Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
1.
Bull Exp Biol Med ; 169(4): 549-557, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32910392

ABSTRACT

Regenerative potential of multipotent mesenchymal stromal cells from the human umbilical cord (MMSC-UC) in the suspension and spheroid form was revealed during the progression of experimental small focal myocardial infarction in rats. In isoproterenol-induced myocardial infarction, foci of necrosis and inflammatory infiltrate and at later terms fibrosis foci were found mainly in the left ventricle of rat heart. In rats receiving MMSC-UC, destructive changes in the myocardium, fibrous scars, and inflammatory process were less pronounced. MMSC-UC also contributed to normalization of the morphofunctional parameters of the heart. Spheroids exhibited higher efficiency in comparison with cell suspension.


Subject(s)
Endomyocardial Fibrosis/prevention & control , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/physiology , Myocardial Infarction/therapy , Regeneration/physiology , Spheroids, Cellular/transplantation , Animals , Disease Models, Animal , Endomyocardial Fibrosis/chemically induced , Endomyocardial Fibrosis/pathology , Endomyocardial Fibrosis/physiopathology , Heart Ventricles/pathology , Heart Ventricles/ultrastructure , Humans , Isoproterenol/administration & dosage , Male , Mesenchymal Stem Cells/cytology , Myocardial Infarction/chemically induced , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/pathology , Myocardium/ultrastructure , Myocytes, Cardiac/pathology , Myocytes, Cardiac/ultrastructure , Primary Cell Culture , Rats , Rats, Wistar , Spheroids, Cellular/cytology , Spheroids, Cellular/physiology , Transplantation, Heterologous , Treatment Outcome , Umbilical Cord/cytology , Umbilical Cord/metabolism
2.
J Vet Med Sci ; 81(1): 127-133, 2019 Jan 25.
Article in English | MEDLINE | ID: mdl-30464077

ABSTRACT

Fibrotic lesion is formed by myofibroblasts capable of producing collagens. The myofibroblasts are characterized by immunoexpressions of vimentin, desmin and α-smooth muscle actin (α-SMA) in varying degrees. The cellular characteristics remain investigated in myocardial fibrosis. We analyzed immunophenotypes of myofibroblasts appearing in isoproterenol-induced myocardial fibrosis in rats until 28 days after injection (10 mg/kg body weight); the lesions developed as interstitial edema and inflammatory cell reaction on 8 hr and days 1 and 3, and fibrosis occurred on days 1, 3, 7, 14, and 21 by gradual deposition of collagens, showing the greatest grade on day 14; the lesions gradually reduced with sporadic scar until day 28. Myofibroblasts expressing vimentin and α-SMA increased with a peak on day 3, and then, gradually decreased onwards. Interestingly, Thy-1 expressing cells appeared in the affected areas, apparently being corresponding to the grade similar to vimentin- and α-SMA-positive cells. Thy-1 is expressed in immature mesenchymal cells such as pericytes with pluripotent nature. The immunoreactivity for A3-antigen, a marker for immature mesenchymal cells, was seen in some surrounding cells. There were no cells reacting with antibodies to nestin or glial fibrillary acidic protein, although hepatic myofibroblats have been reported to react with these antibodies. Collectively, myofibroblasts appearing in rat myocardial fibrosis may have been derived from immature mesenchymal cells positive for Thy-1 or A3-antigen, with thereafter showing expressions of vimentin and α-SMA in differentiation.


Subject(s)
Cardiomyopathies/chemically induced , Endomyocardial Fibrosis/chemically induced , Isoproterenol/toxicity , Myofibroblasts/drug effects , Adrenergic beta-Agonists/toxicity , Animals , Endomyocardial Fibrosis/pathology , Gene Expression Regulation/drug effects , Immunohistochemistry , Male , Myofibroblasts/metabolism , RNA, Messenger , Rats , Rats, Sprague-Dawley
3.
Biochem Biophys Res Commun ; 491(2): 329-336, 2017 09 16.
Article in English | MEDLINE | ID: mdl-28733035

ABSTRACT

Hyperglycemia plays a crucial role in the pathogenesis of diabetic complications; however, the mechanisms underlying diabetic cardiac fibrosis remain unclear. Endothelial cells are known to contribute to cardiac fibrosis through endothelial-mesenchymal transition (EndMT) under high glucose stimulation. Here we investigated the expression of miR-18a-5p and examined its functional role in human aortic valvular endothelial cells (HAVECs). Using HAVECs, we revealed that miR-18a-5p regulated high glucose-induced EndMT. Moreover, high glucose levels induced Notch2 expression, which promoted EndMT, resulting in the downregulation of vascular endothelial cadherin and CD31 and upregulation of fibroblast-specific protein-1, α-smooth muscle actin, fibronectin, and vimentin. Furthermore, Notch2 was identified as a target of miR-18a-5p. Our data showed that the overexpression of miR-18a-5p could downregulate Notch2 expression and subsequently suppress EndMT. In conclusion, our findings demonstrated that miR-18a-5p/Notch2 signaling pathway participates in the regulation of high glucose-induced EndMT, and may act as a novel promising target for myocardial fibrosis in diabetic cardiomyopathy.


Subject(s)
Dependovirus/genetics , Diabetic Cardiomyopathies/genetics , Endomyocardial Fibrosis/genetics , Epithelial-Mesenchymal Transition , MicroRNAs/genetics , Receptor, Notch2/genetics , Actins/genetics , Actins/metabolism , Animals , Aortic Valve/cytology , Aortic Valve/drug effects , Aortic Valve/metabolism , Cadherins/genetics , Cadherins/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Line , Dependovirus/metabolism , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetic Cardiomyopathies/chemically induced , Diabetic Cardiomyopathies/pathology , Diabetic Cardiomyopathies/prevention & control , Endomyocardial Fibrosis/chemically induced , Endomyocardial Fibrosis/pathology , Endomyocardial Fibrosis/prevention & control , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Fibronectins/genetics , Fibronectins/metabolism , Gene Expression Regulation , Genetic Vectors/administration & dosage , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Glucose/pharmacology , Heart Ventricles/metabolism , Heart Ventricles/pathology , Humans , Male , Mice, Inbred C57BL , MicroRNAs/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Receptor, Notch2/metabolism , S100 Calcium-Binding Protein A4 , Signal Transduction , Streptozocin , Vimentin/genetics , Vimentin/metabolism
4.
Phytomedicine ; 30: 10-17, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28545665

ABSTRACT

BACKGROUND: Astragaloside Ⅳ (ASG-Ⅳ, (Fig. 1) is the most active component of Chinese sp. Astragalus membranaceus Bunge (Fabaceae) that has showed antioxidant, antiapoptotic and antiviral activities among others. It is reported to play an important role in cardiac fibrosis (CF), but the mechanism remains unclear. PURPOSE: To investigate the mechanism of ASG-Ⅳ on inhibiting myocardial fibrosis induced by hypoxia. STUDY DESIGN: We studied the relationship between anti-fibrotic effect of ASG-Ⅳ and transient receptor potential cation channel, subfamily M, member 7 (TRPM7) by in vivo and in vitro experiments. METHODS: In vivo, CF was induced by subcutaneous isoproterenol (ISO) for 10 days. Rat hearts were resected for histological experiment and reverse transcription real-time quantitative poly merase chain reaction (RT-qPCR). In vitro, molecular and cellular biology technologies were used to confirm the anti-fibrosis effect underlying mechanism of ASG-Ⅳ. RESULTS: Histological findings and the collagen volume fraction showed that ASG-Ⅳ decreased fibrosis in heart tissues. Hypoxia could stimulate the proliferation and differentiation of cardiac fibroblast which indicated that the degree of fibrosis was increased significantly. Anoxic treatment could also obviously up-regulate the expression of TRPM7 protein and current. ASG-Ⅳ groups showed the opposite results. Knock-down TRPM7 experiment further confirmed the role of TRPM7 channel in hypoxia-induced cardiac fibrosis. CONCLUSION: Our results suggest that the inhibition of hypoxia-induced CF in vivo and in vitro by ASG-IV is associated with reduction of the expression of TRPM7. The moderate inhibition of the TRPM7 channel may be a new strategy for treating cardiac fibrosis.


Subject(s)
Endomyocardial Fibrosis/drug therapy , Endomyocardial Fibrosis/metabolism , Saponins/pharmacology , TRPM Cation Channels/metabolism , Triterpenes/pharmacology , Animals , Cell Differentiation/drug effects , Cell Hypoxia/drug effects , Cell Proliferation/drug effects , Endomyocardial Fibrosis/chemically induced , Fibroblasts/drug effects , Fibroblasts/metabolism , Heart/drug effects , Isoproterenol/pharmacology , Isoproterenol/toxicity , Male , Mice , NIH 3T3 Cells/drug effects , Rats , Rats, Sprague-Dawley , TRPM Cation Channels/genetics , Up-Regulation
5.
Cell Physiol Biochem ; 41(3): 1167-1178, 2017.
Article in English | MEDLINE | ID: mdl-28245473

ABSTRACT

BACKGROUND: The effect of relaxin and spironolactone combined on myocardial fibrosis has not been reported. Thus, we investigated the effect of the combined therapy on isoprenaline-induced myocardial fibrosis and the mechanism. METHODS: Rats were injected subcutaneously with isoprenaline to induce myocardial fibrosis and underwent subcutaneous injection with relaxin (2 µg·kg-1·d-1) and given a gavage of spironolactone (30 mg·kg-1·d-1) alone or combined for 14 days. In vitro, the endothelial-mesenchymal transition was induced with transforming growth factor ß (TGF-ß) in human umbilical vein endothelial cells (HUVECs) pretreated with relaxin, 200 ng/ml, and/or spironolactone, 1uM. RESULTS: Relaxin and spironolactone used alone or combined improved cardiac function and decreased cardiac weight indices; reduced fibrous tissue proliferation; reduced levels of type I and III collagen; decreased the expression of α-smooth muscle actin (α-SMA) and transforming growth factor-ß1 (TGF-ß1), and increased the expression of cluster of differentiation-31 (CD31) in rats with isoprenaline-induced myocardial fibrosis. In vitro, compared with TGF-ß treatment, relaxin and spironolactone used alone or combined with TGF-ß decreased cell mobility, α-SMA and vimentin levels but increased vascular endothelial cadherin (VE-cadherin) and endothelial CD31levels. Especially, combined therapy had more remarkable effect than relaxin and spironolactone used alone both in vitro and in vivo. CONCLUSION: Relaxin and spironolactone combined affected isoprenaline-induced myocardial fibrosis in rats that the mechanism might be inhibition of the cardiac endothelial-mesenchymal transition.


Subject(s)
Cardiotonic Agents/pharmacology , Endomyocardial Fibrosis/drug therapy , Epithelial-Mesenchymal Transition/drug effects , Relaxin/pharmacology , Spironolactone/pharmacology , Actins/genetics , Actins/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Cadherins/genetics , Cadherins/metabolism , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type III/genetics , Collagen Type III/metabolism , Drug Administration Schedule , Drug Synergism , Drug Therapy, Combination , Endomyocardial Fibrosis/chemically induced , Endomyocardial Fibrosis/genetics , Endomyocardial Fibrosis/pathology , Human Umbilical Vein Endothelial Cells , Humans , Isoproterenol , Male , Myocardium/metabolism , Myocardium/pathology , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Rats , Rats, Sprague-Dawley , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/pharmacology , Vimentin/genetics , Vimentin/metabolism
6.
Toxicol Lett ; 241: 143-51, 2016 Jan 22.
Article in English | MEDLINE | ID: mdl-26541207

ABSTRACT

Abuse of anabolic androgenic steroids is linked to a variety of cardiovascular complications. The aim of our study was to investigate the possible cardiovascular effects of nandrolone decanoate on young rabbits using echocardiography, histology and monitoring of telomerase activity, oxidative stress and biochemical markers. Fourteen rabbits were divided into three administration groups and the control group. Doses of 4mg/kg and 10mg/kg of nandrolone decanoate, given intramuscularly and subcutaneously, two days per week for six months were applied. A 4-months wash-out period followed. Focal fibrosis and inflammatory infiltrations of cardiac tissue were observed in the high dose groups. Thiobarbituric acid-reactive species (TBARS) levels were significantly increased in the high dose groups, while catalase activity decreased. Myocardial Performance Index (MPI) is the main echocardiographic index primarily affected by nandrolone administration in rabbits. Despite the preserved systolic performance, histological lesions observed associated with distorted MPI values, point to diastolic impairment of the thickened myocardium due to nandrolone treatment. Oxidative stress accumulates and telomerase activity in cardiac tissue rises. Subcutaneous administration seems to be more deleterious to the cardiovascular system, as oxidative stress, telomerase activity and biochemical markers do not appear to return into normal values in the wash-out period.


Subject(s)
Anabolic Agents/toxicity , Heart Diseases/chemically induced , Nandrolone/analogs & derivatives , Animals , Antioxidants/metabolism , Biomarkers/analysis , Cardiotoxicity , Catalase/metabolism , Endomyocardial Fibrosis/chemically induced , Endomyocardial Fibrosis/pathology , Heart Diseases/diagnostic imaging , Heart Diseases/pathology , Injections, Intramuscular , Injections, Subcutaneous , Male , Myocardium/pathology , Nandrolone/toxicity , Nandrolone Decanoate , Oxidative Stress/drug effects , Rabbits , Telomerase/metabolism , Thiobarbituric Acid Reactive Substances/metabolism , Ultrasonography
7.
Toxicol Lett ; 241: 9-18, 2016 Jan 22.
Article in English | MEDLINE | ID: mdl-26581635

ABSTRACT

Sunitinib (Su) is currently approved for treatment of several malignances. However, along with the benefits of disease stabilization, cardiovascular toxicities have also been increasingly recognized. The aim of this study was to analyze which mechanisms are involved in the cardiotoxicity caused by Su, as well as to explore the potential cardioprotective effects of l-carnitine (LC). To this end, four groups of Wistar rats were used: (1) control; (2) rats treated with 400mg LC/kg/day; (3) rats treated with 25mg Su/kg/day; and (4) rats treated with LC+Su simultaneously. In addition, cultured rat cardiomyocytes were treated with an inhibitor of nuclear factor kappa B (NF-κB), in order to examine the role of this transcription factor in this process. An elevation in the myocardial expression of pro-inflammatory cytokines, together with an increase in the mRNA expression of NF-κB, was observed in Su-treated rats. These results were accompanied by an increase in the expression of pro-fibrotic factors, nitrotyrosine and NOX 2 subunit of NADPH oxidase; and by a decrease in that of collagen degradation factor. Higher blood pressure and heart rate levels were also found in Su-treated rats. All these alterations were inhibited by co-administration of LC. Furthermore, cardiotoxic effects of Su were blocked by NF-κB inhibition. Our results suggest that: (i) inflammatory and fibrotic processes are involved in the cardiac toxicity observed following treatment with Su; (ii) these processes might be mediated by the transcription factor NF-κB; (iii) LC exerts a protective effect against arterial hypertension, cardiac inflammation and fibrosis, which are all observed after Su treatment.


Subject(s)
Antineoplastic Agents/toxicity , Cardiotonic Agents/pharmacology , Carnitine/pharmacology , Heart Diseases/chemically induced , Heart Diseases/prevention & control , Indoles/antagonists & inhibitors , Indoles/toxicity , Myocarditis/chemically induced , Myocarditis/prevention & control , Pyrroles/antagonists & inhibitors , Pyrroles/toxicity , Animals , Blood Pressure/drug effects , Cardiotoxicity , Cytokines/biosynthesis , Endomyocardial Fibrosis/chemically induced , Endomyocardial Fibrosis/pathology , Endomyocardial Fibrosis/prevention & control , Gene Expression/drug effects , Heart Diseases/pathology , Male , Myocarditis/pathology , Myocytes, Cardiac/drug effects , NF-kappa B/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Wistar , Sunitinib
8.
PLoS One ; 10(4): e0125513, 2015.
Article in English | MEDLINE | ID: mdl-25923922

ABSTRACT

MicroRNA-7a/b (miR-7a/b) protects cardiac myocytes from apoptosis during ischemia/reperfusion injury; however, its role in angiotensin II (ANG II)-stimulated cardiac fibroblasts (CFs) remains unknown. Therefore, the present study investigated the anti-fibrotic mechanism of miR-7a/b in ANG II-treated CFs. ANG II stimulated the expression of specific protein 1 (Sp1) and collagen I in a dose- and time-dependent manner, and the overexpression of miR-7a/b significantly down-regulated the expression of Sp1 and collagen I stimulated by ANG II (100 nM) for 24 h. miR-7a/b overexpression effectively inhibited MMP-2 expression/activity and MMP-9 expression, as well as CF proliferation and migration. In addition, miR-7a/b also repressed the activation of TGF-ß, ERK, JNK and p38 by ANG II. The inhibition of Sp1 binding activity by mithramycin prevented collagen I overproduction; however, miR-7a/b down-regulation reversed this effect. Further studies revealed that Sp1 also mediated miR-7a/b-regulated MMP expression and CF migration, as well as TGF-ß and ERK activation. In conclusion, miR-7a/b has an anti-fibrotic role in ANG II-treated CFs that is mediated by Sp1 mechanism involving the TGF-ß and MAPKs pathways.


Subject(s)
Angiotensin II/adverse effects , Endomyocardial Fibrosis/metabolism , Fibroblasts/metabolism , MicroRNAs/biosynthesis , Myocardium/metabolism , Sp1 Transcription Factor/metabolism , Transforming Growth Factor beta/metabolism , Angiotensin II/pharmacology , Animals , Cell Movement/drug effects , Collagen Type I/biosynthesis , Endomyocardial Fibrosis/chemically induced , Endomyocardial Fibrosis/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblasts/pathology , Gene Expression Regulation/drug effects , MAP Kinase Signaling System/drug effects , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 9/biosynthesis , Myocardium/pathology , Plicamycin/pharmacology , Rats , Rats, Wistar
9.
Arch Biochem Biophys ; 564: 74-82, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25263961

ABSTRACT

Na(+)/Mg(2+) exchanger plays an important role in cardiovascular system, but the molecular mechanisms still largely remain unknown. The Solute Carrier family 41A1 (SLC41A1), a novel Mg(2+) transporter, recently was found to function as Na(+)/Mg(2+) exchanger, which mainly regulates the intracellular Mg(2+) ([Mg(2+)]i) homeostasis. Our present studies were designed to investigate whether SLC41A1 impacts on the fibrogenesis of cardiac fibroblasts under Ang II stimulation. Our results showed that quinidine, a prototypical inhibitor of Na(+)/Mg(2+) exchanger, inhibited Ang II-induced cardiac fibrosis via attenuating the overexpression of vital biomarkers of fibrosis, including connective tissue growth factor (CTGF), fibronectin (FN) and α-smooth muscle actin (α-SMA). In addition, quinidine also decreased the Ang II-mediated elevation of concentration of intracellular Ca(2+) ([Ca(2+)]i) and extrusion of intracellular Mg(2+). Meanwhile, silencing SLC41A1 by RNA interference also impaired the elevation of [Ca(2+)]i, [Mg(2+)]i efflux and the upregulation of CTGF, FN and α-SMA provoked by Ang II. Furthermore, we found that Ang II-mediated activation of NFATc4 translocation decreased in SLC41A1-siRNA cells. These results support the notion that rapid extrusion of intracellular Mg(2+) is mediated by SLC41A1 and provide the evidence that the intracellular free Ca(2+) concentration is influenced by extrusion of intracellular Mg(2+) which facilitates fibrosis reaction in cardiac fibroblasts.


Subject(s)
Angiotensin II/adverse effects , Calcium Signaling , Calcium/metabolism , Cation Transport Proteins/metabolism , Endomyocardial Fibrosis/metabolism , Magnesium/metabolism , Muscle Proteins/metabolism , Vasoconstrictor Agents/adverse effects , Angiotensin II/pharmacology , Animals , Cation Transport Proteins/genetics , Endomyocardial Fibrosis/chemically induced , Endomyocardial Fibrosis/genetics , Endomyocardial Fibrosis/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Knockdown Techniques , Ion Transport/genetics , Muscle Proteins/genetics , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Rats , Rats, Sprague-Dawley , Vasoconstrictor Agents/pharmacology
10.
PLoS One ; 9(9): e107556, 2014.
Article in English | MEDLINE | ID: mdl-25233448

ABSTRACT

BACKGROUND: Exposure to subclinical levels of lipopolysaccharide (LPS) occurs commonly and is seemingly well tolerated. However, recurrent LPS exposure induces cardiac fibrosis over 2 to 3 months in a murine model, not mediated by the renin-angiotensin system. Subclinical LPS induces cardiac fibrosis by unique mechanisms. METHODS: In C57/Bl6 mice, LPS (10 mg/kg) or saline (control) were injected intraperitoneally once a week for 1-4 weeks. Mice showed no signs of distress, change in activity, appetite, or weight loss. Mice were euthanized after 3 days, 1, 2, or 4 weeks to measure cardiac expression of fibrosis-related genes and potential mediators (measured by QRT-PCR), including micro-RNA (miR) and NADPH oxidase (NOX). Collagen fraction area of the left ventricle was measured with picrosirius red staining. Cardiac fibroblasts isolated from adult mouse hearts were incubated with 0, 0.1, 1.0 or 10 ng/ml LPS for 48 hours. RESULTS: Cardiac miR expression profiling demonstrated decreased miR-29c after 3 and 7 days following LPS, which were confirmed by QRT-PCR. The earliest changes in fibrosis-related genes and mediators that occurred 3 days after LPS were increased cardiac expression of TIMP-1 and NOX-2 (but not of NOX-4). This persisted at 1 and 2 weeks, with additional increases in collagen Iα1, collagen IIIα1, MMP2, MMP9, TIMP1, TIMP2, and periostin. There was no change in TGF-ß or connective tissue growth factor. Collagen fraction area of the left ventricle increased after 2 and 4 weeks of LPS. LPS decreased miR-29c and increased NOX-2 in isolated cardiac fibroblasts. CONCLUSIONS: Recurrent exposure to subclinical LPS induces cardiac fibrosis after 2-4 weeks. Early changes 3 days after LPS were decreased miR-29c and increased NOX2 and TIMP1, which persisted at 1 and 2 weeks, along with widespread activation of fibrosis-related genes. Decreased miR-29c and increased NOX2, which induce cardiac fibrosis in other conditions, may uniquely mediate LPS-induced cardiac fibrosis.


Subject(s)
Cardiomyopathies/chemically induced , Endomyocardial Fibrosis/chemically induced , Hypertrophy, Left Ventricular/chemically induced , Membrane Glycoproteins/biosynthesis , MicroRNAs/genetics , NADPH Oxidases/biosynthesis , Animals , Cell Adhesion Molecules/biosynthesis , Cells, Cultured , Collagen Type I/biosynthesis , Lipopolysaccharides , Male , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 9/biosynthesis , Mice , Mice, Inbred C57BL , MicroRNAs/biosynthesis , NADPH Oxidase 2 , Tissue Inhibitor of Metalloproteinase-1/biosynthesis , Tissue Inhibitor of Metalloproteinase-2/biosynthesis
11.
PLoS One ; 9(8): e103793, 2014.
Article in English | MEDLINE | ID: mdl-25099270

ABSTRACT

Atrial hypertrophy and fibrosis are essential pathological features of atrial fibrillation. Recently, adiponectin has become a protein of interest due to its beneficial effects on cardiovascular diseases. However, the molecular mechanism of atrial structural remodeling and signaling pathways evoked by adiponectin remain unclear. In the present study, we investigated the cardioprotective effect of globular adiponectin (gAcrp) on angiotensin II-induced atrial hypertrophy and fibrosis in neonatal Sprague-Dawley rat. To further investigate the molecular mechanisms underlying the preventive effect of gAcrp, transfection of cells with siRNA was used to suppress the mRNA expression of adiponectin receptor 1 (AdipoR1) and its downstream adaptor protein APPL1. Non-silencing-Cy-3 labelled siRNA was used to determine transfection efficiency using fluorescence microscopy. The expression of atrial natriuretic peptide and procollagen type1 α-1, hypertrophy marker and fibrosis one, respectively, was detected by real-time PCR. Furthermore, the expression of adenosine monophosphate-activated protein kinase (AMPK), phosphatidylinositol 3-kinase (PI3K) and Akt was detected by western blotting. In addition, nuclear p65 translocation activity was analyzed by EMSA supershift assay. Our results showed that AdipoR1 and the adaptor protein APPL1 mediated the protective effects of gAcrp. In addition, the function of adiponectin and phosphorylation of AMPK were prominently diminished by inhibition of PI3K. Furthermore, nuclear factor-κB (NF-κB) transcription was diminished by the specific inhibition of AMPK. Taken together, AMPK pivotally interacts with NF-κB and PI3K, mediating the cardioprotective effect of adiponectin, and may serve as a therapeutic target for preventing atrial hypertrophy and fibrosis. Our present study suggests that gAcrp could ameliorate AngII-induced cardiac hypertrophy and fibrosis in rat atrial cells, which is mediated by the activation of AMPK signaling pathways. APPL1 and AdipoR1 are the key factors involved in the downstream of gAcrp approach.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adiponectin/metabolism , Angiotensin II/adverse effects , Cardiomegaly/metabolism , Endomyocardial Fibrosis/metabolism , Fibroblasts/metabolism , Myocytes, Cardiac/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Adiponectin/metabolism , Vasoconstrictor Agents/adverse effects , AMP-Activated Protein Kinases/metabolism , Angiotensin II/pharmacology , Animals , Cardiomegaly/chemically induced , Cardiomegaly/pathology , Cardiomegaly/prevention & control , Endomyocardial Fibrosis/chemically induced , Endomyocardial Fibrosis/pathology , Endomyocardial Fibrosis/prevention & control , Fibroblasts/pathology , Heart Atria/metabolism , Heart Atria/pathology , Myocytes, Cardiac/pathology , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Vasoconstrictor Agents/pharmacology
12.
Mol Ther ; 22(5): 974-85, 2014 May.
Article in English | MEDLINE | ID: mdl-24569834

ABSTRACT

Loss of miR-29 is associated with cardiac fibrosis. This study examined the role and therapeutic potential of miR-29 in mouse model of hypertension induced by angiotensin II (AngII). By using microRNA microarray, in situ hybridization, and real-time polymerase chain reaction, we found that AngII-induced cardiac fibrosis in the hypertensive heart and in cultured cardiac fibroblasts were associated with downregulation of miR-29a-c via a Smad3-dependent mechanism. In vitro knockdown of miR-29b enhanced but overexpression of miR-29b inhibited AngII-induced fibrosis, revealing a protective role of miR-29b in cardiac fibrosis in response to AngII. This was further demonstrated in vivo by the ability of overexpressing miR-29b in the mouse heart to prevent AngII-mediated cardiac fibrosis and cardiac dysfunction. Importantly, we also found that restored miR-29b in the established hypertensive heart was capable of blocking progressive cardiac fibrosis and improving cardiac dysfunction, demonstrating a therapeutic potential of miR-29b for chronic heart disease. Further studies revealed that targeting the transforming growth factor (TGF)-ß1 coding sequence region, thereby inhibiting TGF-ß/Smad3 signaling, could be a new mechanism by which miR-29b inhibited AngII-induced cardiac fibrosis. In conclusion, miR-29b plays a protective role in AngII-mediated cardiac remodeling and may be a therapeutic agent for cardiac fibrosis by targeting the TGF-ß/Smad3 pathway.


Subject(s)
Endomyocardial Fibrosis/genetics , Hypertension/therapy , MicroRNAs/biosynthesis , MicroRNAs/genetics , Angiotensin II/toxicity , Animals , Disease Models, Animal , Endomyocardial Fibrosis/chemically induced , Endomyocardial Fibrosis/therapy , Gene Expression Regulation/genetics , Gene Knockdown Techniques , Humans , Hypertension/chemically induced , Hypertension/genetics , Mice , MicroRNAs/antagonists & inhibitors , Molecular Targeted Therapy , Signal Transduction/genetics , Smad3 Protein/antagonists & inhibitors , Smad3 Protein/genetics , Smad3 Protein/metabolism , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
13.
Can J Physiol Pharmacol ; 91(11): 951-9, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24117263

ABSTRACT

Quercetin exhibits numerous pharmacological effects, including the capacity for cardioprotection. This study aimed to investigate whether quercetin or its glycoside derivative rutin has any protective action against isoproterenol (ISO) induced cardiac fibrosis, and investigate the structure-activity relationship. Male Wistar rats were injected subcutaneously with ISO (15 mg·(kg body mass)(-1)) to induce experimental cardiac fibrosis. The cardioprotective effect of co-treatment with quercetin (25 or 50 mg·kg(-1)) or rutin (25 or 50 mg·kg(-1)) was investigated in ISO-induced cardiac fibrosis in rats. The administration of quercetin and rutin signifcantly decreased the cardiac weight index and myocardial enzyme activity, increased the activity of superoxide dismutase in the serum, and inhibited the ISO-induced increase in angiotensin II and aldosterone in the plasma. Furthermore, overexpression of transforming growth factor ß1 (TGF-ß1), connective tissue growth factor (CTGF), and excessive deposition of extracellular matrix (ECM) in isoproterenol-treated myocardial tissues were normalized by quercetin and rutin. Our results suggest that both quercetin and rutin exhibited cardioprotective effects in cardiac fibrosis induced by ISO in the rat heart. Moreover, the effects of rutin are weaker than quercetin at the same dose. The mechanism of these effects may be related to antioxidative stress, inhibition of the renin-angiotensin-aldosterone system, decrease in the expression of TGF-ß1 and CTGF, and the subsequent reduction in the deposition of the ECM.


Subject(s)
Antioxidants/pharmacology , Cardiotonic Agents , Endomyocardial Fibrosis/prevention & control , Quercetin/pharmacology , Rutin/pharmacology , Adrenergic beta-Agonists , Animals , Blotting, Western , Endomyocardial Fibrosis/chemically induced , Endomyocardial Fibrosis/enzymology , Extracellular Matrix/drug effects , Glycosides/pharmacology , Heart Function Tests , Hemodynamics/drug effects , Immunohistochemistry , Isoproterenol , Male , Myocardium/enzymology , Myocardium/pathology , Organ Size/drug effects , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , Renin-Angiotensin System/drug effects , Transforming Growth Factor beta/biosynthesis
14.
J Cardiovasc Magn Reson ; 15: 48, 2013 Jun 10.
Article in English | MEDLINE | ID: mdl-23758789

ABSTRACT

BACKGROUND: The late cardiotoxic effects of anthracycline chemotherapy influence morbidity and mortality in the growing population of childhood cancer survivors. Even with lower anthracycline doses, evidence of adverse cardiac remodeling and reduced exercise capacity exist. We aim to examine the relationship between cardiac structure, function and cardiovascular magnetic resonance (CMR) tissue characteristics with chemotherapy dose and exercise capacity in childhood cancer survivors. METHODS: Thirty patients (15 ± 3 years), at least 2 years following anthracycline treatment, underwent CMR, echocardiography, and cardiopulmonary exercise testing (peak VO(2)). CMR measured ventricular function, mass, T(1) and T(2) values, and myocardial extracellular volume fraction, ECV, a measure of diffuse fibrosis based on changes in myocardial T1 values pre- and post-gadolinium. Cardiac function was also assessed with conventional and speckle tracking echocardiography. RESULTS: Patients had normal LVEF (59 ± 7%) but peak VO(2) was 17% lower than age-predicted normal values and were correlated with anthracycline dose (r = -0.49). Increased ECV correlated with decreased mass/volume ratio (r = -0.64), decreased LV wall thickness/height ratio (r = -0.72), lower peak VO(2)(r = -0.52), and higher cumulative dose (r = 0.40). Echocardiographic measures of systolic and diastolic function were reduced compared to normal values (p < 0.01), but had no relation to ECV, peak VO(2) or cumulative dose. CONCLUSIONS: Myocardial T1 and ECV were found to be early tissue markers of ventricular remodeling that may represent diffuse fibrosis in children with normal ejection fraction post anthracycline therapy, and are related to cumulative dose, exercise capacity and myocardial wall thinning.


Subject(s)
Anthracyclines/adverse effects , Endomyocardial Fibrosis/chemically induced , Endomyocardial Fibrosis/diagnosis , Endomyocardial Fibrosis/physiopathology , Magnetic Resonance Imaging/methods , Physical Endurance/physiology , Adolescent , Child , Contrast Media , Cross-Sectional Studies , Echocardiography , Exercise Test , Female , Gadolinium DTPA , Humans , Linear Models , Male , Neoplasms/drug therapy , Oxygen/blood , Risk Factors , Survivors , Ventricular Remodeling/drug effects
15.
PLoS One ; 7(9): e45260, 2012.
Article in English | MEDLINE | ID: mdl-23028889

ABSTRACT

The innate immune system is responsible for the initial response of an organism to potentially harmful stressors, pathogens or tissue injury, and accordingly plays an essential role in the pathogenesis of many inflammatory processes, including some cardiovascular diseases. Toll like receptors (TLR) and nucleotide-binding oligomerization domain-like receptors (NLRs) are pattern recognition receptors that play an important role in the induction of innate immune and inflammatory responses. There is a line of evidence supporting that activation of TLRs contributes to the development and progression of cardiovascular diseases but less is known regarding the role of NLRs. Here we demonstrate the presence of the NLR member NOD1 (nucleotide-binding oligomerization domain containing 1) in the murine heart. Activation of NOD1 with the specific agonist C12-iEDAP, but not with the inactive analogue iE-Lys, induces a time- and dose-dependent cardiac dysfunction that occurs concomitantly with cardiac fibrosis and apoptosis. The administration of iEDAP promotes the activation of the NF-κB and TGF-ß pathways and induces apoptosis in whole hearts. At the cellular level, both native cardiomyocytes and cardiac fibroblasts expressed NOD1. The NLR activation in cardiomyocytes was associated with NF-κB activation and induction of apoptosis. NOD1 stimulation in fibroblasts was linked to NF-κB activation and to increased expression of pro-fibrotic mediators. The down-regulation of NOD1 by specific siRNAs blunted the effect of iEDAP on the pro-fibrotic TGF-ß pathway and cell apoptosis. In conclusion, our report uncovers a new pro-inflammatory target that is expressed in the heart, NOD1. The specific activation of this NLR induces cardiac dysfunction and modulates cardiac fibrosis and cardiomyocyte apoptosis, pathological processes involved in several cardiac diseases such as heart failure.


Subject(s)
Biological Factors/pharmacology , Endomyocardial Fibrosis/metabolism , Heart Failure/metabolism , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Nod1 Signaling Adaptor Protein/agonists , Animals , Apoptosis/drug effects , Dose-Response Relationship, Drug , Endomyocardial Fibrosis/chemically induced , Endomyocardial Fibrosis/complications , Endomyocardial Fibrosis/pathology , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Regulation/drug effects , Heart Failure/chemically induced , Heart Failure/complications , Heart Failure/pathology , Mice , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , NF-kappa B/genetics , NF-kappa B/metabolism , Nod1 Signaling Adaptor Protein/antagonists & inhibitors , Nod1 Signaling Adaptor Protein/genetics , RNA, Small Interfering/genetics , Rats , Signal Transduction/drug effects , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
16.
Trop Doct ; 42(4): 206-7, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22875807

ABSTRACT

The cause of endomyocardial fibrosis in Uganda remains unknown in spite of studies dating back to 1948. Herbal decoctions have been used by traditional healers for countless generations to treat many complaints, especially fever. We present evidence - ethnic, geographical, tribal, pathological and distribution by age - that the toxic herbal decoctions taken as medicines in Uganda can cause endomyocardial fibrosis.


Subject(s)
Argemone/adverse effects , Endomyocardial Fibrosis/chemically induced , Medicine, African Traditional/adverse effects , Phytotherapy/adverse effects , Adult , Child , Female , Humans , Male , Uganda
17.
PLoS One ; 7(8): e42989, 2012.
Article in English | MEDLINE | ID: mdl-22900077

ABSTRACT

Type I collagen is the most abundant protein in the human body. Its excessive synthesis results in fibrosis of various organs. Fibrosis is a major medical problem without an existing cure. Excessive synthesis of type I collagen in fibrosis is primarily due to stabilization of collagen mRNAs. We recently reported that intermediate filaments composed of vimentin regulate collagen synthesis by stabilizing collagen mRNAs. Vimentin is a primary target of Withaferin-A (WF-A). Therefore, we hypothesized that WF-A may reduce type I collagen production by disrupting vimentin filaments and decreasing the stability of collagen mRNAs. This study is to determine if WF-A exhibits anti-fibrotic properties in vitro and in vivo and to elucidate the molecular mechanisms of its action. In lung, skin and heart fibroblasts WF-A disrupted vimentin filaments at concentrations of 0.5-1.5 µM and reduced 3 fold the half-lives of collagen α1(I) and α2(I) mRNAs and protein expression. In addition, WF-A inhibited TGF-ß1 induced phosphorylation of TGF-ß1 receptor I, Smad3 phosphorylation and transcription of collagen genes. WF-A also inhibited in vitro activation of primary hepatic stellate cells and decreased their type I collagen expression. In mice, administration of 4 mg/kg WF-A daily for 2 weeks reduced isoproterenol-induced myocardial fibrosis by 50%. Our findings provide strong evidence that Withaferin-A could act as an anti-fibrotic compound against fibroproliferative diseases, including, but not limited to, cardiac interstitial fibrosis.


Subject(s)
Collagen Type I/genetics , Endomyocardial Fibrosis/genetics , Withanolides/pharmacology , Animals , Cell Line , Collagen Type I/metabolism , Endomyocardial Fibrosis/chemically induced , Endomyocardial Fibrosis/drug therapy , Endomyocardial Fibrosis/pathology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Regulation/drug effects , Gene Knockout Techniques , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Humans , Isoproterenol/adverse effects , Male , Mice , Phosphorylation/drug effects , Promoter Regions, Genetic , Proteolysis/drug effects , RNA Stability/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta1/pharmacology , Vimentin/genetics , Vimentin/metabolism , Withanolides/administration & dosage , Withanolides/toxicity
18.
Zhongguo Zhong Yao Za Zhi ; 37(3): 344-7, 2012 Feb.
Article in Chinese | MEDLINE | ID: mdl-22568237

ABSTRACT

OBJECTIVE: To detect the effect and mechanism of procyanidin foom vaccinium (PC) on myocardial fibrosis in rats. METHOD: The myocardial fibrosis model in rats was built by subcutaneous injection of 5 mg x kg(-1) x d(-1) of isoprenaline (Iso) for 7 days in vivo while intragastrically administrating PC 100, 200 and 400 mg x kg(-1) x d(-1) for 14 days. Following the model was established, myocardial indexes (heart weight/body weight, HW/BW and left ventricalar weight/body weight, LVW/BW) were measured. Besides, angiotensin II and I , III collagen quantification levels in blood serum were determined respectively by ELISA. The change in the content of nitric oxide (NO) in blood serum was determined with colorimetry. The content of malondialdehyde (MDA) in left ventricle was assayed with spectrophotometry. The contents of lactate dehydrogenase (LDH) and creatine kinase (CK) in blood serum were detected with automatic biochemistry analyzer. RESULT: Compared with the control group, the myocardial indexes, the contents of I , III collagen quantification, angiotensin II in blood serum and malondialdehyde in left ventricle were markedly increased and the content of nitric oxide in blood serum was decreased, the contents of lactate dehydrogenase and creatine kinase in blood serum were markedly increased in Iso model group (P<0.05 or P<0.01). Compared with the model group, the myocardial indexes were decreased, the contents of I , III collagen quantification, angiotensin II in blood serum were reduced in PC 200 and 400 mg x kg(-1) x d(-1) groups (P<0.05 or P<0.01). The content of nitric oxide in blood serum was increased, the content of malondialdehyde in left ventricle was markedly decreased, the contents of lactate dehydrogenase and creatine kinase in blood serum were markedly decreased in PC three groups (P<0.05 or P<0.01). CONCLUSION: PC could inhibit collagen synthesis by decreasing angiotensin II level and increasing the content of nitric oxide and antioxidation, and thereby inhibiting myocardial fibrosis and protect myocardium in rats.


Subject(s)
Antioxidants/pharmacology , Biflavonoids/pharmacology , Catechin/pharmacology , Endomyocardial Fibrosis/drug therapy , Endomyocardial Fibrosis/metabolism , Isoproterenol/adverse effects , Proanthocyanidins/pharmacology , Vaccinium/chemistry , Angiotensins/blood , Animals , Antioxidants/administration & dosage , Biflavonoids/administration & dosage , Catechin/administration & dosage , Endomyocardial Fibrosis/chemically induced , Female , Male , Malondialdehyde/metabolism , Nitric Oxide/blood , Proanthocyanidins/administration & dosage , Rats , Rats, Wistar , Ventricular Remodeling/drug effects
19.
Cardiovasc Res ; 85(4): 773-84, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-19942623

ABSTRACT

AIMS: Here we investigated the mechanisms by which cardiovascular CB1 cannabinoid receptors may modulate the cardiac dysfunction, oxidative stress, and interrelated cell death pathways associated with acute/chronic cardiomyopathy induced by the widely used anti-tumour compound doxorubicin (DOX). METHODS AND RESULTS: Both load-dependent and -independent indices of left-ventricular function were measured by the Millar pressure-volume conductance system. Mitogen-activated protein kinase (MAPK) activation, cell-death markers, and oxidative/nitrosative stress were measured by molecular biology/biochemical methods and flow cytometry. DOX induced left-ventricular dysfunction, oxidative/nitrosative stress coupled with impaired antioxidant defense, activation of MAPK (p38 and JNK), and cell death and/or fibrosis in hearts of wide-type mice (CB1(+/+)), and these effects were markedly attenuated in CB1 knockouts (CB1(-/-)). In human primary cardiomyocytes expressing CB1 receptors (demonstrated by RT-PCR, western immunoblot, and flow cytometry) DOX, likewise the CB1 receptor agonist HU210 and the endocannabinoid anandamide (AEA), induced MAPK activation and cell death. The DOX-induced MAPK activation and cell death were significantly enhanced when DOX was co-administered with CB1 agonists AEA or HU210. Remarkably, cell death and MAPK activation induced by AEA, HU210, and DOX +/- AEA/HU210 were largely attenuated by either CB1 antagonists (rimonabant and AM281) or by inhibitors of p38 and JNK MAPKs. Furthermore, AEA or HU210 in primary human cardiomyocytes triggered increased reactive oxygen species generation. CONCLUSION: CB1 activation in cardiomyocytes may amplify the reactive oxygen/nitrogen species-MAPK activation-cell death pathway in pathological conditions when the endocannabinoid synthetic or metabolic pathways are dysregulated by excessive inflammation and/or oxidative/nitrosative stress, which may contribute to the pathophysiology of various cardiovascular diseases.


Subject(s)
Cardiomyopathies/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Oxidative Stress/physiology , Receptor, Cannabinoid, CB1/metabolism , Amidohydrolases/metabolism , Animals , Antibiotics, Antineoplastic/toxicity , Apoptosis/physiology , Cannabinoid Receptor Modulators/metabolism , Cardiomyopathies/chemically induced , Cardiomyopathies/pathology , Caspase 3/metabolism , Caspase 7/metabolism , Cells, Cultured , Cytochromes c/metabolism , Disease Models, Animal , Doxorubicin/toxicity , Endomyocardial Fibrosis/chemically induced , Endomyocardial Fibrosis/metabolism , Endomyocardial Fibrosis/pathology , Humans , MAP Kinase Signaling System/physiology , Male , Mice , Mice, Knockout , Poly(ADP-ribose) Polymerases/metabolism , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Receptor, Cannabinoid, CB1/genetics , Ventricular Function, Left/physiology
20.
Mov Disord ; 24(1): 129-33, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-19170199

ABSTRACT

There is growing evidence that the ergot-derived dopamine agonists cabergoline and pergolide can cause fibrotic cardiac valvulopathy. Data on other fibrotic reactions and nonergot-derived dopamine agonists are sparse. Aim of this study was to investigate whether there are signals that dopamine agonists are related to cardiac and other fibrotic reactions. We identified all reports of fibrotic reactions at the heart, lung, and retroperitoneal space associated with dopamine agonists within the US Adverse Event Reporting System database. Disproportionality analyses were used to calculate adjusted reporting odds ratios (RORs). For ergot-derived dopamine agonists (bromocriptine, cabergoline, pergolide), the RORs of all reactions under study were increased, whereas no such increases were observed for nonergot-derived drugs (apomorphine, pramipexole, ropinirole, rotigotine). Fibrotic reactions due to ergot-derived dopamine agonists may not be limited to heart valves. For nonergot-derived dopamine agonists, no drug safety signals were evident.


Subject(s)
Bromocriptine/adverse effects , Dopamine Agonists/adverse effects , Endomyocardial Fibrosis/chemically induced , Ergolines/adverse effects , Heart Valve Diseases/chemically induced , Pergolide/adverse effects , Pericarditis/chemically induced , Pleural Diseases/chemically induced , Pulmonary Fibrosis/chemically induced , Retroperitoneal Fibrosis/chemically induced , Aged , Aged, 80 and over , Apomorphine/adverse effects , Benzothiazoles/adverse effects , Cabergoline , Databases, Factual , Endomyocardial Fibrosis/epidemiology , Female , Fibrosis/chemically induced , Fibrosis/epidemiology , Heart Valve Diseases/epidemiology , Heart Valve Diseases/pathology , Humans , Indoles/adverse effects , Male , Middle Aged , Pericarditis/epidemiology , Pericarditis/pathology , Pleural Diseases/epidemiology , Pramipexole , Pulmonary Fibrosis/epidemiology , Retroperitoneal Fibrosis/epidemiology , Tetrahydronaphthalenes/adverse effects , Thiophenes/adverse effects , United States/epidemiology
SELECTION OF CITATIONS
SEARCH DETAIL
...