enhancing ACE2 action for preventing atherosclerosis and fibrosis-associated kidney disorders.


Subject(s)
Angiotensin I/metabolism , Kidney Diseases/enzymology , Kidney/pathology , MAP Kinase Signaling System , Peptide Fragments/metabolism , Peptidyl-Dipeptidase A/pharmacology , Angiotensin-Converting Enzyme 2 , Animals , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Atherosclerosis/complications , Atherosclerosis/drug therapy , Fibrosis , Kidney/enzymology , Kidney Diseases/drug therapy , Kidney Diseases/etiology , Male , Mice, Inbred C57BL , Mice, Knockout , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/metabolism
12.
FEBS Lett ; 590(6): 769-78, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26910302

ABSTRACT

Beta3 adrenergic receptor (ADRB3) mediates vessel relaxation in the endothelium while it modulates lipolysis in the adipose tissue. However, the function and regulation mechanism of ADRB3 in the perivascular adipose tissue (PVAT), especially in hypertension, is still unclear. We show that ADRB3 protein is upregulated in the PVAT of deoxycorticosterone acetate-salt (DOCA-salt) hypertensive mice, with the characteristics of PVAT browning and increased uncoupling protein 1 (UCP1) expression. Inhibition of ADRB3 with selective antagonist SR59230A caused serious vascular injury in vivo, even though UCP1 expression was downregulated. ADRB3 protein was regulated by let-7b, which was decreased in the PVAT of the DOCA-salt group. These data reveal that ADRB3 in PVAT contributes to vascular function in the progression of hypertension.


Subject(s)
Hypertension/complications , Hypertension/metabolism , Receptors, Adrenergic, beta-3/metabolism , Vascular System Injuries/etiology , Vascular System Injuries/metabolism , 3' Untranslated Regions , 3T3-L1 Cells , Adipose Tissue/blood supply , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Adrenergic beta-3 Receptor Antagonists/pharmacology , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/injuries , Aorta, Thoracic/metabolism , Base Sequence , Binding Sites/genetics , Cells, Cultured , Desoxycorticosterone Acetate , Humans , Hypertension/genetics , Ion Channels/metabolism , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Mitochondrial Proteins/metabolism , Molecular Sequence Data , Propanolamines/pharmacology , Receptors, Adrenergic, beta-3/genetics , Uncoupling Protein 1 , Vascular System Injuries/genetics
13.
J Transl Med ; 13: 255, 2015 Aug 06.
Article in English | MEDLINE | ID: mdl-26245758

ABSTRACT

BACKGROUND: The renin-angiotensin system (RAS) has been implicated in atherosclerotic lesions and progression to chronic kidney diseases. We examined regulatory roles of angiotensin-converting enzyme 2 (ACE2) in the apolipoprotein E (ApoE) knockout (KO) kidneys. METHODS: The 3-month-old wild-type, ApoEKO, ACE2KO and ApoE/ACE2 double-KO (DKO) mice in a C57BL/6 background were used. The ApoEKO mice were randomized to daily deliver either Ang II (1.5 mg/kg) and/or human recombinant ACE2 (rhACE2; 2 mg/kg) for 2 weeks. We examined changes in pro-inflammatory cytokines, renal ultrastructure, and pathological signaling in mouse kidneys. RESULTS: Downregulation of ACE2 and nephrin levels was observed in ApoEKO kidneys. Genetic ACE2 deletion resulted in modest elevations in systolic blood pressure levels and Ang II type 1 receptor expression and reduced nephrin expression in kidneys of the ApoE/ACE2 DKO mice with a decrease in renal Ang-(1-7) levels. These changes were linked with marked increases in renal superoxide generation, NADPH oxidase (NOX) 4 and proinflammatory factors levels, including interleukin (IL)-1beta, IL-6, IL-17A, RANTES, ICAM-1, Tumor necrosis factor-alpha (TNF-alpha) and TNFRSF1A. Renal dysfunction and ultrastructure injury were aggravated in the ApoE/ACE2 DKO mice and Ang II-infused ApoEKO mice with increased plasma levels of creatinine, blood urea nitrogen and enhanced levels of Ang II in plasma and kidneys. The Ang II-mediated reductions of renal ACE2 and nephrin levels in ApoEKO mice were remarkably rescued by rhACE2 supplementation, along with augmentation of renal Ang-(1-7) levels. More importantly, rhACE2 treatment significantly reversed Ang II-induced renal inflammation, superoxide generation, kidney dysfunction and adverse renal injury in ApoEKO mice with suppression of the NOX4 and TNF-alpha-TNFRSF1A signaling. However, rhACE2 had no effect on renal NOX2 and TNFRSF1B expression and circulating lipid levels. CONCLUSIONS: ACE2 deficiency exacerbates kidney inflammation, oxidative stress and adverse renal injury in the ApoE-mutant mice through modulation of the nephrin, NOX4 and TNF-alpha-TNFRSF1A signaling. While rhACE2 supplementation alleviates inflammation, renal dysfunction and glomerulus injury in the ApoE-mutant mice associated with upregulations of Ang-(1-7) levels and nephrin expression and suppression of the TNF-alpha-TNFRSF1A signaling. Strategies aimed at enhancing the ACE2/Ang-(1-7) actions may have important therapeutic potential for atherosclerotic renal injury and kidney diseases.


Subject(s)
Apolipoproteins E/deficiency , Gene Deletion , Kidney/pathology , Membrane Proteins/metabolism , Peptidyl-Dipeptidase A/deficiency , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , Angiotensin I/metabolism , Angiotensin II/metabolism , Angiotensin II/pharmacology , Angiotensin-Converting Enzyme 2 , Animals , Apolipoproteins E/metabolism , Humans , Inflammation/pathology , Kidney/drug effects , Kidney/physiopathology , Kidney/ultrastructure , Male , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , NADPH Oxidase 4 , NADPH Oxidases/metabolism , Oxidative Stress/drug effects , Peptide Fragments/metabolism , Peptidyl-Dipeptidase A/pharmacology , Real-Time Polymerase Chain Reaction , Receptor, Angiotensin, Type 1/metabolism , Recombinant Proteins/pharmacology , Signal Transduction/drug effects , Superoxides/metabolism
14.
Arterioscler Thromb Vasc Biol ; 35(7): 1687-95, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25977569

ABSTRACT

OBJECTIVE: To investigate whether rs12731181 (A→G) interrupted miR-590-3p-mediated suppression of the prostaglandin F2α receptor (FP) and whether it is associated with essential hypertension in the Chinese population. APPROACH AND RESULTS: We found that miR-590-3p regulates human FP gene expression by binding to its 3'-untranslated region. rs12731181 (A→G) altered the binding affinity between miR-590-3p and its FP 3'-untranslated region target, thus reducing the suppression of FP expression, which, in turn, enhanced FP receptor-mediated contractility of vascular smooth muscle cells. Overexpression of FP augmented vascular tone and elevated blood pressure in mice. An association study was performed to analyze the relationship between the FP gene and essential hypertension in the Han Chinese population. The results indicated that the rs12731181 G allele was associated with susceptibility to essential hypertension. Carriers of the AG genotype exhibited significantly higher blood pressure than those of the AA genotype. FP gene expression was significantly higher in human peripheral leukocytes from individuals with the AG genotype than that in leukocytes from individuals with the AA genotype. CONCLUSIONS: rs12731181 in the seed region of the miR-590-3p target site is associated with increased risk of essential hypertension and represents a new paradigm for FP involvement in blood pressure regulation.


Subject(s)
Asian People/genetics , Hypertension/genetics , MicroRNAs/genetics , Receptors, Prostaglandin/genetics , 3' Untranslated Regions , Animals , Binding Sites , China/ethnology , Essential Hypertension , Genetic Predisposition to Disease , Humans , Mice , Polymorphism, Single Nucleotide , Transcription, Genetic
15.
Arterioscler Thromb Vasc Biol ; 35(3): 598-606, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25573852

ABSTRACT

OBJECTIVE: We have previously shown an increased expression of complement 3 (C3) in the perivascular adipose tissue (PVAT) in the deoxycorticosterone acetate (DOCA)-salt hypertensive model. This study aims to examine the role and underlying mechanism of C3 in PVAT for understanding the pathogenesis of hypertensive vascular remodeling further. APPROACH AND RESULTS: The role of C3 in macrophage polarization was investigated using peritoneal macrophages from wild-type and C3-deficient (C3KO) mice because we found that C3 was primarily expressed in macrophages in PVAT of blood vessels from DOCA-salt mice, and results showed a decreased expression of M1 phenotypic marker in contrast to an increased level of M2 marker in the C3KO macrophages. Bone marrow transplantation studies further showed in vivo that DOCA-salt recipient mice had fewer M1 but more M2 macrophages in PVAT when the donor bone marrows were from C3KO compared with those from wild-type mice. Of note, this macrophage polarization shift was accompanied with an ameliorated vascular injury. Furthermore, we identified the complement 5a (C5a) as the major C3 activation product that was involved in macrophage polarization and DOCA-salt-induced vascular injury. Consistently, in vivo depletion of macrophages prevented the induction of C3 and C5a in PVAT, and ameliorated hypertensive vascular injury as well. CONCLUSIONS: The presence and activation of bone marrow-derived macrophages in PVAT are crucial for complement activation in hypertensive vascular inflammation, and C5a plays a critical role in DOCA-salt-induced vascular injury by stimulating macrophage polarization toward a proinflammatory M1 phenotype in PVAT.


Subject(s)
Adipose Tissue/metabolism , Complement C3/metabolism , Complement C5a/metabolism , Desoxycorticosterone Acetate , Hypertension/metabolism , Macrophages, Peritoneal/metabolism , Vascular Diseases/metabolism , Vascular Remodeling , 3T3-L1 Cells , Adipocytes/immunology , Adipocytes/metabolism , Adipose Tissue/immunology , Animals , Bone Marrow Transplantation , Cell Communication , Complement Activation , Complement C3/deficiency , Complement C3/genetics , Disease Models, Animal , Hypertension/chemically induced , Hypertension/genetics , Hypertension/immunology , Hypertension/pathology , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Signal Transduction , Time Factors , Vascular Diseases/chemically induced , Vascular Diseases/genetics , Vascular Diseases/immunology , Vascular Diseases/pathology , Vascular Diseases/prevention & control
16.
Hypertension ; 65(3): 622-8, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25624344

ABSTRACT

Recent studies have shown that somatic mutations in the KCNJ5, ATP1A1, ATP2B3, and CACNA1D genes are associated with the pathogenesis of aldosterone-producing adenoma. Clinical profile and biochemical characteristics of the mutations in Chinese patients with aldosterone-producing adenoma remain unclear. In this study, we performed DNA sequencing in 168 Chinese patients with aldosterone-producing adenoma and found 129 somatic mutations in KCNJ5, 4 in ATP1A1, 1 in ATP2B3, and 1 in CACNA1D. KCNJ5 mutations were more prevalent in female patients and were associated with larger adenomas, higher aldosterone excretion, and lower minimal serum K(+) concentration. More interestingly, we identified a novel somatic KCNJ5 mutation (c.445-446insGAA, p.T148-T149insR) that could enhance CYP11B2 mRNA upregulation and aldosterone release. This mutation could also cause membrane depolarization and intercellular Ca(2+) increase. In conclusion, somatic KCNJ5 mutations are conspicuously more popular than mutations of other genes in aldosterone-producing adenomas of Chinese patients. The T148-T149insR mutation in KCNJ5 may influence K(+) channel selectivity and autonomous aldosterone production.


Subject(s)
Adrenal Cortex Neoplasms/genetics , Adrenal Cortex Neoplasms/metabolism , Adrenocortical Adenoma/genetics , Adrenocortical Adenoma/metabolism , Aldosterone/metabolism , Asian People/genetics , Mutation/genetics , Phenotype , Adult , Base Sequence , Calcium Channels, L-Type/genetics , Female , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , Humans , Male , Middle Aged , Molecular Sequence Data , Plasma Membrane Calcium-Transporting ATPases/genetics , Potassium/blood , Prevalence , Retrospective Studies , Sex Factors , Sodium-Potassium-Exchanging ATPase/genetics , Tumor Burden
17.
Ren Fail ; 36(5): 666-72, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24575880

ABSTRACT

A positive association between inflammation and chronic kidney disease (CKD) has been reported but the impact of hypertension on this relation remains unclear. The aim of this study is to investigate the association of various inflammation markers with risk of CKD in hypertensive patients. 387 hypertensive patients (mean age 55.5 years) were recruited. Serum matrix metalloproteinase-2 (MMP-2), matrix metalloproteinase-9 (MMP-9), tissue inhibitor of metalloproteinase-1(TIMP-1), high-sensitivity C-reactive protein (hsCRP) and osteopontin (OPN) were measured by ELISA. CKD was diagnosed either as evidence of kidney damage, including microalbuminuria, or by low glomerular filtration rate (GFR) (<60 mL/min/1.73 m(2)), which was estimated using the Modification of Diet in Renal Disease (MDRD) abbreviated equation. Compared with the reference groups (eGFR ≥ 60 mL/min/1.73 m(2)), the serum levels of TIMP-1, OPN, hsCRP were significantly higher, and the MMP-9/TIMP-1 ratio was lower in the risk group (eGFR < 60 mL/min/1.73 m(2)). Multiple logistic regression analysis showed that TIMP-1, MMP-9/TIMP-1 ratio, OPN and hsCRP were associated with low GFR separately after adjustment, whereas MMP-9/TIMP-1 ratio, OPN and hsCRP were associated with microalbuminuria. The significant association of MMP-9/TIMP-1 ratio and OPN with low GFR and microalbuminuria persisted after additional adjustment for other studied inflammatory biomarkers. Our data suggest that inflammation is strongly and independently associated with renal damage in hypertensive patients. MMP-9/TIMP-1 ratio and OPN may serve as novel risk factors and therapeutic targets for the treatment of CKD in hypertensive patients.


Subject(s)
Biomarkers/blood , Hypertension/complications , Inflammation/blood , Renal Insufficiency, Chronic/blood , Aged , C-Reactive Protein/metabolism , Female , Humans , Hypertension/blood , Male , Matrix Metalloproteinase 2/blood , Matrix Metalloproteinase 9/blood , Middle Aged , Osteopontin/blood , Regression Analysis , Renal Insufficiency, Chronic/diagnosis , Tissue Inhibitor of Metalloproteinase-1/blood
18.
Hypertension ; 63(1): 61-7, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24144646

ABSTRACT

It is unclear at what time-window of the day blood pressure (BP) is most closely associated with cerebrovascular damage. In this cross-sectional study, we examined the strength of association between intracranial arterial stenosis (ICAS) and systolic BP (SBP) across different time-windows using 24-hour ambulatory BP monitoring in 757 consecutively recruited patients with hypertension. ICAS was diagnosed with computerized tomographic angiography in 127 (16.8%) patients, of whom 64 (50.4%) had stenosis ≥50% and 82 (64.6%) had ICAS in ≥2 vessels. Patients with ICAS (142 mm Hg), especially of multiple vessels (145 mm Hg), compared with patients without ICAS (126 mm Hg), had significantly (P<0.001) higher early morning (05:00-07:59 am) SBP. The differences remained significant (P≤0.015) after adjustment for cardiovascular risk factors and SBPs at other time-windows of the day. Multivariate regression analysis showed that consecutive 3-hourly mean SBPs during the day were significantly associated with ICAS (odds ratio for each 10-mm Hg increase, 1.28-1.38; P≤0.001). However, only mean SBP obtained between 05:00 am and 07:59 am remained significant for ICAS (odds ratio, 1.30; P=0.019) when all consecutive 3-hourly mean SBPs were forced into a single multivariate model. In conclusion, the present study showed a significant association between early morning SBP and asymptomatic ICAS in patients with hypertension after accounting for conventional cardiovascular risk factors. Our findings highlight the importance of morning SBP as a cardiovascular risk factor and should be validated in prospective studies.


Subject(s)
Circadian Rhythm , Hypertension/physiopathology , Intracranial Arterial Diseases/physiopathology , Aged , Blood Pressure/physiology , Blood Pressure Monitoring, Ambulatory , Brain/blood supply , Cerebral Angiography , Cerebrovascular Circulation , Constriction, Pathologic , Cross-Sectional Studies , Female , Humans , Intracranial Arterial Diseases/diagnostic imaging , Male , Middle Aged , Systole , Tomography, X-Ray Computed
19.
Chin Med J (Engl) ; 126(21): 4072-7, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24229676

ABSTRACT

BACKGROUND: Hypertension is an important issue in Asia, responsible for up to 66% of cardiovascular disease cases. This randomized controlled trial subgroup analysis compared telmisartan 80 mg (T80)/hydrochlorothiazide 25 mg (H25) singlepill combination with T80 monotherapy, specifically in Chinese and Korean patients. METHODS: Patients with grade 2/3 hypertension were randomized to receive telmisartan 40 mg (T40)/hydrochlorothiazide 12.5 mg (H12.5) combination or T40 monotherapy for one week, before uptitrating the dose to T80/H25 or T80, respectively, for the remaining 6 weeks. The primary endpoint was systolic blood pressure (SBP) mean change from baseline. Secondary endpoints included mean diastolic blood pressure (DBP) change from baseline, and blood pressure (BP) goal achievement. Adverse events were recorded. RESULTS: Of a total 888 patients who were treated, efficacy analyses for Chinese and Korean patients included 127 patients treated with T80/H25 and 54 patients treated with T80. At week 7, mean SBP reductions from baseline were -37.5 mmHg (1 mmHg = 0.133 kPa) and -26.9 mmHg in the T80/H25 and T80 groups (adjusted mean difference, -10.6 mmHg; 95% confidence interval (CI), -15.6 to -5.7). Mean DBP reductions were -19.0 and -14.1 mmHg in the T80/H25 and T80 groups (adjusted mean difference, -4.9 mmHg; 95% CI, -8.0 to -1.8). In total, 56.7% of patients receiving T80/H25 achieved BP goal (<140/90 mmHg) compared with 35.2% receiving T80. SBP goal attainment (<140 mmHg) and DBP goal attainment (<90 mmHg) were also higher in the T80/H25 group compared with the T80 group (SBP: 69.3% vs. 48.1%; DBP: 62.2% vs. 46.3%). A small number of treatment-related adverse events were observed in both T80/H25 (nine patients, 6.9%) and T80 monotherapy (two patients, 3.6%) groups. CONCLUSIONS: In Chinese and Korean patients with moderate-to-severe hypertension, treatment with T80/H25 provided large reductions in mean SBP and DBP, and high BP goal attainment rates. This once-daily combination is effective and well tolerated in this patient group.


Subject(s)
Benzimidazoles/therapeutic use , Benzoates/therapeutic use , Hydrochlorothiazide/therapeutic use , Hypertension/drug therapy , Adult , Aged , Antihypertensive Agents , Blood Pressure/drug effects , Double-Blind Method , Female , Humans , Male , Middle Aged , Telmisartan , Treatment Outcome
20.
Circ J ; 77(12): 2997-3006, 2013.
Article in English | MEDLINE | ID: mdl-24161906

ABSTRACT

BACKGROUND: Angiotensin-converting enzyme 2 (ACE2) has been implicated in human heart failure, but the mechanism remains elusive. We hypothesized that ACE2 deficiency would exacerbate angiotensin (Ang) II-mediated myocardial injury. METHODS AND RESULTS: 10-week-old ACE2 knockout (ACE2KO) and wild-type mice received by mini-osmotic pump either AngII (1.5 mg·kg(-1)·day(-1)) or saline for 2 weeks. ACE2 deficiency triggered greater increases in the expression of connective tissue growth factor (CTGF), fractalkine (FKN) and phosphorylated ERK1/2 in AngII-treated ACE2KO hearts. These changes were associated with greater activation of matrix metalloproteinase (MMP) 2, MMP9 and MT1-MMP and exacerbation of myocardial injury and dysfunction. In cultured cardiofibroblasts, exposure to AngII (100 nmol/L) for 30 min resulted in marked increases in superoxide production and expression of CTGF, FKN and phosphorylated ERK1/2, which were strikingly prevented by recombinant human ACE2 (rhACE2; 1mg/ml) and the CTGF-neutralizing antibody (5 µg/ml), but were aggravated by ACE2 inhibitor DX600 (0.5 µmol/L). These protective effects of rhACE2 were eradicated by the Ang-(1-7) antagonist A779 (1 µmol/L). More intriguingly, rhACE2 treatment significantly abolished AngII-mediated increases in MMP2, MMP9 and MT1-MMP in cardiofibroblasts. CONCLUSIONS: Loss of ACE2 exacerbates AngII-mediated inflammation, myocardial injury and dysfunction in ACE2-deficient hearts via activation of the CTGF-FKN-ERK and MMP signaling. ACE2 gene may represent a potential candidate to prevent and treat myocardial injury and heart diseases.


Subject(s)
Chemokine CX3CL1/biosynthesis , Connective Tissue Growth Factor/biosynthesis , Heart Injuries/metabolism , MAP Kinase Signaling System , Myocardium/metabolism , Peptidyl-Dipeptidase A/metabolism , Angiotensin II/adverse effects , Angiotensin II/pharmacology , Angiotensin-Converting Enzyme 2 , Animals , Chemokine CX3CL1/genetics , Connective Tissue Growth Factor/genetics , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Heart Injuries/chemically induced , Heart Injuries/genetics , Heart Injuries/pathology , Humans , Matrix Metalloproteinase 14/biosynthesis , Matrix Metalloproteinase 14/genetics , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/biosynthesis , Matrix Metalloproteinase 9/genetics , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Myocardium/pathology , Peptidyl-Dipeptidase A/genetics , Phosphorylation/drug effects , Phosphorylation/genetics , Vasoconstrictor Agents/adverse effects , Vasoconstrictor Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL