Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Antioxidants (Basel) ; 9(10)2020 Oct 06.
Article in English | MEDLINE | ID: mdl-33036258

ABSTRACT

Vascular calcification (VC) is highly prevalent in patients with atherosclerosis, chronic kidney disease, diabetes mellitus, and hypertension. In blood vessels, VC is associated with major adverse cardiovascular events. Xanthohumol (XN), a main prenylated chalcone found in hops, has antioxidant effects to inhibit VC. This study aimed to investigate whether XN attenuates VC through in vivo study. A rat VC model was established by four weeks oral administration of vitamin D3 plus nicotine in Sprague Dawley (SD) rats. In brief, 30 male SD rats were randomly divided into three groups: control, 25 mg/kg nicotine in 5 mL corn oil and 3 × 105 IU/kg vitamin D3 administration (VDN), and combination of VDN with 20 mg/L in 0.1% ethanol of XN (treatment group). Physiological variables such as body and heart weight and drinking consumption were weekly observed, and treatment with XN caused no differences among the groups. In comparison with the control group, calcium content and alkaline phosphatase (ALP) activity were increased in calcified arteries, and XN treatment reduced these levels. Dihydroethidium (DHE) and 2',7'-dichloroflurescin diacetate (DCFH-DA) staining to identify Superoxide and reactive oxygen species generation from aorta tissue showed increased production in VDN group compared with the control and treatment groups. Hematoxylin eosin (HE) and Alizarin Red S staining were determined to show medial vascular thickness and calcification of vessel wall. Administration of VDN resulted in VC, and XN treatment showed improvement in vascular structure. Moreover, overexpression of osteogenic transcription factors bone morphogenetic protein 2 (BMP-2) and runt-related transcription factor 2 (Runx2) were significantly suppressed by XN treatment in VC. Moreover, downregulation of vascular phenotypic markers alpha-smooth muscle actin (α-SMA) and smooth muscle 22 alpha (SM22α) were increased by XN treatment in VC. Furthermore, XN treatment in VC upregulated nuclear translocation of nuclear factor-E2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) expressions. Otherwise, Kelch-like ECH-associated protein 1 (Keap1) was alleviated by XN treatment in VC. In conclusion, our findings suggested that XN enhances antioxidant capacity to improve VC by regulating the Nrf2/Keap1/HO-1 pathway. Therefore, XN may have potential effects to decrease cardiovascular risk by reducing VC.

2.
J Tradit Complement Med ; 10(4): 378-388, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32695655

ABSTRACT

BACKGROUND AND AIM: Centella asiatica, Justicia gendarussa and Imperata cylindrica decoction (CJID) is efficacious for hypertension. NADPH (nicotinamide adenine dinucleotide phosphate) oxidase (NOX)-induced reactive oxygen species (ROS) generation modulates nuclear factor kappa B (NF-κB) activation and thus mediates hypertension-induced vascular remodeling. This research aims to investigate the anti-remodeling effect of CJID through the mechanism of NOXs-ROS-NF-κB pathway in spontaneously hypertensive rats (SHRs). EXPERIMENTAL PROCEDURE: CJID was orally administered once a day for five weeks in SHRs and normotensive-WKY (Wistar Kyoto) rats. All rats were sacrificed at the end of study and different assays were performed to determine whether CJID ameliorates vascular remodeling in SHRs, such as histological examination; lactate dehydrogenase (LDH), nitric oxide (NO), malondialdehyde (MDA) and superoxide dismutase (SOD) assays; superoxide and hydrogen peroxide (H2O2) generation assays, immunohistochemistry and immunofluorescence assays. . Changes in levels of inducible nitric oxide synthase (iNOS), NF-κB-p65, NF-κB inhibitor alpha/IκBα (inhibitory kappa B- alpha), phosphorylation of IκBα (p-IκBα) and NOX1, NOX2, NOX4 in the thoracic aorta were determined. RESULTS: Vascular remodeling indicators, media thickness, collagen and elastic accumulation in the thoracic aorta, of SHRs-treated CJID were attenuated. Redox homeostasis, aortic superoxide and hydrogen peroxide generation were decreased in SHRs-treated group. Aortic iNOS, p-IκBα, NF-κB-p65 and NOX1, NOX2, NOX4 expressions were suppressed. CONCLUSIONS: CJI treatment diminishes oxidative stress response in the thoracic aorta of SHRs via regulation of NOXs-ROS-NF-κB signaling pathway. These findings indicate that CJI possess protective effect against hypertension-induced vascular remodeling in SHRs.

3.
Clin Sci (Lond) ; 132(18): 2045-2058, 2018 09 28.
Article in English | MEDLINE | ID: mdl-30219798

ABSTRACT

The physiologic process of postnatal ductus arteriosus (DA) closure consists of vasoconstriction followed by vascular remodeling. We have recently reported that B-type natriuretic peptide (BNP), a potent vasodilator, also has anti-remodeling effects in pulmonary vasculature. However, its effects on DA have not been elucidated. We investigated whether BNP can prevent DA closure, and if so, the underlying mechanisms. Using in vivo studies, we examined effects of BNP (10 mg/kg, ip at birth) on DA closure in neonatal rats within 4 h after birth. We found that in control rats, the DA spontaneously closed at 4 h with a decreased DA diameter, enhanced intimal thickening, and luminal occlusion. BNP prevented DA closure at 4 h with a preserved DA diameter, attenuated intimal thickening, and preserved luminal patency. Ex vivo, BNP attenuated oxygen-induced vasoconstriction of isolated DA rings of newborn rats. These vasodilating effects were blunted by Rp-8-Br-PET-cGMPS, a cGMP inhibitor. In vitro, BNP inhibited angiotensin II (Ang II)-induced proliferation and migration of DA smooth muscle cells (DASMCs). BNP inhibited Ang II-induced mitochondrial reactive oxygen species (ROS) production and calcium overload in DASMCs. Finally, BNP inhibited Ang II-induced ERK1/2 activation. These in vitro effects were antagonized by Rp-8-Br-PET-cGMPS. In conclusion, BNP prevents postnatal DA closure by both vasodilation and anti-remodeling through the cGMP pathway. The mechanisms underlying anti-remodeling effects include anti-poliferation and anti-migration, with attenuation of mitochondrial ROS production and intracellular calcium and ERK1/2 signaling. Therefore, the BNP/cGMP pathway can be a promising therapeutic target for clinical management of DA patency.


Subject(s)
Ductus Arteriosus/drug effects , Natriuretic Peptide, Brain/pharmacology , Vascular Remodeling/drug effects , Vasodilation/drug effects , Angiotensin II/pharmacology , Animals , Animals, Newborn , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Ductus Arteriosus/cytology , Ductus Arteriosus/physiology , MAP Kinase Signaling System/drug effects , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/physiology , Rats, Wistar , Thionucleotides/pharmacology , Time Factors , Vascular Remodeling/physiology , Vasodilation/physiology
4.
Int J Biol Sci ; 12(9): 1063-73, 2016.
Article in English | MEDLINE | ID: mdl-27570480

ABSTRACT

Patent ductus arteriosus (PDA) can cause morbidity and mortality in neonates. Vascular remodeling, characterized by proliferation and migration of smooth muscle cells (SMCs), is an essential process for postnatal DA closure. Notch signaling is an important mediator of vascular remodelling but its role in DA is unkonwn. We investigated the effects and underlying mechanisms of γ-secretase inhibitor DAPT, a Notch signaling inhibitor on angiotensin II (Ang II)-induced proliferation and migration of DASMCs. Proliferation and migration of DASMCs cultured from neonatal Wistar rats were induced by Ang II, with or without DAPT pre-treatment. In addition, potential underlying mechanisms including cell cycle progression, Ca(2+) influx, reactive oxygen species (ROS) production, signal transduction of MAPK and Akt, and Notch receptor with its target gene pathway were examined. We found that DAPT inhibited Ang II-induced DASMCs proliferation and migration dose dependently. DAPT also arrested the cell cycle progression in the G0/G1-phase, and attenuated calcium overload and ROS production caused by Ang II. Moreover, DAPT inhibited nuclear translocation of Notch3 receptor intracellular domain, with decreased expression of its down-stream genes including HES1, HES2 and HES5. Finally, Ang II-activated ERK1/2, JNK and Akt were also counteracted by DAPT. In conclusion, DAPT inhibits Ang II-induced DASMCs proliferation and migration. These effects are potentially mediated by decreased calcium influx, reduced ROS production, and down-regulation of ERK1/2, JNK and Akt, through the Notch3-HES1/2/5 pathway. Therefore, Notch signaling has a role in DA remodeling and may provide a target pathway for therapeutic intervention of PDA.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Diamines/pharmacology , Ductus Arteriosus/cytology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Receptor, Notch3/metabolism , Repressor Proteins/metabolism , Thiazoles/pharmacology , Transcription Factor HES-1/metabolism , Angiotensin II/pharmacology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Female , Myocytes, Smooth Muscle/metabolism , Pregnancy , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Receptor, Notch3/genetics , Receptors, Notch/genetics , Receptors, Notch/metabolism , Repressor Proteins/genetics , Signal Transduction/drug effects , Transcription Factor HES-1/genetics
5.
J Pharm Pharmacol ; 68(6): 810-8, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27109251

ABSTRACT

OBJECTIVES: Endothelial damage is strongly associated with cardiovascular diseases such as atherosclerosis, thrombosis and hypertension. Endothelial progenitor cells (EPCs) are primitive bone marrow (BM) cells that possess the capacity to mature into endothelial cells and play a role in neovascularization and vascular remodelling. This study aimed to investigate whether KMUP-1, a synthetic xanthine-based derivative, atorvastatin and simvastatin, can prevent endothelial dysfunction and apoptosis induced by hypoxia and to elucidate the underlying mechanisms. METHODS: Mononuclear cells were separated and were induced to differentiate into EPCs. KMUP-1, atorvastatin or simvastatin were administered prior to hypoxia. KEY FINDINGS: We found that EPCs exposed to hypoxia increased apoptosis as well as diminished proliferation. Pretreatment with KMUP-1, atorvastatin and simvastatin significantly prevented hypoxia-induced EPCs death and apoptosis, with associated increased of the Bcl-2/Bax ratio, and reduced caspase-3 and caspase-9 expression. We also assessed the nitrite production and Ser(1177)-phospho-eNOS expression and found that KMUP-1, atorvastatin and simvastatin not only increased the secretion of NO compared with the hypoxia group but also upregulated the eNOS activation. CONCLUSIONS: KMUP-1 inhibited hypoxia-induced dysfunction and apoptosis in EPCs, which may be mediated through suppressing oxidative stress, upregulating eNOS and downregulating the caspase-3 signalling pathway.


Subject(s)
Apoptosis/drug effects , Endothelial Progenitor Cells/drug effects , Enzyme Activators/pharmacology , Nitric Oxide Synthase Type III/metabolism , Piperidines/pharmacology , Xanthines/pharmacology , Animals , Apoptosis Regulatory Proteins/metabolism , Atorvastatin/pharmacology , Cell Hypoxia , Cell Proliferation/drug effects , Cells, Cultured , Cytoprotection , Endothelial Progenitor Cells/enzymology , Endothelial Progenitor Cells/pathology , Enzyme Activation , Nitric Oxide/metabolism , Nitrites/metabolism , Oxidative Stress/drug effects , Phosphorylation , Rats, Sprague-Dawley , Signal Transduction/drug effects , Simvastatin/pharmacology
6.
Kaohsiung J Med Sci ; 32(2): 55-67, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26944323

ABSTRACT

KMUP-3 (7-{2-[4-(4-nitrobenzene) piperazinyl]ethyl}-1, 3-dimethylxanthine) displays cardioprotection and increases cardiac output, and is suggested to increase cardiac performance and improve myocardial infarction. To determine whether KMUP-3 improves outcomes in hypoperfused myocardium by inducing Ca(2+) sensitization to oppose protein kinase (PK)G-mediated Ca(2+) blockade, we measured left ventricular systolic blood pressure, maximal rates of pressure development, mean arterial pressure and heart rate in rats, and measured contractility and expression of PKs/RhoA/Rho kinase (ROCK)II in beating guinea pig left atria. Hemodynamic changes induced by KMUP-3 (0.5-3.0 mg/kg, intravenously) were inhibited by Y27632 [(R)-(+)-trans-4-1-aminoethyl)-N-(4-Pyridyl) cyclohexane carboxamide] and ketanserin (1 mg/kg, intravenously). In electrically stimulated left guinea pig atria, positive inotropy induced by KMUP-3 (0.1-100µM) was inhibited by the endothelial NO synthase (eNOS) inhibitors N-nitro-l-arginine methyl ester (L-NAME) and 7-nitroindazole, cyclic AMP antagonist SQ22536 [9-(terahydro-2-furanyl)-9H-purin-6-amine], soluble guanylyl cyclase (sGC) antagonist ODQ (1H-[1,2,4] oxadiazolo[4,3-a] quinoxalin-1-one), RhoA inhibitor C3 exoenzyme, ß-blocker propranolol, 5-hydroxytryptamine 2A antagonist ketanserin, ROCK inhibitor Y27632 and KMUP-1 (7-{2-[4-(2-chlorobenzene) piperazinyl]ethyl}-1, 3-dimethylxanthine) at 10µM. Western blotting assays indicated that KMUP-3 (0.1-10µM) increased PKA, RhoA/ROCKII, and PKC translocation and CIP-17 (an endogenous 17-kDa inhibitory protein) activation. In spontaneous right atria, KMUP-3 induced negative chronotropy that was blunted by 7-nitroindazole and atropine. In neonatal myocytes, L-NAME inhibited KMUP-3-induced eNOS phosphorylation and RhoA/ROCK activation. In H9c2 cells, Y-27632 (50µM) and PKG antagonist KT5823 [2,3,9,10,11,12-hexahydro-10R- methoxy-2,9-dimethyl-1-oxo-9S,12R-epoxy-1H-diindolo(1,2,3-fg:3',2',1'-kl) pyrrolo(3,4-i)(1,6)benzodiazocine-10-carboxylic acid, methyl ester] (3µM) reversed KMUP-3 (1-100µM)-induced Ca(2+)-entry blockade. GPCR agonist activity of KMUP-3 appeared opposed to KMUP-1, and increased cardiac output via Ca(2+) sensitization, and displayed cardioprotection via cyclic GMP/PKG-mediated myocardial preconditioning in animal studies.


Subject(s)
Cardiac Output/drug effects , Cardiotonic Agents/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Piperidines/pharmacology , Xanthines/pharmacology , Animals , Blood Pressure , Calcium Signaling , Carbazoles/pharmacology , Cell Line , Cyclic GMP-Dependent Protein Kinases/metabolism , Drug Evaluation, Preclinical , Female , Guinea Pigs , Heart Atria/physiopathology , Male , Protein Transport , Rats, Wistar , Receptors, G-Protein-Coupled/metabolism , Ventricular Pressure
7.
Molecules ; 20(6): 10435-49, 2015 Jun 05.
Article in English | MEDLINE | ID: mdl-26056815

ABSTRACT

The signaling cascades of the mitogen activated protein kinase (MAPK) family, calcineurin/NFATc4, and PI3K/Akt/GSK3, are believed to participate in endothelin-1 (ET-1)-induced cardiac hypertrophy. The aim of this study was to investigate whether KMUP-1, a synthetic xanthine-based derivative, prevents cardiomyocyte hypertrophy induced by ET-1 and to elucidate the underlying mechanisms. We found that in H9c2 cardiomyocytes, stimulation with ET-1 (100 nM) for 4 days induced cell hypertrophy and enhanced expressions of hypertrophic markers, including atrial natriuretic peptide and brain natriuretic peptide, which were all inhibited by KMUP-1 in a dose-dependent manner. In addition, KMUP-1 prevented ET-1-induced intracellular reactive oxygen species generation determined by the DCFH-DA assay in cardiomyocytes. KMUP-1 also attenuated phosphorylation of ERK1/2 and Akt/GSK-3ß, and activation of calcineurin/NFATc4 and RhoA/ROCK pathways induced by ET-1. Furthermore, we found that the expression of heme oxygenase-1 (HO-1), a stress-response enzyme implicated in cardio-protection, was up-regulated by KMUP-1. Finally, KMUP-1 attenuated ET-1-stimulated activator protein-1 DNA binding activity. In conclusion, KMUP-1 attenuates cardiomyocyte hypertrophy induced by ET-1 through inhibiting ERK1/2, calcineurin/NFATc4 and RhoA/ROCK pathways, with associated cardioprotective effects via HO-1 activation. Therefore, KMUP-1 may have a role in pharmacological therapy of cardiac hypertrophy.


Subject(s)
Calcineurin/metabolism , Endothelin-1/metabolism , Glycogen Synthase Kinase 3/metabolism , Heme Oxygenase-1/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , NFATC Transcription Factors/metabolism , Piperidines/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Xanthines/pharmacology , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Enzyme Activation , Glycogen Synthase Kinase 3 beta , Hypertrophy , Mitogen-Activated Protein Kinases/metabolism , Models, Biological , Myocytes, Cardiac/pathology , Protein Binding , Rats , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Transcription Factor AP-1
8.
J Cell Physiol ; 230(9): 2038-48, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25536014

ABSTRACT

Phosphodiesterase (PDE) inhibitors have been suggested as a possible candidate for the treatment of osteopenia, including osteoporosis. KMUP-1 is a novel xanthine derivative with inhibitory activities on the PDE 3, 4, and 5 iso-enzymes to suppress the degradation of cAMP and cGMP. This study aimed to investigate the effect of KMUP-1 on osteoblast differentiation and the underlying cellular and molecular mechanisms. Primary osteoblasts and osteoblastic MC3T3-E1 cells were examined. KMUP-1 enhanced alkaline phosphatase (ALP) activity and mineralization compared to untreated controls in primary osteoblasts and MC3T3-E1 cells. KMUP-1 also increased the mRNA expression of the osteoblastic differentiation markers, including collagen type 1a, ALP, osteocalcin, osteoprotegerin, BMP-2, and Runx2, a key transcription regulator for osteoblastic differentiation. The osteogenic effect of KMUP-1 was abolished by BMP signaling inhibitor, noggin. Furthermore, we found that KMUP-1 upregulated Smad1/5/8 phosphorylations with subsequent BRE-Luc activation confirmed by transient transfection assay. In addition, KMUP-1 inactivated glycogen synthase kinase-3ß (GSK-3ß), with associated nuclear translocation of ß-catenin. Co-treatment with H89 and KT5823, cAMP and cGMP pathway inhibitors, respectively, reversed the KMUP-1-induced activations of Smad1/5/8, ß-catenin, and Runx2. The findings demonstrate for the first time that KMUP-1 can promote osteoblast maturation and differentiation in vitro via BMP-2/Smad1/5/8 and Wnt/ß-catenin pathways. These effects are mediated, in part, by the cAMP and cGMP signaling. Thus, KMUP-1 may be a novel osteoblast activator and a potential new therapy for osteoporosis.


Subject(s)
Bone Morphogenetic Protein 2/biosynthesis , Cell Differentiation/drug effects , Piperidines/administration & dosage , Smad1 Protein/biosynthesis , Smad5 Protein/biosynthesis , Smad8 Protein/biosynthesis , Xanthines/administration & dosage , Animals , Bone Morphogenetic Protein 2/genetics , Calcification, Physiologic , Cell Line , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Gene Expression Regulation, Developmental/drug effects , Mice , Osteoblasts/drug effects , RNA, Messenger/biosynthesis , Smad1 Protein/genetics , Smad5 Protein/genetics , Smad8 Protein/genetics , Wnt Signaling Pathway
9.
Shock ; 42(6): 540-7, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25243423

ABSTRACT

It has been recently demonstrated that intracellular heat shock cognate protein 70 (HSC70) can be released into extracellular space with physiologic effects. However, its extracellular function in sepsis is not clear. In this study, we hypothesize that extracellular HSC70 can protect against lipopolysaccharide (LPS)-induced myocardial and hepatic dysfunction because of its anti-inflammatory actions. In Wistar rats, septic shock developed with hypotension, tachycardia, and myocardial and hepatic dysfunction at 4 h following LPS administration (10 mg/kg, i.v.). Pretreatment with recombinant bovine HSC70 (20 µg/kg, i.v.) attenuated LPS-induced hypotension and tachycardia by 21% and 23%, respectively (P < 0.05), improved myocardial dysfunction (left ventricular systolic pressure: 33%; max dP/dt: 20%; min dP/dt: 33%, P < 0.05), and prevented hepatic dysfunction (glutamic-oxaloacetic transaminase: 81 vs. 593 IU/L; glutamic-pyruvic transaminase: 15 vs. 136 IU/L, P < 0.05) compared with LPS-treated rats at 4 h. Heat shock cognate protein 70 also prevented LPS-induced hypoglycemia (217 vs. 59 mg/dL, P < 0.05) and elevated lactate dehydrogenase (1,312 vs. 6,301 IU/L, P < 0.05). Furthermore, HSC70 decreased LPS-induced elevation of circulating tumor necrosis factor α and nitrite/nitrate, and tissue expression of inducible nitric oxide synthase, cyclooxygenase 2, and matrix metalloproteinase 9 in the heart and liver. To investigate underlying mechanisms, we found that HSC70 attenuated LPS-induced nuclear translocation of nuclear factor κB subunit p65 by blocking the phosphorylation of inhibitor of nuclear factor κB. Finally, we showed that HSC70 repressed the activation of MAPKs caused by LPS. These results demonstrate that in LPS-induced septic shock, extracellular HSC70 conveys pleiotropic protection on myocardial, hepatic, and systemic derangements, with associated inhibition of proinflammatory mediators including tumor necrosis factor α, nitric oxide, cyclooxygenase 2, and matrix metalloproteinase 9, through mitogen-activated protein kinase/nuclear factor κB signaling pathways. Therefore, extracellular HSC70 may have a promising role in the prophylactic treatment of sepsis.


Subject(s)
HSP70 Heat-Shock Proteins/pharmacology , Liver/physiopathology , Shock, Septic/metabolism , Active Transport, Cell Nucleus , Animals , Cattle , Endotoxemia/metabolism , Gene Expression Regulation , Heart Ventricles/metabolism , Lipopolysaccharides/chemistry , Liver/enzymology , Male , Matrix Metalloproteinase 9/metabolism , Myocardium/enzymology , Nitric Oxide/metabolism , Rats , Rats, Wistar , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/metabolism
10.
BMC Complement Altern Med ; 14: 233, 2014 Jul 09.
Article in English | MEDLINE | ID: mdl-25012390

ABSTRACT

BACKGROUND: Lysophosphatidylcholine (lysoPC), a metabolite from membrane phospholipids, accumulates in the ischemic myocardium and plays an important role in the development of myocardial dysfunction ventricular arrhythmia. In this study, we investigated if baicalein, a major component of Huang Qui, can protect against lysoPC-induced cytotoxicity in rat H9c2 embryonic cardiomyocytes. METHODS: Cell viability was detected by the MTT assay; ROS levels were assessed using DCFH-DA; and intracellular free calcium concentrations were assayed by spectrofluorophotometer. Cell apoptosis and necrosis were evaluated by the flow cytometry assay and Hoechst staining. Mitogen-Activated Protein Kinases (MAPKs), which included the ERK, JNK, and p38, and the apoptotic mechanisms including Bcl-2/Bax, caspase-3, caspase-9 and cytochrome c pathways were examined by Western blot analysis. The activation of MAPKs was examined by enzyme-linked immunosorbent assay. RESULTS: We found that lysoPC induced death and apoptosis of H9c2 cells in a dose-dependent manner. Baicalein could prevent lysoPC-induced cell death, production of reactive oxygen species (ROS), and increase of intracellular calcium concentration in H9c2 cardiomyoctes. In addition, baicalein also inhibited lysoPC-induced apoptosis, with associated decreased pro-apoptotic Bax protein, increased anti-apoptotic Bcl-2 protein, resulting in an increase in the Bcl-2/Bax ratio. Finally, baicalein attenuated lysoPC-induced the expression of cytochrome c, casapase-3, casapase-9, and the phosphorylations of ERK1/2, JNK, and p38. LysoPC-induced ERK1/2, JNK, and p38 activations were inhibited by baicalein. CONCLUSIONS: Baicalein protects cardiomyocytes from lysoPC-induced apoptosis by reducing ROS production, inhibition of calcium overload, and deactivations of MAPK signaling pathways.


Subject(s)
Calcium/metabolism , Flavanones/pharmacology , MAP Kinase Signaling System/drug effects , Myocytes, Cardiac/drug effects , Reactive Oxygen Species/metabolism , Scutellaria baicalensis/chemistry , Animals , Apoptosis/drug effects , Apoptosis/physiology , Apoptosis Regulatory Proteins/metabolism , Cell Line , Cell Survival/drug effects , Lysophosphatidylcholines , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinases/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/metabolism , Rats
11.
Am J Chin Med ; 42(4): 785-97, 2014.
Article in English | MEDLINE | ID: mdl-25004875

ABSTRACT

Myocardial dysfunction, a common complication after sepsis, significantly contributes to the death of patients with septic shock. In the search for potentially effective drugs to decrease mortality from sepsis, we investigated the cardioprotective effects of baicalein, a flavonoid present in the root of Scutellaria baicalensis, on lipopolysaccharide (LPS)-induced pro-inflammatory cytokine production and matrix metalloproteinase-2 and -9 (MMP-2/-9) expression. We found that baicalein significantly attenuated LPS-induced cardiac hypertrophy and counteracted reactive oxygen species (ROS) generation in neonatal rat cardiomyocytes. In addition, pretreatment with baicalein inhibited LPS-induced early (e.g., tumor necrosis factor-α (TNF-α) and interleukin-6) and late (e.g., high mobility group box 1 (HMGB1) pro-inflammatory cytokine release, inducible nitric oxide synthase (iNOS) expression and NO production. Finally, baicalein also significantly down-regulated the expression of MMP-2/-9 and attenuated HMGB1 translocation from the nucleus to the cytoplasm. These results suggest that baicalein can protect cardiomyocytes from LPS-induced cardiac injury via the inhibition of ROS and inflammatory cytokine production. These cardioprotective effects are possibly mediated through the inhibition of the HMGB1 and MMP-2/-9 signaling pathways.


Subject(s)
Antioxidants/pharmacology , Antioxidants/therapeutic use , Cardiomegaly/drug therapy , Cardiomegaly/genetics , Flavanones/pharmacology , Flavanones/therapeutic use , HMGB1 Protein/metabolism , Lipopolysaccharides , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Phytotherapy , Animals , Cardiomegaly/chemically induced , Cardiomegaly/pathology , Cells, Cultured , Down-Regulation/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Nitric Oxide Synthase Type II/metabolism , Protein Biosynthesis/drug effects , Rats, Wistar , Reactive Oxygen Species/metabolism , Scutellaria baicalensis
12.
Pediatr Pulmonol ; 49(8): 734-44, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24167111

ABSTRACT

Pulmonary vascular remodeling, characterized by disordered proliferation and migration of pulmonary arterial smooth muscle cells (PASMCs), is a pathognomonic feature of pulmonary arterial hypertension. Thus, pharmacologic strategy targeting on anti-proliferation and anti-migration of PASMCs may have therapeutic implications for PAH. Here we investigated the effects and underlying mechanisms of B-type natriuretic peptide (BNP) on angiotensin II (Ang II)-induced proliferation and migration of PASMCs. Proliferation and migration of PASMCs cultured from Wistar rats were induced by Ang II, with or without BNP treatment. In addition, potential underlying mechanisms including cell cycle progression, Ca(2+) overload, reactive oxygen species (ROS) production, signal transduction of MAPK and Akt, and the cGMP/PKG pathway were examined. We found that BNP inhibited Ang II-induced PASMCs proliferation and migration dose dependently. BNP could also arrest the cell cycle progression in the G0/G1-phase. In addition, BNP attenuated intracellular calcium overload caused by Ang II. Moreover, Ang II-induced ROS production was mitigated by BNP, with associated down-regulation of NAD(P)H oxidase 1 (Nox1) and reduced mitochondrial ROS production. Finally, Ang II-activated MAPKs and Akt were also counteracted by BNP. Of note, all these effects of BNP were abolished by a PKG inhibitor (Rp-8-Br-PET-cGMPS). In conclusion, BNP inhibits Ang II-induced PASMCs proliferation and migration. These effects are potentially mediated by decreased calcium influx, reduced ROS production by Nox1 and mitochondria, and down-regulation of MAPK and Akt signal transduction, through the cGMP/PKG pathway. Therefore, this study implicates that BNP may have a therapeutic role in pulmonary vascular remodeling.


Subject(s)
Cell Movement/drug effects , Cell Proliferation/drug effects , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/drug effects , Natriuretic Agents/pharmacology , Natriuretic Peptide, Brain/pharmacology , Pulmonary Artery/cytology , Angiotensin II/pharmacology , Animals , Calcium/metabolism , Cells, Cultured , Down-Regulation , MAP Kinase Signaling System/drug effects , Mitochondria/drug effects , Muscle, Smooth, Vascular/drug effects , NADH, NADPH Oxidoreductases/drug effects , NADH, NADPH Oxidoreductases/metabolism , NADPH Oxidase 1 , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Pulmonary Artery/drug effects , Rats , Rats, Wistar , Reactive Oxygen Species , Vasoconstrictor Agents/pharmacology
13.
PLoS One ; 8(7): e69468, 2013.
Article in English | MEDLINE | ID: mdl-23936022

ABSTRACT

BACKGROUND: KMUP-1 is a xanthine derivative with inhibitory activities on the phosphodiesterase (PDE) 3,4 and 5 isoenzymes to suppress the degradation of cyclic AMP and cyclic GMP. However, the effects of KMUP-1 on osteoclast differentiation are still unclear. In this study, we investigated whether KMUP-1 inhibits osteoclastogenesis induced by RANKL in RAW 264.7 cells and bone loss induced by ovariectomy in mice, and the underlying mechanisms. PRINCIPAL FINDINGS: In vitro, KMUP-1 inhibited RANKL-induced TRAP activity, the formation of multinucleated osteoclasts and resorption-pit formation. It also inhibited key mediators of osteoclastogenesis including IL-1ß, IL-6, TNF-α and HMGB1. In addition, KMUP-1 inhibited RANKL-induced activation of signaling molecules (Akt, MAPKs, calcium and NF-κB), mRNA expression of osteoclastogensis-associated genes (TRAP, MMP-9, Fra-1, and cathepsin K) and transcription factors (c-Fos and NFATc1). Furthermore, most inhibitory effects of KMUP-1 on RANKL-mediated signal activations were reversed by a protein kinase A inhibitor (H89) and a protein kinase G inhibitor (KT5823). In vivo, KMUP-1 prevented loss of bone mineral content, preserved serum alkaline phosphate and reduced serum osteocalcin in ovariectomized mice. CONCLUSIONS: KMUP-1 inhibits RANKL-induced osteoclastogenesis in vitro and protects against ovariectomy-induced bone loss in vivo. These effects are mediated, at least in part, by cAMP and cGMP pathways. Therefore, KMUP-1 may have a role in pharmacologic therapy of osteoporosis.


Subject(s)
Bone Density Conservation Agents/pharmacology , Calcium/metabolism , Osteoclasts/drug effects , Osteoporosis/prevention & control , Ovariectomy , Piperidines/pharmacology , RANK Ligand/genetics , Xanthines/pharmacology , Animals , Bone Density , Calcineurin/genetics , Calcineurin/metabolism , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Female , Gene Expression Regulation , Humans , Mice , Mice, Inbred BALB C , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Osteoclasts/metabolism , Osteoclasts/pathology , Osteoporosis/genetics , Osteoporosis/metabolism , Osteoporosis/pathology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RANK Ligand/metabolism , Signal Transduction
14.
Atherosclerosis ; 226(2): 364-72, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23290263

ABSTRACT

OBJECTIVE: Inflammation is an important molecular basis of atherosclerosis. Recent studies have shown that dihydropyridine calcium channel blockers (CCBs) can exert potent anti-inflammatory effects in models of vascular dysfunction. The purpose of the present study was to evaluate anti-inflammatory effects and mechanisms of lercanidipine and labedipinedilol-A, new generation dihydropyridine CCBs, in rat vascular smooth muscle cells (VSMCs) exposed to lipopolysaccharide (LPS) and interferon-γ (IFN-γ). METHODS AND RESULTS: MTT, Griess reagent, RT-PCR, ELISA, gelatin zymography, immunocytochemistry and Western blotting were employed. We found that lercanidipine and labedipinedilol-A attenuated production of NO, ROS and TNF-α from LPS/IFN-γ-stimulated VSMCs. In addition, they both diminished the LPS/IFN-γ-induced expression of iNOS protein and mRNA, with attenuation of HMGB1 cytosolic translocation and subsequent extracellular release. Furthermore, they down-regulated MMP-2/MMP-9 activities, whereas expression of tissue inhibitor of matrix metalloproteinase-1 (TIMP-1), an inhibitor of MMP-9, was up-regulated. Finally, we found that lercanidipine and labedipinedilol-A inhibited the nuclear translocation of NF-κB and suppressed the phosphorylation of JNK, p38 MAPK and Akt. CONCLUSION: Lercanidipine and labedipinedilol-A can exert their anti-inflammatory effects through suppression of NO, ROS and TNF-α through down-regulation of iNOS, MMP-2/MMP-9, and HMGB1, with inhibition of signaling transduction of MAPKs, Akt/IkB-α and NF-κB pathways. These findings implicate a valuable role of new generation dihydropyridine CCBs lercanidipine and labedipinedilol-A for the treatment of inflammatory vascular diseases.


Subject(s)
Anisoles/pharmacology , Anti-Inflammatory Agents/pharmacology , Dihydropyridines/pharmacology , HMGB1 Protein/metabolism , Inflammation/drug therapy , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 9/biosynthesis , Muscle, Smooth, Vascular/drug effects , Animals , Inflammation/chemically induced , Interferon-gamma , Lipopolysaccharides/pharmacology , Male , Muscle, Smooth, Vascular/cytology , Rats , Tissue Inhibitor of Metalloproteinase-1/biosynthesis
15.
J Nat Med ; 66(2): 311-20, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21979292

ABSTRACT

Oxidative stress has been widely implicated in the pathogenesis of hypoxia/reoxygenation (H/R) injury. San-Huang-Xie-Xin-Tang (SHXT), a widely used traditional Chinese medication, has been shown to possess antioxidant effects. Here, we investigated whether SHXT and its main component baicalin can attenuate oxidative stress induced by H/R injury. H9c2 rat ventricular cells were exposed to SHXT or baicalin followed by hypoxia for 24 h and/or reoxygenation for 8 h. Pretreatment with SHXT and baicalin both significantly prevented cell death and production of reactive oxygen species induced by hypoxia or H/R in H9c2 cardiomyoctes. In addition, SHXT and baicalin also inhibited hypoxia- or H/R-induced apoptosis, with associated decreased Bax protein, increased Bcl-2 protein, and decreased caspase-3 activity. Furthermore, we found that hypoxia and H/R decreased endothelial nitric oxide synthase (eNOS) expression and nitrite production, and these effects were counteracted by SHXT and baicalein. Finally, SHXT inhibited H/R-induced activation of p38 mitogen activated protein kinase (MAPK) and c-Jun N-terminal kinase (JNK) phosphorylation in H9c2 rat ventricular cells. The present study demonstrates for the first time that SHXT can protect cardiomyocytes from H/R injury via inhibition of oxidative stress-induced apoptosis. These cardioprotective effects are possibly mediated through eNOS enhancement and p38 MAPK and JNK-dependent signaling pathways.


Subject(s)
Apoptosis/drug effects , Cell Survival/drug effects , Drugs, Chinese Herbal/pharmacology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Reactive Oxygen Species/metabolism , Reperfusion Injury/prevention & control , Animals , Blotting, Western , Cell Hypoxia/physiology , Cell Line , Nitrites/metabolism , Oxidative Stress/drug effects , Rats
16.
Article in English | MEDLINE | ID: mdl-21785641

ABSTRACT

San-Huang-Xie-Xin-Tang (SHXT) is a traditional Chinese medication consisting of three herbs, namely Coptidis rhizome, Scutellariae radix and Rhei rhizome. This study aimed to examine the cardioprotective effects of SHXT in a rat model of acute myocardial apoptosis induced by ischemia/reperfusion (I/R). Vehicle (intravenous saline) or SHXT (intravenous or oral) was administered prior to I/R (occlusion of left coronary artery for 45 min followed by reperfusion for 2 h). In the vehicle group, myocardial I/R caused myocardial infarction with increased plasma cardiac enzymes, severe arrhythmia and mortality. Myocardial apoptosis was induced by I/R as evidenced by DNA ladder and Bcl-2/Bax ratio. In the SHXT group, we found that SHXT significantly reduced plasma levels of cardiac enzymes, arrhythmia scores (from 5 ± 1 to 2 ± 1, P < .01) and mortality rate (from 53 to 0%, P < .01). In addition, pretreatment with intravenous SHXT reduced the infarct size dose-dependently when compared with the vehicle group (10 mg kg(-1): 14.0 ± 0.2 versus 44.5 ± 5.0%, and 30 mg kg(-1): 6.2 ± 1.2% versus 44.5 ± 5.0%, both P < .01). Similarly, oral administration of SHXT reduced the infarct size dose-dependently. Furthermore, SHXT markedly decreased the apoptosis induced by I/R with increased Bcl-2/Bax ratio. Finally, we found that SHXT counteracted the I/R-induced downstream signaling, resulting in increased myocardial eNOS expression and plasma nitrite, and decreased activation of ERK1/2, p38 and JNK. These data suggest that SHXT has cardioprotective effects against I/R-induced apoptosis, and that these effects are mediated, at least in part, by eNOS and MAPK pathways.

17.
Atherosclerosis ; 217(2): 379-86, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21601858

ABSTRACT

OBJECTIVE: Labedipinedilol-A, a novel calcium antagonist, has been previously demonstrated to have pleiotropic protective effects in the cardiovascular system. This study aimed to investigate its cytoprotective effects in rat vascular smooth muscle cells (VSMCs) treated with lysophosphatidylcholine (lysoPC), a key lipid component mediating atherogenesis. METHODS AND RESULTS: VSMCs were incubated with lysoPC with or without labedipinedilol-A pretreatment to determine its effects on lysoPC-induced cell death, Ca(2+) influx, oxidative stress, MAPK signaling and apoptosis. Labedipinedilol-A attenuated lysoPC-induced cell death and Ca(2+) influx. It also reduced reactive oxygen species (ROS) production evoked by lysoPC and down-regulated expressions of NAD(P)H oxidase subunits, Nox1 and Rac1. Moreover, it inhibited lysoPC-induced phosphorylation of MAPK including ERK1/2, JNK, and p38. It mitigated the dissipation of mitochondrial transmembrane potential induced by lysoPC. Lastly, labedipinedilol-A inhibited lysoPC-induced apoptosis with attenuation of caspase-3/-9 activations and modulation of Bax/Bcl-2 protein expressions. CONCLUSION: Labedipinedilol-A can suppress lysoPC-induced VSMCs death via reducing ROS production and anti-apoptosis. These protective effects are potentially mediated through the inhibition of Ca(2+) influx, down-regulation of the NAD(P)H oxidase subunits (Nox1/Rac1) and MAPK signaling, and attenuation of mitochondrial depolarization. Thus, labedipinedilol-A may have a valuable role in the preventing atherosclerosis associated with hyperlipidemia.


Subject(s)
Anisoles/pharmacology , Antioxidants/pharmacology , Apoptosis/drug effects , Dihydropyridines/pharmacology , Lysophosphatidylcholines/metabolism , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Calcium Signaling/drug effects , Cells, Cultured , Cytoprotection , Dose-Response Relationship, Drug , Male , Membrane Potential, Mitochondrial/drug effects , Mitogen-Activated Protein Kinase Kinases/metabolism , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , NADH, NADPH Oxidoreductases/metabolism , NADPH Oxidase 1 , Rats , Rats, Wistar , Time Factors , rac1 GTP-Binding Protein/metabolism
18.
Regul Toxicol Pharmacol ; 60(1): 112-9, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21397653

ABSTRACT

UV exposure is known to induce premature aging, which is mediated by matrix metalloproteinase-1 (MMP-1) activity. MMP-1 mRNA expression is up-regulated by elevated cysteine-rich 61 (CYR61) and monocyte chemoattractant protein-1 (MCP-1) via action of transcription factor AP-1. Collagen is degraded by MMP-1 activity but synthesized by transforming growth factor-ß (TGF-ß) signal. Chlorella has been shown to inhibit UVB-induced MMP-1 level, however its regulatory molecular mechanisms have not been studied. In this study, Chlorella derived peptide (CDP) was added to skin fibroblasts after UVB irradiation and the expression of MMP-1, CYR61, procollagen, c-fos, c-jun, and TGF-ß receptor (TbRII) mRNA and MCP-1 production were investigated. CDP (10 or 5mg/ml) diminished UVB-induced MMP-1 and CYR61 mRNA expression and MCP-1 production, whereas, UVB-suppressed procollagen and TbRII mRNA was restored by CDP treatment. UVB-induced c-fos and c-jun expressions were also inhibited by the CDP treatment. Taken together, CDP inhibits UVB-induced MMP-1 expression in skin fibroblasts by suppressing expression of AP-1 and CYR61 and MCP-1 production.


Subject(s)
Chlorella/chemistry , Fibroblasts/drug effects , Gene Expression/drug effects , Matrix Metalloproteinase 1/metabolism , Peptides/pharmacology , Procollagen/metabolism , Skin/drug effects , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Cells, Cultured , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Cysteine-Rich Protein 61/genetics , Cysteine-Rich Protein 61/metabolism , Fibroblasts/metabolism , Fibroblasts/radiation effects , Gene Expression/radiation effects , Humans , Matrix Metalloproteinase 1/genetics , Peptides/isolation & purification , Plant Extracts/pharmacology , Plant Proteins/isolation & purification , Plant Proteins/pharmacology , Procollagen/genetics , Procollagen/radiation effects , Skin/metabolism , Skin/radiation effects , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Ultraviolet Rays
19.
Br J Pharmacol ; 159(5): 1151-60, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20132211

ABSTRACT

BACKGROUND AND PURPOSE: To determine whether KMUP-1, a novel xanthine-based derivative, attenuates isoprenaline (ISO)-induced cardiac hypertrophy in rats, and if so, whether the anti-hypertrophic effect is mediated by the nitric oxide (NO) pathway. EXPERIMENTAL APPROACH: In vivo, cardiac hypertrophy was induced by injection of ISO (5 mg.kg(-1).day(-1), s.c.) for 10 days in Wistar rats. In the treatment group, KMUP-1 was administered 1 h before ISO. After 10 days, effects of KMUP-1 on survival, cardiac hypertrophy and fibrosis, the NO/guanosine 3'5'-cyclic monophosphate (cGMP)/protein kinase G (PKG) and hypertrophy signalling pathways [calcineurin A and extracellular signal-regulated kinase (ERK)1/2] were examined. To investigate the role of nitric oxide synthase (NOS) in the effects of KMUP-1, a NOS inhibitor, N(omega)-nitro-L-arginine (L-NNA) was co-administered with KMUP-1. In vitro, anti-hypertrophic effects of KMUP-1 were studied in ISO-induced hypertrophic neonatal rat cardiomyocytes. KEY RESULTS: In vivo, KMUP-1 pretreatment attenuated the cardiac hypertrophy and fibrosis and improved the survival of ISO-treated rats. Plasma NOx (nitrite and nitrate) and cardiac endothelial NOS, cGMP and PKG were all increased by KMUP-1. The activation of hypertrophic signalling by calcineurin A and ERK1/2 in ISO-treated rats was also attenuated by KMUP-1. All these effects of KMUP-1 were inhibited by simultaneous administration of L-NNA. Similarly, in vitro, KMUP-1 attenuated hypertrophic responses and signalling induced by ISO in neonatal rat cardiomyocytes. CONCLUSIONS AND IMPLICATIONS: KMUP-1 attenuates the cardiac hypertrophy in rats induced by administration of ISO. These effects are mediated, at least in part, by NOS activation. This novel agent, which targets the NO/cGMP pathway, has a potential role in the prevention of cardiac hypertrophy.


Subject(s)
Cardiomegaly/prevention & control , Nitric Oxide Synthase/drug effects , Nitric Oxide/metabolism , Piperidines/pharmacology , Xanthines/pharmacology , Animals , Calcineurin/metabolism , Cardiomegaly/mortality , Cyclic GMP/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Disease Models, Animal , Drug Delivery Systems , Fibrosis , Isoproterenol/toxicity , Male , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Nitric Oxide Synthase/metabolism , Rats , Rats, Wistar , Signal Transduction/drug effects , Survival Rate
20.
Pharmacol Res ; 59(1): 48-56, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18973813

ABSTRACT

Lercanidipine, a calcium channel antagonist, is currently employed in the treatment of essential hypertension and angina pectoris. The purpose of this study was to elucidate the anti-proliferative effect of lercanidipine and to investigate the molecular role of this agent. Both in vitro studies and in a balloon injury rat carotid artery model were employed to study the effect of lercanidipine on smooth muscle cell proliferation. Lercanidipine-inhibited rat vascular smooth muscle cell (VSMC) proliferation and migration in a dose-dependent manner following stimulation of VSMC cultures with 10% fetal bovine serum (FBS) and 20 ng/ml platelet-derived growth factor (PDGF)-BB. FBS- and PDGF-BB-stimulated intracellular Ras, MEK1/2, ERK1/2, proliferative cell nuclear antigen (PCNA), and Akt activations were significantly inhibited by lercanidipine; however, lercanidipine did not affect FBS- and PDGF-BB-induced STAT3 phosphorylation. Lercanidipine also inhibited PDGF-receptor beta chain phosphorylation and reactive oxygen species (ROS) production induced by PDGF-BB. Lercanidipine blocked the FBS-inducible progression through the G(0)/G(1) to the S-phase of the cell cycle in synchronized cells. In vivo, 14 days after balloon injury, treatment with 3 and 10 mg/kg lercanidipine resulted in significant inhibition of the neointima/media ratio. Suppression of neointima formation by lercanidipine was dependent on its influence on ERK1/2 phosphorylation. These results demonstrate that lercanidipine can suppress the proliferation of VSMCs via inhibiting cellular ROS, Ras-MEK1/2-ERK1/2, and PI3K-Akt pathways, and suggesting that it may have therapeutic relevance in the prevention of human restenosis.


Subject(s)
Calcium Channel Blockers/pharmacology , Dihydropyridines/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , MAP Kinase Signaling System/drug effects , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/drug effects , Reactive Oxygen Species/metabolism , Tunica Intima/drug effects , ras Proteins/antagonists & inhibitors , Animals , Becaplermin , Cell Movement/drug effects , Cell Proliferation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , G1 Phase/drug effects , Male , Muscle, Smooth, Vascular/drug effects , Phosphorylation , Platelet-Derived Growth Factor/pharmacology , Proliferating Cell Nuclear Antigen/analysis , Proto-Oncogene Proteins c-sis , Rats , Rats, Wistar , STAT3 Transcription Factor/metabolism , Tunica Intima/pathology , ras Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...