Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Type of study
Language
Publication year range
1.
Mediators Inflamm ; 2019: 6168340, 2019.
Article in English | MEDLINE | ID: mdl-31686985

ABSTRACT

Andrographolide (Andr) is a major component isolated from the plant Andrographis paniculata. Inflammation, apoptosis, and impaired angiogenesis are implicated in the pathogenesis of high glucose (HG)-induced injury of vascular endotheliocytes. Our study is aimed at evaluating the effect of Andr on HG-induced HUVEC injury and the underlying mechanism. HUVECs were exposed to HG levels (33 mM) and treated with Andr (0, 12.5, 25, and 50 µM). Western blot analysis, real-time PCR, immunofluorescence staining, the scratch test, and the tube formation assay were performed to assess the effects of Andr. We discovered that Andr inhibited the inflammatory response (IL-1ß, IL-6, and TNFα), decreased the apoptosis ratio and cell migration, and promoted tube formation in response to HG stimulation. Andr ameliorated the levels of phosphorylated PI3K (p-PI3K), phosphorylated AKT (p-AKT), and phosphorylated eNOS (p-eNOS). The expression of vascular endothelial growth factor (VEGF) protein, a vital factor in angiogenesis, was improved by Andr treatment under HG stimulation. LY294002 is a blocker of PI3K, MK-2206 2HCI (MK-2206) is a highly selective AKT inhibitor, and L-NAME is a suppressor of eNOS, all of which significantly reduce Andr-mediated protective effects in vitro. Hence, Andr may be involved in regulating HG-induced injury by activating PI3K/AKT-eNOS signalling in HUVECs.


Subject(s)
Diterpenes/pharmacology , Human Umbilical Vein Endothelial Cells/metabolism , Inflammation/prevention & control , Apoptosis/drug effects , Blotting, Western , Cell Movement/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Humans , In Situ Nick-End Labeling , Inflammation/chemically induced , Inflammation/metabolism , L-Lactate Dehydrogenase/metabolism , Malondialdehyde/metabolism , Nitric Oxide Synthase Type III/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects
2.
J Cell Mol Med ; 23(9): 6466-6478, 2019 09.
Article in English | MEDLINE | ID: mdl-31293067

ABSTRACT

Cardiac remodelling refers to a series of changes in the size, shape, wall thickness and tissue structure of the ventricle because of myocardial injury or increased pressure load. Studies have shown that cardiac remodelling plays a significant role in the development of heart failure. Zingerone, a monomer component extracted from ginger, has been proven to possess various properties including antioxidant, anti-inflammatory, anticancer and antidiabetic properties. As oxidative stress and inflammation contribute to acute and chronic myocardial injury, we explored the role of zingerone in cardiac remodelling. Mice were subjected to aortic banding (AB) or sham surgery and then received intragastric administration of zingerone or saline for 25 days. In vitro, neonatal rat cardiomyocytes (NRCMs) were treated with zingerone (50 and 250 µmol/L) when challenged with phenylephrine (PE). We observed that zingerone effectively suppressed cardiac hypertrophy, fibrosis, oxidative stress and inflammation. Mechanistically, Zingerone enhanced the nuclear factor (erythroid-derived 2)-like 2 (Nrf2)/antioxidant response element (ARE) activation via increasing the phosphorylation of endothelial nitric oxide synthase (eNOS) and nitric oxide (NO) production. Additionally, we used Nrf2-knockout (KO) and eNOS-KO mice and found that Nrf2 or eNOS deficiency counteracts these cardioprotective effects of zingerone in vivo. Together, we concluded that zingerone may be a potent treatment for cardiac remodelling that suppresses oxidative stress via the eNOS/Nrf2 pathway.


Subject(s)
Aorta/drug effects , Guaiacol/analogs & derivatives , NF-E2-Related Factor 2/metabolism , Nitric Oxide Synthase Type III/metabolism , Signal Transduction/drug effects , Ventricular Remodeling/drug effects , Animals , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Aorta/metabolism , Cardiomegaly/drug therapy , Cardiomegaly/metabolism , Cells, Cultured , Fibrosis/drug therapy , Fibrosis/metabolism , Guaiacol/pharmacology , Heart Failure/drug therapy , Heart Failure/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardium/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Oxidative Stress/drug effects , Phenylephrine/pharmacology , Rats
3.
J Cell Mol Med ; 23(8): 5715-5727, 2019 08.
Article in English | MEDLINE | ID: mdl-31225721

ABSTRACT

Increase of myocardial oxidative stress is closely related to the occurrence and development of cardiac hypertrophy. Cordycepin, also known as 3'-deoxyadenosine, is a natural bioactive substance extracted from Cordyceps militaris (which is widely cultivated for commercial use in functional foods and medicine). Since cordycepin suppresses oxidative stress both in vitro and in vivo, we hypothesized that cordycepin would inhibit cardiac hypertrophy by blocking oxidative stress-dependent related signalling. In our study, a mouse model of cardiac hypertrophy was induced by aortic banding (AB) surgery. Mice were intraperitoneally injected with cordycepin (20 mg/kg/d) or the same volume of vehicle 3 days after-surgery for 4 weeks. Our data demonstrated that cordycepin prevented cardiac hypertrophy induced by AB, as assessed by haemodynamic parameters analysis and echocardiographic, histological and molecular analyses. Oxidative stress was estimated by detecting superoxide generation, superoxide dismutase (SOD) activity and malondialdehyde levels, and by detecting the protein levels of gp91phox and SOD. Mechanistically, we found that cordycepin activated activated protein kinase α (AMPKα) signalling and attenuated oxidative stress both in vivo in cordycepin-treated mice and in vitro in cordycepin treated cardiomyocytes. Taken together, the results suggest that cordycepin protects against post-AB cardiac hypertrophy through activation of the AMPKα pathway, which subsequently attenuates oxidative stress.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Cardiomegaly/drug therapy , Deoxyadenosines/therapeutic use , Signal Transduction , Angiotensin II/pharmacology , Animals , Cardiomegaly/diagnostic imaging , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Cardiotonic Agents/pharmacology , Cardiotonic Agents/therapeutic use , Deoxyadenosines/pharmacology , Fibrosis , Hemodynamics/drug effects , Male , Mice, Inbred C57BL , Models, Biological , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Oxidative Stress/drug effects , Phosphorylation/drug effects , Pressure , Signal Transduction/drug effects
4.
Oxid Med Cell Longev ; 2018: 4327901, 2018.
Article in English | MEDLINE | ID: mdl-30046377

ABSTRACT

Whether aucubin could protect myocardial infarction- (MI-) induced cardiac remodeling is not clear. In this study, in a mouse model, cardiac remodeling was induced by left anterior descending coronary artery ligation surgery. Mice were intraperitoneally injected with aucubin (10 mg/kg) 3 days post-MI. Two weeks post-MI, mice in the aucubin treatment group showed decreased mortality, decreased infarct size, and improved cardiac function. Aucubin also decreased cardiac remodeling post-MI. Consistently, aucubin protected cardiomyocytes against hypoxic injury in vitro. Mechanistically, we found that aucubin inhibited the ASK1/JNK signaling. These effects were abolished by the JNK activator. Moreover, we found that the oxidative stress was attenuated in both in vivo aucubin-treated mice heart and in vitro-treated cardiomyocytes, which caused decreased thioredoxin (Trx) consumption, leading to ASK1 forming the inactive complex with Trx. Aucubin increased nNOS-derived NO production in vivo and vitro. The protective effects of aucubin were reversed by the NOS inhibitors L-NAME and L-VINO in vitro. Furthermore, nNOS knockout mice also reversed the protective effects of aucubin on cardiac remodeling. Taken together, aucubin protects against cardiac remodeling post-MI through activation of the nNOS/NO pathway, which subsequently attenuates the ROS production, increases Trx preservation, and leads to inhibition of the ASK1/JNK pathway.


Subject(s)
Iridoid Glucosides/therapeutic use , Myocardial Infarction/drug therapy , Myocardial Infarction/metabolism , Animals , Cell Line , Disease Models, Animal , Echocardiography , Hemodynamics/drug effects , Immunohistochemistry , In Situ Nick-End Labeling , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardium/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase Type I/metabolism , Nitrogen Oxides/metabolism , Oxidative Stress/drug effects , Rats , Reactive Oxygen Species/metabolism , Thioredoxins/metabolism , Ventricular Remodeling/drug effects
5.
Br J Pharmacol ; 175(9): 1548-1566, 2018 05.
Article in English | MEDLINE | ID: mdl-29447430

ABSTRACT

BACKGROUND AND PURPOSE: Aucubin, the predominant component of Eucommia ulmoides Oliv., has been shown to have profound effects on oxidative stress. As oxidative stress has previously been demonstrated to contribute to acute and chronic myocardial injury, we tested the effects of aucubin on cardiac remodelling and heart failure. EXPERIMENTAL APPROACH: Initially, H9c2 cardiomyocytes and neonatal rat cardiomyocytes pretreated with aucubin (1, 3, 10, 25 and 50 µM) were challenged with phenylephrine. Secondly, the transverse aorta was constricted in C57/B6 and neuronal NOS (nNOS)-knockout mice, then aucubin (1 or 5 mg·kg-1 body weight day-1 ) was injected i.p. for 25 days. Hypertrophy was evaluated by assessing morphological changes, echocardiographic parameters, histological analyses and hypertrophic markers. Oxidative stress was evaluated by examining ROS generation, oxidase activity and NO generation. NOS expression was determined by Western blotting. KEY RESULTS: Aucubin effectively suppressed cardiac remodelling; in mice, aucubin substantially inhibited pressure overload-induced cardiac hypertrophy, fibrosis and inflammation, whereas knocking out nNOS abolished these cardioprotective effects of aucubin. Blocking or knocking down the ß3 -adrenoceptor abolished the protective effects of aucubin in vitro. Furthermore, aucubin enhanced the protective effects of a ß3 -adrenoceptor agonist in vitro by increasing cellular cAMP levels, whereas treatment with an adenylate cyclase (AC) inhibitor abolished the cardioprotective effects of aucubin. CONCLUSIONS AND IMPLICATIONS: Aucubin suppresses oxidative stress during cardiac remodelling by increasing the expression of nNOS in a process that requires activation of the ß3 -adrenoceptor/AC/cAMP pathway. These findings suggest that aucubin could have potential as a treatment for cardiac remodelling and heart failure.


Subject(s)
Heart Failure/prevention & control , Iridoid Glucosides/pharmacology , Nitric Oxide Synthase Type I/metabolism , Receptors, Adrenergic, beta-3/metabolism , Ventricular Remodeling/drug effects , Adrenergic beta-3 Receptor Agonists/pharmacology , Adrenergic beta-3 Receptor Antagonists/pharmacology , Animals , Cardiotonic Agents/pharmacology , Cardiotonic Agents/therapeutic use , Cells, Cultured , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Drug Interactions , Hypertrophy/drug therapy , Hypertrophy/pathology , Iridoid Glucosides/therapeutic use , Male , Mice , Mice, Knockout , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Nitric Oxide Synthase Type I/biosynthesis , Nitric Oxide Synthase Type I/genetics , Oxidative Stress/drug effects , Phenylephrine/pharmacology , Protective Agents/pharmacology , Rats , Receptors, Adrenergic, beta-3/genetics
6.
Biochim Biophys Acta Mol Basis Dis ; 1864(5 Pt A): 1728-1743, 2018 May.
Article in English | MEDLINE | ID: mdl-29476905

ABSTRACT

Diabetic cardiomyopathy is associated with suppressed autophagy and augmented inflammation in the heart. The effects of Tax1 binding protein 1 (TAX1BP1) on both autophagy and inflammation suggest that it may participate in the progression of diabetic cardiomyopathy. Mice were injected with streptozotocin (STZ) to induce experimental diabetes. An adenovirus system was used to induce heart specific TAX1BP1 overexpression 12 weeks after STZ injection. TAX1BP1 expression was significantly decreased in STZ-induced diabetic mouse hearts. TAX1BP1 overexpression in the heart alleviated cardiac hypertrophy and fibrosis, attenuated inflammation, oxidative stress, and apoptosis, and improved cardiac function in STZ-induced diabetic mice. Diabetic mice exhibited decreased autophagy. By contrast, increased autophagy was observed in diabetic mice overexpressing TAX1BP1. TAX1BP1 overexpression promoted autophagic flux, as demonstrated by increased LC3-RFP fluorescence in vitro. Furthermore, the autophagy inhibitor 3-MA abolished the protective effects of TAX1BP1 in vivo. Interestingly, we found that TAX1BP1 increased autophagy via the activation of a non-canonical NF-κB signaling pathway. Conversely, RelB knockdown disrupted the protective effects of TAX1BP1 in cardiomyocytes. TAX1BP1 thus restores the decreased autophagy level, leading to decreased inflammatory responses and oxidative stress and reduced apoptosis in cardiomyocytes.


Subject(s)
Autophagy , Diabetes Mellitus, Experimental/metabolism , Diabetic Cardiomyopathies/metabolism , Intracellular Signaling Peptides and Proteins/biosynthesis , Myocytes, Cardiac/metabolism , Neoplasm Proteins/biosynthesis , Oxidative Stress , Animals , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Diabetic Cardiomyopathies/genetics , Diabetic Cardiomyopathies/pathology , Gene Knockdown Techniques , Intracellular Signaling Peptides and Proteins/genetics , Male , Mice , Myocytes, Cardiac/pathology , Neoplasm Proteins/genetics
7.
Planta Med ; 84(2): 91-99, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28841738

ABSTRACT

Fibrosis is a key feature of various cardiovascular diseases and compromises cardiac systolic and diastolic performance. The lack of effective anti-fibrosis drugs is a major contributor to the increasing prevalence of heart failure. The present study was performed to investigate whether the iridoid aucubin alleviates cardiac fibroblast activation and its underlying mechanisms. Neonatal rat cardiac fibroblasts were incubated with aucubin (1, 10, 20, 50 µM) followed by transforming growth factor ß1 (TGFß1, 10 ng/mL) stimulation for 24 h. Fibrosis proliferation was measured by cell counting kit-8 assay. The differentiation of fibroblasts into myofibroblasts was determined by measuring the expression of α-smooth muscle actin. Then, the expressions levels of cardiac fibrosis-related proteins in myofibroblasts were analyzed by western blot and real-time PCR to confirm the anti-fibrosis effect of aucubin. As a result, aucubin suppressed TGFß1-induced proliferation in fibroblasts and inhibited the TGFß1-induced activation of fibroblasts to myofibroblasts. In addition, aucubin further attenuated fibrosis-related protein expression in myofibroblasts. Furthermore, this protective effect was related to increased adenosine 5'-monophosphate-activated protein kinase (AMPK) phosphorylation and decreased mammalian target of rapamycin (mTOR) phosphorylation, which was confirmed by an mTOR inhibitor (rapamycin), an AMPK agonist (AICAR) and an AMPKα inhibitor compound C. Collectively, our findings suggest that aucubin protects against TGFß1-induced fibroblast proliferation, activation and function by regulating the AMPKα/mTOR signal axis.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Fibroblasts/drug effects , Iridoid Glucosides/pharmacology , Myocardium/cytology , TOR Serine-Threonine Kinases/metabolism , Transforming Growth Factor beta1/metabolism , AMP-Activated Protein Kinases/antagonists & inhibitors , Animals , Cell Proliferation/drug effects , Cells, Cultured , Fibroblasts/metabolism , Fibrosis/prevention & control , Heart/drug effects , Myocardium/metabolism , Rats , Rats, Sprague-Dawley , TOR Serine-Threonine Kinases/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...