Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Exp Ther Med ; 10(2): 797-803, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26622396

ABSTRACT

The aim of the present study was to investigate the effects of simvastatin on the protein kinase B (PKB) signaling pathway and the expression of phosphatase and tensin homolog (PTEN). The effects of simvastatin were analyzed by administering the drug orally to male spontaneously hypertensive rats (SHRs) at a dose of 10 mg/kg/day, while the control animals received an equal volume of saline. The systolic pressure (mmHg) of the rat tail artery was measured prior to the initiation of the experiment, and once a week until the end of the experiment. At the end of the experiment, the animals were euthanized and the hearts were removed. The left ventricular and interventricular septum were weighed, after which the left ventricular mass/body mass ratio was calculated. In addition, cardiomyocytes isolated from Sprague Dawley rats were cultured with 15% fetal bovine serum to induce hypertrophy, following which the cells were treated with different doses of simvastatin. The in vitro effects were assessed by measuring the surface area of the cardiomyocytes, while the rate of protein synthesis was measured using a 3H-leucine incorporation assay and western blot analyses. Simvastatin was demonstrated to inhibit cardiomyocyte hypertrophy in the in vivo and in vitro experiments. Notably, simvastatin increased PTEN expression and inhibited PKB expression in the SHR model, as well as in the cardiomyocytes in culture. In addition, the use of PTEN antisense oligodeoxynucleotides was revealed to inhibit the effects of simvastatin on cardiomyocytes. Therefore, these results indicated that simvastatin was able to reverse cardiomyocyte hypertrophy in vivo and in vitro, possibly by increasing the expression of PTEN.

2.
Zhonghua Xin Xue Guan Bing Za Zhi ; 41(4): 333-6, 2013 Apr.
Article in Chinese | MEDLINE | ID: mdl-23906407

ABSTRACT

OBJECTIVE: To investigate glucose metabolism status and its relationship with blood pressure, obesity, renal function and cardio-cerebral vascular events in Chinese essential hypertensive patients. METHODS: Essential hypertensive patients without diabetic history were enrolled in this cross-sectional survey. All patients filled in questionnaires and received physical examination and laboratory tests. Oral glucose tolerance test (OGTT, fasting and 2 hours glucose level after drinking the 75 g glucose solution) was performed in patients who signed the informed consent. RESULTS: (1) The control rate of systolic BP was lower in patients with dysglycemia than in patients without dysglycemia (41.0% vs. 46.4%, P = 0.000). (2) The albuminuria detection rate and the abnormal rate of estimated glumerular filtration rate (eGFR) increased significantly with the deterioration of glucose metabolism. (3) Multifactor-analysis showed that abnormal waist circumference, decreased eGFR and presence of albuminuria were independent risk factors for abnormal glucose metabolism. Cardiovascular events was significantly higher in patients with abnormal glucose metabolism than patients with normal glucose metabolism. CONCLUSION: Abnormal glucose metabolism is common in Chinese essential hypertensive patients. When complicated with abnormal glucose metabolism, essential hypertensive patients had poor blood pressure control rate and were related to higher cardiovascular risk.


Subject(s)
Blood Glucose/metabolism , Glucose Metabolism Disorders/diagnosis , Hypertension/blood , Aged , Cross-Sectional Studies , Essential Hypertension , Female , Glucose Metabolism Disorders/complications , Glucose Tolerance Test , Humans , Hypertension/complications , Male , Middle Aged , Risk Factors
3.
Biomarkers ; 16(8): 657-62, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21988056

ABSTRACT

CONTEXT: Omentin-1, an adipokine secreted from visceral adipose tissue, has been reported to be associated with coronary artery disease (CAD) and metabolic disorders. OBJECTIVE: To clarify the relationship between serum omentin-1 levels and the presence and severity of CAD in patients with metabolic syndrome (MetS). METHODS: We measured serum omentin-1 levels in 175 consecutive patients with MetS and in 46 controls. RESULTS: Serum omentin-1 levels are inversely associated with the presence and angiographic severity of CAD in MetS patients. CONCLUSIONS: Serum omentin-1 might be a potential biomarker to predict the development and progression of CAD in MetS patients.


Subject(s)
Coronary Artery Disease/blood , Cytokines/metabolism , Lectins/metabolism , Metabolic Syndrome/blood , Case-Control Studies , Coronary Artery Disease/complications , GPI-Linked Proteins/metabolism , Humans , Metabolic Syndrome/complications
4.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 27(1): 7-10, 2011 Jan.
Article in Chinese | MEDLINE | ID: mdl-21208554

ABSTRACT

AIM: To investigate the role of p38 mitogen-activated protein kinase(MAPK) in lipopolysaccharide (LPS)-induced tumor necrosis factor-α (TNF-α) expression in neonatal rat cardiomyocytes and to determine the relationship between reactive oxygen species (ROS) and p38 MAPK activation. METHODS: Cardiomyocytes were isolated from neonatal Sprague-Dawley rats and cultured by differential adhesion. Expression of TNF-α was determined in culture medium by ELISA. Activation of p38 MAPK was determined by Western blot analysis with phospho-specific antibody. ROS generation in cardiomyocytes was determined by peroxide specific probe 2', 7'-dichlorofluorescin diacetate (DCF-DA). RESULTS: In cardiomyocytes stimulated with LPS, the content of TNF-α in culture medium correlated with the activity of p38 MAPK in a time-dependent manner. The activation of p38 was observed after stimulation of 1 mg/L LPS for 1 h. TNF-α accumulated significantly in culture medium at 3 h after stimulation of LPS (P<0.05), which was remarkably attenuated by pretreatment with p38 MAPK specific inhibitor SB203580 (P<0.01). Furthermore, the production of ROS in cardiomyocytes stimulated with LPS was also increased at 1 h after stimulation of LPS, consistent with p38 MAPK activity. Pretreatment with antioxidants such as N-acetylcysteine and diphenyleneiodonium significantly inhibited the activation of p38 MAPK compared with LPS control (P<0.05). There was no significance in the activity of p38 MAPK among antioxidants pretreatment and non-LPS control groups. CONCLUSION: The activation of p38 MAPK plays an important role in TNF-α expression in LPS-stimulated cardiomyocytes and the increase of ROS production is prerequisite for the activation of p38 MAPK.


Subject(s)
Myocytes, Cardiac/metabolism , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , p38 Mitogen-Activated Protein Kinases/metabolism , Acetylcysteine/pharmacology , Animals , Female , Imidazoles/pharmacology , Lipopolysaccharides/pharmacology , Male , Myocytes, Cardiac/drug effects , Onium Compounds/pharmacology , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Time Factors , Tumor Necrosis Factor-alpha/genetics , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
5.
Sheng Li Xue Bao ; 60(3): 333-40, 2008 Jun 25.
Article in English | MEDLINE | ID: mdl-18560723

ABSTRACT

Arginine vasopressin (AVP), a neurohormone and hemodynamic factor implicated in the pathophysiology of hypertension and congestive heart failure, can also act as a growth-stimulating factor. Our previous work demonstrated that AVP is a mitogen for neonatal rat cardiac fibroblasts (CFs). In the present study, we extended our investigations to adult rat CFs to explore whether AVP could induce adult rat CF proliferation and, if so, to identify the mechanism involved. Adult rat CFs were isolated, cultured and subjected to AVP treatment. DNA synthesis and cell cycle distribution were analyzed by [(3)H]-thymidine incorporation and flow cytometry. Cellular extracellular signal-regulated kinase 1/2 (ERK1/2) activity was measured by in vitro kinase assay using myelin basic protein (MBP) as a substrate. Protein expressions of total- and phospho-ERK1/2, p27(Kip1), cyclins D1, A, E were assessed by Western blot. The results showed that AVP stimulated DNA synthesis in adult rat CFs, and the effect was abolished by a V1 receptor antagonist, d(CH(2))(5)[Tyr(2)(Me), Arg(8)]-vasopressin (0.1 µmol/L), but not by a V2 receptor antagonist, desglycinamide-[d(CH(2))(5), D-Ile(2), Ile(4), Arg8]-vasopressin (0.1 µmol/L). AVP induced an activation of ERK1/2, which could be mimicked by the protein kinase C (PKC) activator, phorbol 12-myristate 13-acetate (PMA, 30 nmol/L, 5 min), but abolished by depletion of PKC via chronic PMA incubation (2.5 µmol/L, 24 h). In addition, AVP down-regulated protein expression of p27(Kip1), increased protein expressions of cyclins D1, A and E, and induced cell cycle progression from G(0)/G(1) into S stage. Inhibition of ERK1/2 activation by PD98059 (30 µmol/L) abolished the effect of AVP on DNA synthesis, protein expressions of p27(Kip1), cyclins D1, A and E as well as cell cycle progression. These results suggest that AVP is also a growth factor for adult rat CFs. The mitogenic effect of AVP is mediated via V1 receptors and PKC-ERK1/2 pathway. Moreover, AVP modulates the expressions of cell cycle regulatory proteins p27(Kip1) and cyclins D1, A and E, which lie downstream of ERK1/2 activation, and induces cell cycle progression in adult rat CFs.


Subject(s)
Arginine Vasopressin/pharmacology , Fibroblasts/drug effects , Mitogen-Activated Protein Kinase 3/metabolism , Protein Kinase C/metabolism , Signal Transduction , Animals , Antidiuretic Hormone Receptor Antagonists/pharmacology , Cell Cycle , Cell Cycle Proteins/metabolism , Cell Proliferation , Fibroblasts/cytology , Myocardium/cytology , Phosphorylation , Rats , Tetradecanoylphorbol Acetate/pharmacology
6.
Mol Cell Biochem ; 317(1-2): 33-41, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18581203

ABSTRACT

Arginine vasopressin (AVP) has been shown to directly induce neonatal rat cardiac fibroblasts (CFs) proliferation, a major component involved in cardiac hypertrophy. Herein, we explored whether AVP is also a growth factor for adult rat CFs and, if so, whether the growth effect could be inhibited by simvastatin, a 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitor. AVP significantly increased DNA synthesis in adult rat CFs by 73.5 +/- 5.1% (P < or = 0.05), an effect inhibited by V1 receptor antagonist, d(CH(2))(5)[Tyr(2)(Me), Arg(8)]-vasopressin. AVP also activated extracellular signal-regulated kinase 1/2 (ERK1/2) as assessed by MBP phosphotransferase activity (5.1 +/- 0.6 fold over basal level, P < or = 0.05) and Western blot analysis, and effects were mimicked by protein kinase C (PKC) activator, phorbol 12-myristate 13-acetate (PMA), but abolished by inhibiting cellular PKC through chronic PMA incubation. In addition, AVP induced PKC activation (27.2 +/- 3.8% from a basal value of 9.3 +/- 0.7%, P < or = 0.05). AVP-induced increase in DNA synthesis could be attenuated by the specific inhibitors of ERK1/2 (PD98059), PI3K (LY294002), and AKT (1L-6-hydroxymethyl-chiro-inositol 2-(R)-2-O-methyl-3-O-octadecylcarbonate, HIMO). Simvastatin inhibited the effects of AVP on DNA synthesis, ERK1/2, and PKC activation in a dose-dependent manner. Phosphatidylinositol-3-kinase (PI3K)-dependent AKT activation induced by AVP was also inhibited by simvastatin. The effects of simvastatin on ERK1/2, PKC, and AKT activation and DNA synthesis could be reversed by mevalonate. These results support a growth-inducing effect of AVP on adult rat CFs through ERK and AKT signalings and the growth effect could be attenuated by simvastatin via inhibiting these two pathways.


Subject(s)
Arginine Vasopressin/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblasts/enzymology , Myocardium/cytology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Simvastatin/pharmacology , Animals , Cell Proliferation/drug effects , DNA/biosynthesis , Enzyme Activation/drug effects , Fibroblasts/cytology , Fibroblasts/drug effects , Male , Mevalonic Acid/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase C/metabolism , Rats , Rats, Sprague-Dawley
7.
Mol Cell Biochem ; 310(1-2): 159-66, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18057996

ABSTRACT

Mast cell-derived chymase is implicated in myocardial fibrosis (MF), but the underlying mechanism of intracellular signaling remains unclear. Transforming growth factor-beta 1 (TGF-beta1) is identified as the most important profibrotic cytokine, and Smad proteins are essential, but not exclusive downstream components of TGF-beta 1 signaling. Moreover, novel evidence indicates that there is a cross talk between Smad and mitogen-activated protein kinase (MAPK) signaling cascade. We investigated whether chymase activated TGF-beta 1/Smad pathway and its potential role in MF by evaluating cardiac fibroblasts (CFs) proliferation and collagen synthesis in neonatal rats. MTT assay and 3H-Proline incorporation revealed that chymase induced CFs proliferation and collagen synthesis in a dose-dependent manner. RT-PCR and Western blot assay demonstrated that chymase not only increased TGF-beta1 expression but also upregulated phosphorylated-Smad2/3 protein. Furthermore, pretreatment with TGF-beta 1 neutralizing antibody suppressed chymase-induced cell growth, collagen production, and Smad activation. In contrast, the blockade of angiotensin II receptor had no effects on chymase-induced production of TGF-beta 1 and profibrotic action. Additionally, the inhibition of MAPK signaling had no effect on Smad activation elicited by chymase. These results suggest that chymase can promote CFs proliferation and collagen synthesis via TGF-beta 1/Smad pathway rather than angiotensin II, which is implicated in the process of MF.


Subject(s)
Chymases/pharmacology , Fibroblasts/drug effects , Fibroblasts/pathology , Myocardium/pathology , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Cell Proliferation/drug effects , Collagen/biosynthesis , Fibroblasts/metabolism , Fibrosis , Gene Expression Regulation/drug effects , MAP Kinase Signaling System/drug effects , Myocardium/metabolism , Phosphorylation/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Transforming Growth Factor beta1/genetics
8.
Life Sci ; 81(4): 327-35, 2007 Jul 04.
Article in English | MEDLINE | ID: mdl-17604059

ABSTRACT

Previous studies have shown that arginine vasopressin (AVP) promotes myocardial fibrosis (MF), whereas nitric oxide (NO) inhibits MF. Cardiac fibroblasts (CFs) are the main target cells of MF. However, the modulatory effect of AVP on NO production in CFs and the role of this effect in MF are still unknown. In the present study, CFs obtained from Sprague-Dawley rats were stimulated with or without AVP and pyrrolidine dithiocarbamate (PDTC), a specific inhibitor of nuclear factor kappa-B (NF-kappaB). NO production and NOS activity were detected with absorption spectrometry, inducible nitric oxide synthase (iNOS) protein with Western blot analysis, iNOS mRNA with real-time PCR, CF collagen synthesis with [(3)H]proline incorporation, and NF-kappaB activation with immunofluorescence staining and Western blot analysis. The results showed that AVP increased NO production in a dose- and time-dependent manner, with maximal effects at 10(-7) mol/l after 24-h stimulation. AVP also increased NOS activity, protein and mRNA levels of iNOS in a coincident manner. Furthermore, AVP also increased CF collagen synthesis in a dose- and time-dependent manner. In addition, it was found that NF-kappaB was activated by AVP, and that PDTC could inhibit NO production, NOS activity, protein and mRNA levels of iNOS stimulated by AVP in a dose-dependent manner. The inhibitory effects of PDTC on NF-kappaB translocation were coincident with the effects of PDTC on iNOS-NO system activity. It is suggested that AVP increases NO production via the regulation of iNOS gene expression, and the upregulation of iNOS gene expression stimulated by AVP is mediated through NF-kappaB activation. NO production induced by AVP may counteract the profibrotic effects of AVP, thus the development of MF perhaps depends on the balance between profibrotic AVP and antifibrotic NO effects on MF.


Subject(s)
Arginine Vasopressin/metabolism , Cardiomyopathies/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide/metabolism , Animals , Arginine Vasopressin/pharmacology , Cardiomyopathies/physiopathology , Cells, Cultured , Collagen/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Induction/genetics , Enzyme Inhibitors/pharmacology , Fibroblasts/ultrastructure , Fibrosis/metabolism , Fibrosis/physiopathology , Fluorescent Antibody Technique , Gene Expression Regulation, Enzymologic , In Vitro Techniques , Myocardium/cytology , Myocardium/metabolism , Myocardium/ultrastructure , NF-kappa B/drug effects , Nitric Oxide Synthase Type II/genetics , Pyrrolidines/pharmacology , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Spectrophotometry , Thiocarbamates/pharmacology
10.
Nan Fang Yi Ke Da Xue Xue Bao ; 26(2): 205-7, 213, 2006 Feb.
Article in Chinese | MEDLINE | ID: mdl-16503530

ABSTRACT

OBJECTIVE: To investigate the effects of simvastatin (Sim) and the interference by mevalonate (MVA) against its effect on DNA synthesis in rat cardiac fibroblasts (CFs). METHODS: CFs were isolated from neonatal SD rats by trypsin digestion and growth-arrested CFs were stimulated with Sim and/or MVA at varied concentrations for different time lengths, and the DNA synthesis in the cells was measured by (3)H-thymidine ((3)H-TdR) incorporation assay. RESULTS: Sim decreased (3)H-TdR incorporation in the CFs in a concentration-dependent manner, and (3)H-TdR incorporation was significantly lower in cells treated with 1 x 10(-6) and 1 x 10(-5) mol/L Sim (1,175+/-202.66 and 771+/-164.86 cpm/2000 cells, respectively) than in the control cells (1,608+/-204.32 cpm/2000 cells, P<0.01). As the treatment time with 1 x 10(-5) mol/L Sim prolonged (for 6, 12, 18, 24, 36, 42, and 48 h), (3)H-TdR incorporation in CFs decreased gradually, showing an obvious inverse correlation with the treatment time (r=-919, P<0.01). (3)H-TdR incorporation in cells treated with 1 x 10(-6) to 1 x 10(-3) mol/L MVA and 1 x 10(-5) mol/L Sim rose steadily as MVA concentration increased. A significant difference in the incorporation was found between cells treated with both 1 x 10(-4)/1 x 10(-3) mol/L MVA and 1 x 10(-5) mol/L Sim (1,612+/-308.57 and 1,995+/-353.83 cpm/2000 cells, respectively) and the cells with 1 x 10(-5) mol/L Sim treatment alone (P<0.01); difference was also noted between cells treated with 1 x 10(-5) mol/L MVA and the control cells (P<0.05), but treatment with 1 x 10(-6) mol/L MVA did not produce much difference in comparison with the control cells (P>0.05) With the increase of treatment time (for 6, 12, 18, 24, 36, 42, 48 h), 1 x 10(-3) mol/L MVA caused steady increase in (3)H-TdR incorporation in the CFs, showing a significant positive correlation with the treatment time (r=0.968, P<0.01). CONCLUSION: Sim can decrease DNA synthesis in rat CFs and postpone the occurrence of myocardial fibrosis, which can be reversed by MVA.


Subject(s)
DNA/biosynthesis , Fibroblasts/drug effects , Myocytes, Cardiac/drug effects , Simvastatin/pharmacology , Animals , Animals, Newborn , Cells, Cultured , Dose-Response Relationship, Drug , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Fibrosis/prevention & control , Hypolipidemic Agents/pharmacology , Male , Mevalonic Acid/pharmacology , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Rats , Rats, Sprague-Dawley , Time Factors
11.
Yao Xue Xue Bao ; 41(11): 1044-9, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17262945

ABSTRACT

AIM: To investigate the effects of cyclosporin A (CsA) on growth and collagen synthesis of cardiac fibroblasts (CFs) induced by arginine vasopressin (AVP). METHODS: CFs of neonatal Sprague-Dawley rats were isolated by trypsinization and cultured; growth-arrested CFs were stimulated with 1 x 10(-7) mol x L(-1) AVP in the presence or absence of CsA (0.05, 0.5 and 5 micromol x L(-1)). MTT and flow cytometry techniques were adopted to measure cell number and analyze cell cycle respectively. Collagen synthesis was determined by measurement of hydroxyproline content in culture supernatant with colorimetry. Calcineurin activity was estimated by chemiluminescence. Trypan blue staining to test the viability of CFs. RESULTS: 0.05, 0.5 and 5 micromol x L(-1) CsA inhibited the increase of CFs number induced by 1 x 10(-7) mol x L(-1) AVP in a dose-dependent manner, with the inhibitory rates by 12%, 24% and 29%, respectively (P < 0.05). Furthermore, cell cycle analysis showed 0.5 micromol x L(-1) CsA decreased the S stage percentage and proliferation index of CFs stimulated by AVP (P < 0.05). In culture medium, the hydroxyproline content induced by AVP decreased by 0.5 and 5 micromol x L(-1) CsA (P < 0.05), with the inhibitory rates of 29% and 33%, respectively. CsA completely inhibited the increment of calcineurin activity induced by AVP (P < 0.01), but CsA itself had no effect on the baseline of calcineurin activity and CFs viability. CONCLUSION: CsA inhibits proliferation and collagen synthesis of CFs by virtue of blocking calcineurin signaling pathway and might provide a novel target for prevention and treatment to cardiac fibrosis.


Subject(s)
Arginine Vasopressin/pharmacology , Cell Proliferation/drug effects , Collagen/biosynthesis , Cyclosporine/pharmacology , Fibroblasts/drug effects , Animals , Animals, Newborn , Calcineurin/metabolism , Cell Cycle/drug effects , Cell Survival/drug effects , Cells, Cultured , Cyclosporine/administration & dosage , Dose-Response Relationship, Drug , Fibroblasts/cytology , Fibroblasts/metabolism , Hydroxyproline/metabolism , Myocardium/cytology , Rats , Rats, Sprague-Dawley
12.
Zhonghua Yi Xue Za Zhi ; 85(19): 1344-7, 2005 May 25.
Article in Chinese | MEDLINE | ID: mdl-16029635

ABSTRACT

OBJECTIVE: To investigate the effects of simvastatin on left ventricular hypertrophy (LVH) in spontaneously hypertensive rats (SHRs) and its possible mechanism. METHODS: Sixteen male SHRs were randomly divided into 2 equal groups: treatment group and SHR control to be given simvastatin or glucose-normal saline by oral gavage for 10 weeks. Eight Wistar-Kyoto (WKY) rats were given normal saline as normal controls. Blood pressure was measured before the experiment and then once every week after the beginning of experiment. By the end of the experiment the rats were killed and their hearts were taken out to measure the left ventricle weight/body weight. RT-PCR was used to detect the mRNA expression of atrial natriuretic peptide (ANP) and of protein kinase B (PKB) in myocardium. Western blotting was used to examine the protein expression of PKB. RESULTS: (1) The systolic blood pressure of the SHR normal control and treatment groups were 221 mm Hg +/- 10 mm Hg and 217 mm Hg +/- 8 mm Hg respectively (P > 0.05) and the systolic pressure of the normal control group was 126 +/- 6 mm Hg, significantly lower than those of the 2 SHR groups (both P < 0.01). (2) The LVW/BW values of the SHR control group were 3.04 mg/g +/- 0.12 mg/g, 3.73 mg/g +/- 0.08 mg/g, and 4.10 mg/g +/- 0.13 mg/g in the normal control group, SHR treatment group and SHR control group respectively with significant difference between any 2 groups (all P < 0.01). (3) The mRNA expression levels of ANP were 0.44 +/- 0.33, 0.27 +/- 0.03, and 0.17 +/- 0.33 in the SHR control group, SHR treatment group, and normal control group respectively (P < 0.01 or P < 0.05). (4) The mRNA expression levels of PKB were 0.45 +/- 0.05, 0.32 +/- 0.03, and 0.19 +/- 0.02 in the SHR control group, SHR treatment group, and normal control group respectively (P < 0.01 or P < 0.05). CONCLUSION: Simvastatin reverses LVH and myocyte phenocyte transformation in the SHRs with the possible mechanism of decreasing the expression level of PKB.


Subject(s)
Hypertension/complications , Hypertrophy, Left Ventricular/enzymology , Proto-Oncogene Proteins c-akt/biosynthesis , Simvastatin/pharmacology , Animals , Male , Proto-Oncogene Proteins c-akt/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Inbred SHR , Rats, Inbred WKY
13.
Zhonghua Yi Xue Za Zhi ; 85(1): 49-53, 2005 Jan 05.
Article in Chinese | MEDLINE | ID: mdl-15808078

ABSTRACT

OBJECTIVES: The phenotypic modulation of vascular smooth muscle cells (VSMC) plays a central role in the pathogenesis of arteriosclerosis. The purpose of this study was to investigate the expression of the cellular repressor of E1A-activated genes (CREG) at the transcriptional and protein level in human internal thoracic artery smooth muscle cells (HITASY), which express different patterns of differentiation markers after serum withdrawal. METHODS: After cloning and recombining the CREG vector, the antiserum against the CREG protein was produced from the rabbits immunized by the purification CREG protein. The specificity of purified polyclonal antibody was detected by Western blot assay. The DNA synthesis of HITASY cultured in serum-free and serum-supplemented medium was measured by the [(3)H]-thymidine incorporation. Western blot analysis detected the expression of smooth muscle-specific markers (smooth muscle alpha-actin, calponin). The localization of CREG in cells was examined with immunohistochemistry staining and expression of CREG mRNA and protein were analyzed by RT-PCR and Western blot in HITASY after serum withdrawal. RESULTS: The high specificity of polyclonal antibody against CREG obtained from rabbits was confirmed by Western blot assay. In response to serum withdrawal, cultured HITASY cells exhibited phenotypic conversion from synthetic into contractile one as evidenced by the data of [(3)H]-thymidine incorporation and Western blot. The DNA synthesis of HITASY precipitously dropped to background levels after serum withdrawal and nearly restored after reintroduction of serum to culture medium 2 days later. Western blot revealed a reversible upregulation of smooth muscle alpha-actin and calponin in HITASY after serum deprivation. Moreover, serum withdrawal also induced a prominent increase of CREG mRNA and protein expression which reached a peak on 3 days and decreased gradually on 5 approximately 7 days after serum withdrawal. Immunohistochemistry stain indicated the CREG protein mainly localizes in a perinuclear pattern in HITASY cells. CONCLUSIONS: Those data provide evidence that the coordinated changes in CREG gene and protein expression as well as smooth muscle-specific markers may take place in connection with the process of phenotypic modulation of VSMC in vitro.


Subject(s)
Arteriosclerosis/metabolism , Muscle, Smooth, Vascular/metabolism , Repressor Proteins/biosynthesis , Actins/biosynthesis , Animals , Aorta, Thoracic/pathology , Calcium-Binding Proteins/biosynthesis , Cell Division , Cells, Cultured , Microfilament Proteins , Muscle, Smooth, Vascular/pathology , Phenotype , Rabbits , Repressor Proteins/genetics , Calponins
14.
Sheng Li Xue Bao ; 55(4): 417-21, 2003 Aug 25.
Article in Chinese | MEDLINE | ID: mdl-12937821

ABSTRACT

To investigate the changes in the nitric oxide (NO) contents, nitric oxide synthase (NOS) activity and inducible nitric oxide (iNOS) mRNA expression in arginine vasopressin (AVP)-induced cardiac fibroblasts (CFs) in vitro and its relation to nuclear factor-kappaB (NF-kappaB), CFs were isolated by trypsin digestion method. Nitric acid reductase method, spectrophotometry, reverse transcription-polymerase chain reaction (RT-PCR), immunofluorescence-interactive laser cytometer techniques and Western blotting were used respectively to detect NO contents, NOS activity, iNOS mRNA expression and the activation of NF-kappaB in CFs. AVP increased NO contents, NOS activity and iNOS mRNA expressions in a concentration-dependent manner; NF-kappaB was activated and mobilized from cytoplasm to nucleus in AVP-induced CFs; PDTC, one of the inhibitors of NF-kappaB, could inhibit aforementioned increments. It is suggested that the increases in NO contents, elevation of NOS activity and increment of iNOS mRNA expression may be mediated through NF-kappaB activation pathway in cultured CFs induced by AVP, and that NF-kappaB is involved in the occurrence and development of myocardial fibrosis.


Subject(s)
Arginine Vasopressin/pharmacology , Fibroblasts/metabolism , Myocytes, Cardiac/metabolism , NF-kappa B/metabolism , Nitric Oxide/metabolism , Animals , Animals, Newborn , Cells, Cultured , Fibroblasts/cytology , Myocytes, Cardiac/cytology , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley
15.
Zhonghua Yi Xue Za Zhi ; 83(5): 421-4, 2003 Mar 10.
Article in Chinese | MEDLINE | ID: mdl-12820922

ABSTRACT

OBJECTIVE: To study the effects of chelerythrine, a protein kinase C (PKC) inhibitor, on the cell proliferation and p27 expression of cardiac fibroblasts (CFs) modulated by arginine vasopressin (AVP) and to investigate the intracellular signal transduction mechanisms of AVP in CFs. METHODS: The cultured CFs of neonatal Sprague-Dawley rats were divided into 3 groups: AVP group (10(-7) mol/L AVP was added into the culture), chelerythrine group (10(-6) mol/L chelerythrine and 10(-7) mol/L AVP were added into the culture), and control group. MTT assay was used to evaluate the cell proliferation. The cultured cells were collected and propidium iodide was used to label the DNA so as to identify the cell cycle. Specific mouse-versus-rat p27 protein monoclonal antibody and fluorescein isothiocyanate-labeled secondary antibody were added into the cell suspension to label the p27 protein in the cells. Flow cytometry was used to determine the distribution of cell cycles and p27 expression. RESULTS: (1). The A value of CFs measured by MTT assay in AVP + chelerythrine group was 0.32 +/- 0.01, significantly lower than that of the AVP group (0.39 +/- 0.01, P < 0.01). (2). The percentage of CFs in S stage was 4.4% +/- 1.7% in the AVP + chelerythrine group, lower than those of the AVP group (15.5% +/- 1.4%, P < 0.01) and control group (7.5% +/- 1.0%). The PI of CFs was 20.9% +/- 1.2% in the AVP + chelerythrine group, significantly lower than that of the AVP group (31.4% +/- 1.5%, P < 0.01). The PI of the AVP group was significantly lower than that of the control group (26.0% +/- 1.0%, P < 0.01). The percentage of CFs in G(0)/G(1) stage was 79.1% +/- 1.2% in the AVP + 1 chelerythrine group, significantly higher than that in the AVP group (68.6% +/- 1.5%, P < 0.01). The percentage of CFs in G(0)/G(1) stage in the AVP group was significantly lower than that in the control group (74.0% +/- 1.0%, P < 0.01) too. (3). The expression rate of p27 protein was 91.7% +/- 2.2% in the AVP + chelerythrine group, significantly higher than that in the AVP group (63.3% +/- 1.9%, P < 0.01). CONCLUSION: PKC inhibitor remarkably reverses the CFs proliferation and p27 downregulation induced by AVP. It may be involved in the intracellular signal transduction pathway of AVP in CFs.


Subject(s)
Arginine Vasopressin/pharmacology , Cell Proliferation/drug effects , Myocardium/cytology , Phenanthridines/pharmacology , Alkaloids , Animals , Animals, Newborn , Benzophenanthridines , Cell Cycle Proteins , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p27 , Enzyme Inhibitors/pharmacology , Fibroblasts/metabolism , Protein Kinase C/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Signal Transduction , Tumor Suppressor Proteins
16.
Zhonghua Yi Xue Za Zhi ; 83(2): 118-22, 2003 Jan 25.
Article in Chinese | MEDLINE | ID: mdl-12812679

ABSTRACT

OBJECTIVE: To investigate the effects of atorvastatin on the proliferation and collagen synthesis of rat cardiac fibroblasts (CFs). METHODS: Isolated and cultured CFs of neonatal Sprague-Dawley (SD) rats were isolated and cultured. The DNA synthesis of CFs was measured by (3)H-TdR uptake test. CFs proliferation was measured by thiazolyl blue (MTT) assay. Cell cycle distribution was determined with flow cytometer (FCM). Collagen synthesis was measured by 3H-proline uptake test. RESULTS: (1) The (3)H-TdR uptake in the 10(-7) mol/L, 10(-6) mol/L, 10(-5) mol/L, and 10(-4) mol/L atorvastatin groups was (cpm/5,000 cells) 314 +/- 68, 253 +/- 52, 201 +/- 53, and 170 +/- 48 respectively, all significantly lower than that of the control group (378 +/- 65, all P < 0.001). (2) MTT colorimetry showed that the A(490) values were 0.288 +/- 0.008, 0.252 +/- 0.007, 0.225 +/- 0.008, and 0.216 +/- 0.013 respectively in the 10(-7) mol/L, 10(-6) mol/L, 10(-5) mol/L and 10(-4) mol/L atorvastatin groups, all significantly lower than that in the control group (0.311 +/- 0.005, all P < 0.01). (3) The percentage of cells in G(0)/G(1) phase was 54.8% +/- 2.5%, 61.4% +/- 2.7%, 70.4% +/- 3.2%, and 82.0% +/- 4.0% in the 10(-7) - 10(-4) mol/L atorvastatin groups, all significantly higher than that in the control group (46.5 +/- 2.9, all P < 0.01). The percentage of cells in S phase was 18.8% +/- 2.3%, 15.8% +/- 2.1%, 12.5% +/- 1.8%, and 7.3% +/- 2.0% in the 10(-7) - 10(-4) mol/L atorvastatin groups, all significantly lower than that in the control group (23.1% +/- 2.0%, all P < 0.01). The percentage of cells in G(2)/M phase was 26.5% +/- 0.8%, 22.8% +/- 1.2%, 17.2% +/- 1.4%, and 10.7% +/- 2.0% in the 10(-7) - 10(-4) mol/L atorvastatin groups respectively, all significantly lower than that in the control group (30.5% +/- 1.4%, all P < 0.01). The proliferation index (PI) was 45.3% +/- 2.5%, 38.6% +/- 2.7%, 29.6% +/- 3.2%, and 18.0% +/- 4.0% in the 10(-7) - 10(-4) mol/L atorvastatin groups respectively, all significantly lower than that in the control group (53.5% +/- 2.9%, all P < 0.01). (4) The (3)H-proline uptake was (cpm/5 000 cells) 422 +/- 59, 332 +/- 67, 252 +/- 53, and 184 +/- 48 in the 10(-7) mol/L, 10(-6) mol/L, 10(-5) mol/L, and 10(-4) mol/L atorvastatin groups respectively, all significantly lower than that in the control group (566 +/- 62, all P < 0.01). CONCLUSION: Atorvastatin inhibits effectively the proliferation of cultured rat CFs and the collagen synthesis therein, which may play a role in the regression of heart remodeling.


Subject(s)
Collagen/biosynthesis , Fibroblasts/metabolism , Heptanoic Acids/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Myocardium/cytology , Pyrroles/pharmacology , Animals , Animals, Newborn , Atorvastatin , Cell Division/drug effects , Cells, Cultured , Rats , Rats, Sprague-Dawley , Ventricular Remodeling
17.
J Cardiovasc Pharmacol ; 42(1): 132-5, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12827038

ABSTRACT

The abnormal proliferation of cardiac fibroblasts is involved in the pathophysiologic process of left ventricular hypertrophy (LHV) associated with essential hypertension. Arginine vasopressin (AVP) has been reported to contribute significantly to the pathogenesis of hypertension. In this study, the authors investigated the effects of AVP and its V1 receptor antagonist [d(CH2)5Tyr2(Me)]AVP on the growth of rat cardiac fibroblasts. Cardiac fibroblasts of neonatal Sprague-Dawley rats were isolated, and growth-arrested cardiac fibroblasts were stimulated with 2.5% fetal calf serum in the presence and absence of AVP (0.001, 0.01, 0.1, and 1 microM) and [d(CH2)5Tyr2(Me)]AVP (0.1 microM). DNA synthesis was measured by [3H]thymidine incorporation. Thiazolyl blue assay and flow cytometry techniques were adopted to measure cell numbers and analyze cell cycle, respectively. Arginine vasopressin (0.1 and 1 microM) significantly increased DNA synthesis in cardiac fibroblasts. Moreover, AVP (0.1 and 1 microM) significantly increased the number of cardiac fibroblasts. Analysis of cell cycle showed that AVP (0.1 microM) increased S-stage percentage and proliferation index (PI). The V1 receptor antagonist [d(CH2)5Tyr2(Me)]AVP (0.1 microM) significantly inhibited DNA synthesis in cardiac fibroblasts. The cell number, S-stage percentage, and PI induced by AVP (0.1 microM) were significantly decreased by [d(CH2)5Tyr2(Me)]AVP (0.1 microM). These findings suggest that AVP might promote the proliferation of rat cardiac fibroblasts, which seems to be mediated via the V1 receptor. Arginine vasopressin may be involved in the pathophysiologic process of LVH by promoting cardiac fibroblast proliferation.


Subject(s)
Arginine Vasopressin/analogs & derivatives , Arginine Vasopressin/toxicity , Heart/drug effects , Hypertrophy, Left Ventricular/etiology , Myocardium/metabolism , Receptors, Vasopressin/metabolism , Animals , Antidiuretic Hormone Receptor Antagonists , Arginine Vasopressin/pharmacology , Cell Cycle/drug effects , Cell Survival/drug effects , Cells, Cultured , DNA/biosynthesis , Fibroblasts/cytology , Fibroblasts/metabolism , Heart/growth & development , Myocardium/cytology , Rats , Rats, Sprague-Dawley
18.
Di Yi Jun Yi Da Xue Xue Bao ; 21(12): 920-922, 2001.
Article in English | MEDLINE | ID: mdl-12426166

ABSTRACT

OBJECTIVE: To investigate the effects of simvastatin on the proliferation of rat cardiac fibroblasts (CFs) induced by arginine vasopressin (AVP). METHODS: CFs of neonatal Sprague-Dawley (SD) rats were isolated by trypsin digestion method and growth-arrested CFs were stimulated with 1x10-7 mol/L AVP in the presence of simvastatin (Sim) with varied concentrations. MTT assay was employed to measure CFs proliferation and determine the cell number, and the cell cycle distribution was determined with flow cytometer (FCM). RESULTS: With the increase of Sim concentration, D490 of CFs as shown by MTT assay gradually decreased, and for the cells treated with 1x10-6 mol/L Sim or 1x10-5 mol/L Sim, D490 (0.215+/-0.041and 0.163+/-0.018, respectively) was significantly lower than that of the control (0.939+/-0.048, P<0.01). In a dose-dependent manner, Sim decreased the cell percentage at S stage and the proliferation index (PI) as its concentration increased, but acted to the contrary effect with the percentage of cells at G0/G1 stage, and in CFs treated with 1x10-5 mol/L or 1x10-6 mol/l Sim, the 3 parameters were significantly different from those measured in the CFs with 1x10-7 mol/L AVP treatment (P<0.01). CONCLUSION: The results indicate that Sim can inhibit the proliferation of CFs induced by AVP, possibly through the mechanism of regulating the cell cycle distribution.

SELECTION OF CITATIONS
SEARCH DETAIL
...