Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
1.
Arterioscler Thromb Vasc Biol ; 40(11): 2649-2664, 2020 11.
Article in English | MEDLINE | ID: mdl-32938216

ABSTRACT

OBJECTIVE: Phenotypic switching of vascular smooth muscle cells (VSMCs) plays a critical role in atherosclerosis, vascular restenosis, and hypertension. Choline exerts cardioprotective effects; however, little is known about its effects on VSMC phenotypic switching and vascular remodeling. Here, we investigated whether choline modulates VSMC phenotypic changes and explored the underlying mechanisms. Approach and Results: In cultured VSMCs, choline promoted Nrf2 (nuclear factor erythroid 2-related factor 2) nuclear translocation, inducing the expression of HO-1 (heme oxygenase-1) and NQO-1 (NAD[P]H quinone oxidoreductase-1). Consequently, choline ameliorated Ang II (angiotensin II)-induced increases in NOX (NAD[P]H oxidase) expression and the mitochondrial reactive oxygen species level, thereby attenuating Ang II-induced VSMC phenotypic switching, proliferation, and migration, presumably via M3AChRs (type 3 muscarinic acetylcholine receptors). Downregulation of M3AChR or Nrf2 diminished choline-mediated upregulation of Nrf2, HO-1, and NQO-1 expression, as well as inhibition of VSMC phenotypic transformation, suggesting that M3AChR and Nrf2 activation are responsible for the protective effects of choline. Moreover, activation of the Nrf2 pathway by sulforaphane suppressed Ang II-induced VSMC phenotypic switching and proliferation, indicating that Nrf2 is a key regulator of VSMC phenotypic switching and vascular homeostasis. In a rat model of abdominal aortic constriction in vivo, choline attenuated VSMC phenotypic transformation and vascular remodeling in a manner related to activation of the Nrf2 pathway. CONCLUSIONS: These results reveal that choline impedes VSMC phenotypic switching, proliferation, migration, and vascular remodeling by activating M3AChR and Nrf2-antioxidant signaling and suggest a novel role for Nrf2 in VSMC phenotypic modulation.


Subject(s)
Cell Plasticity/drug effects , Choline/pharmacology , Muscarinic Agonists/pharmacology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , NF-E2-Related Factor 2/metabolism , Receptor, Muscarinic M3/agonists , Vascular Remodeling/drug effects , Animals , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Male , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , NF-E2-Related Factor 2/genetics , Phenotype , Rats, Sprague-Dawley , Receptor, Muscarinic M3/genetics , Receptor, Muscarinic M3/metabolism , Signal Transduction
2.
Free Radic Biol Med ; 145: 103-117, 2019 12.
Article in English | MEDLINE | ID: mdl-31553938

ABSTRACT

Mitochondrial dysfunction is associated with obesity-induced cardiac remodelling. Recent research suggests that the cristae are the true bioenergetic components of cells. Acetylcholine (ACh), the major neurotransmitter of the vagus nerve, exerts cardio-protective effects against ischaemia. This study investigated the role of cristae remodelling in palmitate (PA)-induced neonatal rat cardiomyocyte hypertrophy and explored the beneficial effects of ACh. We found loose, fragmented and even lysed cristae in PA-treated neonatal cardiomyocytes along with declines in mitochondrial network and complex expression and overproduction of mitochondrial reactive oxygen species (ROS); these changes ultimately resulted in increased myocardial size. Overexpression of mitofilin by adenoviral infection partly improved cristae shape, mitochondrial network, and ATP content and attenuated cell hypertrophy. Interestingly, siRNA-mediated AMP-activated protein kinase (AMPK) silencing increased the number of cristae with a balloon-like morphology without disturbing mitofilin expression. Furthermore, AMPK knockdown abolished the effects of mitofilin overexpression on cristae remodelling and inhibited the interaction of mitofilin with sorting and assembly machinery 50 (Sam50) and coiled-coil helix coiled-coil helix domain-containing protein 3 (CHCHD3), two core components of the mitochondrial contact site and cristae organizing system (MICOS) complex. Intriguingly, ACh upregulated mitofilin expression and AMPK phosphorylation via the muscarinic ACh receptor (MAChR). Moreover, ACh enhanced protein-protein interactions between mitofilin and other components of the MICOS complex, thereby preventing PA-induced mitochondrial dysfunction and cardiomyocyte hypertrophy; however, these effects were abolished by AMPK silencing. Taken together, our data suggest that ACh improves cristae remodelling to defend against PA-induced myocardial hypertrophy, presumably by increasing mitofilin expression and activating AMPK to form the MICOS complex through MAChR. These results suggest new and promising therapeutic approaches targeting mitochondria to prevent lipotoxic cardiomyopathy.


Subject(s)
G-Protein-Coupled Receptor Kinase 2/genetics , Hypertrophy/drug therapy , Mitochondria/genetics , Mitochondrial Proteins/genetics , Muscle Proteins/genetics , Protein Kinases/genetics , AMP-Activated Protein Kinase Kinases , Acetylcholine/metabolism , Animals , Animals, Newborn/genetics , Atrial Remodeling/drug effects , Atrial Remodeling/genetics , Disease Models, Animal , G-Protein-Coupled Receptor Kinase 2/metabolism , Gene Expression Regulation/drug effects , Humans , Hypertrophy/chemically induced , Hypertrophy/metabolism , Hypertrophy/pathology , Mitochondria/drug effects , Mitochondria/pathology , Mitochondrial Proteins/metabolism , Muscle Proteins/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Obesity/drug therapy , Obesity/genetics , Obesity/metabolism , Obesity/pathology , Palmitates/toxicity , Phosphorylation , Protein Interaction Maps/drug effects , Protein Transport , RNA, Small Interfering/pharmacology , Rats , Vagus Nerve/drug effects , Vagus Nerve/pathology
3.
Am J Physiol Endocrinol Metab ; 317(2): E312-E326, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31211620

ABSTRACT

Diabetic patients are more susceptible to myocardial ischemia damage than nondiabetic patients, with worse clinical outcomes and greater mortality. The mechanism may be related to glucose metabolism, mitochondrial homeostasis, and oxidative stress. Pyridostigmine may improve vagal activity to protect cardiac function in cardiovascular diseases. Researchers have not determined whether pyridostigmine regulates glucose metabolism and mitochondrial homeostasis to reduce myocardial vulnerability to injury in diabetic mice. In the present study, autonomic imbalance, myocardial damage, mitochondrial dysfunction, and oxidative stress were exacerbated in isoproterenol-stimulated diabetic mice, revealing the myocardial vulnerability of diabetic mice to injury compared with mice with diabetes or exposed to isoproterenol alone. Compared with normal mice, the expression of glucose transporters (GLUT)1/4 phosphofructokinase (PFK) FB3, and pyruvate kinase isoform (PKM) was decreased in diabetic mice, but increased in isoproterenol-stimulated normal mice. Following exposure to isoproterenol, the expression of (GLUT)1/4 phosphofructokinase (PFK) FB3, and PKM decreased in diabetic mice compared with normal mice. The downregulation of SIRT3/AMPK and IRS-1/Akt in isoproterenol-stimulated diabetic mice was exacerbated compared with that in diabetic mice or isoproterenol-stimulated normal mice. Pyridostigmine improved vagus activity, increased GLUT1/4, PFKFB3, and PKM expression, and ameliorated mitochondrial dysfunction and oxidative stress to reduce myocardial damage in isoproterenol-stimulated diabetic mice. Based on these results, it was found that pyridostigmine may reduce myocardial vulnerability to injury via the SIRT3/AMPK and IRS-1/Akt pathways in diabetic mice with isoproterenol-induced myocardial damage. This study may provide a potential therapeutic target for myocardial damage in diabetic patients.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Cardiomyopathies/prevention & control , Glucose/metabolism , Mitochondria, Heart/drug effects , Myocardial Ischemia/prevention & control , Pyridostigmine Bromide/pharmacology , Animals , Carbohydrate Metabolism/drug effects , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetic Cardiomyopathies/metabolism , Diabetic Cardiomyopathies/pathology , Isoproterenol/pharmacology , Male , Mice , Mice, Inbred C57BL , Mitochondria, Heart/physiology , Myocardial Ischemia/metabolism , Myocardial Ischemia/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Oxidative Stress/drug effects , Pyridostigmine Bromide/therapeutic use
4.
Sheng Li Xue Bao ; 71(2): 216-224, 2019 Apr 25.
Article in Chinese | MEDLINE | ID: mdl-31008481

ABSTRACT

Obesity is an important risk factor for cardiovascular diseases, which can lead to a variety of cardiovascular diseases including myocardial remodeling. Obesity may induce myocardial dysfunction by affecting hemodynamics, inducing autonomic imbalance, adipose tissue dysfunction, and mitochondrial dyshomeostasis. The key necessary biochemical functions for metabolic homeostasis are performed in mitochondria, and mitochondrial homeostasis is considered as one of the key determinants for cell viability. Mitochondrial homeostasis is regulated by dynamic regulation of mitochondrial fission and fusion, as well as mitochondrial cristae remodeling, biogenesis, autophagy, and oxidative stress. The mitochondrial fission-fusion and morphological changes of mitochondrial cristae maintain the integrity of the mitochondrial structure. The mitochondria maintain a "healthy" state by balancing biogenesis and autophagy, while reactive oxygen species can act as signaling molecules to regulate intracellular signaling. The excessive accumulation of lipids and lipid metabolism disorder in obesity leads to mitochondrial dyshomeostasis, which activate the apoptotic cascade and lead to myocardial remodeling. In this review, we provide an overview of the recent research progress on obesity-induced myocardial remodeling and its possible mechanism of mitochondrial dyshomeostasis.


Subject(s)
Mitochondria/pathology , Mitochondrial Dynamics , Myocardium/pathology , Obesity/physiopathology , Humans , Reactive Oxygen Species
5.
Life Sci ; 222: 1-12, 2019 Apr 01.
Article in English | MEDLINE | ID: mdl-30786250

ABSTRACT

AIMS: Obesity is associated with increased cardiovascular morbidity and mortality. It is accompanied by augmented O-linked ß-N-acetylglucosamine (O-GlcNAc) modification of proteins via increasing hexosamine biosynthetic pathway (HBP) flux. However, the changes and regulation of the O-GlcNAc levels induced by obesity are unclear. MAIN METHODS: High fat diet (HFD) model was induced obesity in mice with or without the cholinergic drug pyridostigmine (PYR, 3 mg/kg/d) for 22 weeks and in vitro human umbilical vein endothelial cells (HUVECs) was treated with high glucose (HG, 30 mM) with or without acetylcholine (ACh). KEY FINDINGS: PYR significantly reduced body weight, blood glucose, and O-GlcNAcylation levels and attenuated vascular endothelial cells detachment in HFD-fed mice. HG addition induced endoplasmic reticulum (ER) stress and increased O-GlcNAcylation levels and apoptosis in HUVECs in a time-dependent manner. Additionally, HG decreased levels of phosphorylated AMP-activated protein kinase (AMPK). Interestingly, ACh significantly blocked damage to HUVECs induced by HG. Furthermore, the effects of ACh on HG-induced ER stress, O-GlcNAcylation, and apoptosis were prevented by treating HUVECs with 4-diphenylacetoxy-N-methylpiperidine methiodide (4-DAMP, a selective M3 AChR antagonist) or compound C (Comp C, an AMPK inhibitor). Treatment with 5-aminoimidazole-4-carboxamide ribose (AICAR, an AMPK activator), 4-phenyl butyric acid (4-PBA, an ER stress inhibitor), and 6-diazo-5-oxonorleucine (DON, a GFAT antagonist) reproduced a similar effect with ACh. SIGNIFICANCE: Activation of cholinergic signaling ameliorated endothelium damage, reduced levels of ER stress, O-GlcNAcylation, and apoptosis in mice and HUVECs under obese conditions, which may function through M3 AChR-AMPK signaling.


Subject(s)
Acetylglucosamine/metabolism , Cholinergic Agents/pharmacology , Endoplasmic Reticulum Stress/physiology , Endothelium, Vascular/metabolism , Protein Kinases/metabolism , Receptor, Muscarinic M3/metabolism , AMP-Activated Protein Kinase Kinases , Acetylcholine/pharmacology , Acetylglucosamine/antagonists & inhibitors , Animals , Cholinesterase Inhibitors/pharmacology , Diet, High-Fat/adverse effects , Dose-Response Relationship, Drug , Endoplasmic Reticulum Stress/drug effects , Endothelium, Vascular/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Pyridostigmine Bromide/pharmacology , Receptor, Muscarinic M3/antagonists & inhibitors
6.
Free Radic Biol Med ; 134: 119-132, 2019 04.
Article in English | MEDLINE | ID: mdl-30633969

ABSTRACT

Insulin resistance and autonomic imbalance are important pathological processes in metabolic syndrome-induced cardiac remodeling. Recent studies determined that disruption of mitochondrial cristae shape is associated with myocardial ischemia; however, the change in cristae shape in metabolic syndrome-induced cardiac remodeling remains unclear. This study determined the effect of pyridostigmine (PYR), which reversibly inhibits cholinesterase to improve autonomic imbalance, on high-fat diet (HFD)-induced cardiac insulin resistance and explored the potential effect on the shape of mitochondrial cristae. Feeding of a HFD for 22 weeks led to an irregular and even lysed cristae structure in cardiac mitochondria, which contributed to decreased mitochondrial content and ATP production and increased oxygen species production, ultimately impairing insulin signaling and lipid metabolism. Interestingly, PYR enhanced vagal activity by increasing acetylcholine production and exerted mito-protective effects by activating the LKB1/AMPK/ACC signal pathway. Specifically, PYR upregulated OPA1 and Mfn1/2 expression, promoted the formation of the mitofilin/CHCHD3/Sam50 complex, and decreased p-Drp1 and Fis1 expression, resulting in tight and parallel cristae and increasing cardiac mitochondrial complex subunit expression and ATP generation as well as decreasing release of cytochrome C from mitochondria and oxidative damage. Furthermore, PYR improved glucose and insulin tolerance and insulin-stimulated Akt phosphorylation, decreased lipid toxicity, and ultimately ameliorated HFD-induced cardiac remodeling and dysfunction. In conclusion, PYR prevented cardiac and insulin insensitivity and remodeling by stimulating vagal activity to regulate mitochondrial cristae shape and function in HFD-induced metabolic syndrome in mice. These results provide novel insights for the development of a therapeutic strategy for obesity-induced cardiac dysfunction that targets mitochondrial cristae.


Subject(s)
Disease Models, Animal , Heart Diseases/prevention & control , Insulin Resistance , Metabolic Syndrome/prevention & control , Mitochondria, Heart/physiology , Mitochondrial Membranes/chemistry , Pyridostigmine Bromide/pharmacology , Animals , Cholinesterase Inhibitors/pharmacology , Diet, High-Fat/adverse effects , Heart Diseases/etiology , Male , Metabolic Syndrome/complications , Mice , Mitochondrial Proteins/metabolism , Organelle Shape
7.
Cardiovasc Res ; 115(3): 530-545, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30165480

ABSTRACT

AIMS: Cardiac hypertrophy is characterized by a shift in metabolic substrate utilization, but the molecular events underlying the metabolic remodelling remain poorly understood. We explored metabolic remodelling and mitochondrial dysfunction in cardiac hypertrophy and investigated the cardioprotective effects of choline. METHODS AND RESULTS: The experiments were conducted using a model of ventricular hypertrophy by partially banding the abdominal aorta of Sprague Dawley rats. Cardiomyocyte size and cardiac fibrosis were significantly increased in hypertrophic hearts. In vitro cardiomyocyte hypertrophy was induced by exposing neonatal rat cardiomyocytes to angiotensin II (Ang II) (10-6 M, 24 h). Choline attenuated the mito-nuclear protein imbalance and activated the mitochondrial-unfolded protein response (UPRmt) in the heart, thereby preserving the ultrastructure and function of mitochondria in the context of cardiac hypertrophy. Moreover, choline inhibited myocardial metabolic dysfunction by promoting the expression of proteins involved in ketone body and fatty acid metabolism in response to pressure overload, accompanied by the activation of sirtuin 3/AMP-activated protein kinase (SIRT3-AMPK) signalling. In vitro analyses demonstrated that SIRT3 siRNA diminished choline-mediated activation of ketone body metabolism and UPRmt, as well as inhibition of hypertrophic signals. Intriguingly, serum from choline-treated abdominal aorta banding models (where ß-hydroxybutyrate was increased) attenuated Ang II-induced myocyte hypertrophy, which indicates that ß-hydroxybutyrate is important for the cardioprotective effects of choline. CONCLUSION: Choline attenuated cardiac dysfunction by modulating the expression of proteins involved in ketone body and fatty acid metabolism, and induction of UPRmt; this was likely mediated by activation of the SIRT3-AMPK pathway. Taken together, these results identify SIRT3-AMPK as a key cardiac transcriptional regulator that helps orchestrate an adaptive metabolic response to cardiac stress. Choline treatment may represent a new therapeutic strategy for optimizing myocardial metabolism in the context of hypertrophy and heart failure.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Choline/pharmacology , Energy Metabolism/drug effects , Hypertrophy, Left Ventricular/prevention & control , Mitochondria, Heart/drug effects , Myocytes, Cardiac/drug effects , Sirtuins/metabolism , Unfolded Protein Response/drug effects , Animals , Cells, Cultured , Disease Models, Animal , Fatty Acids/metabolism , Fibrosis , Hypertrophy, Left Ventricular/enzymology , Hypertrophy, Left Ventricular/pathology , Hypertrophy, Left Ventricular/physiopathology , Ketone Bodies/metabolism , Mitochondria, Heart/enzymology , Mitochondria, Heart/pathology , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Rats, Sprague-Dawley , Signal Transduction , Ventricular Function, Left/drug effects , Ventricular Remodeling/drug effects
8.
ACS Appl Mater Interfaces ; 10(11): 9774-9781, 2018 Mar 21.
Article in English | MEDLINE | ID: mdl-29517231

ABSTRACT

Along with an unexpected conducting interface between nonmagnetic insulating perovskites LaAlO3 and SrTiO3 (LaAlO3/SrTiO3), striking interfacial magnetisms have been observed in LaAlO3/SrTiO3 heterostructures. Interestingly, the strength of the interfacial magnetic moment is found to be dependent on oxygen partial pressures during the growth process. This raises an important, fundamental question on the origin of these remarkable interfacial magnetic orderings. Here, we report a direct evidence of room-temperature stable magnetism in a LaAlO3/SrTiO3 heterostructure prepared at high oxygen partial pressure by using element-specific soft X-ray magnetic circular dichroism at both Ti L3,2 and O K edges. By combining X-ray absorption spectroscopy at both Ti L3,2 and O K edges and first-principles calculations, we qualitatively ascribe that this strong magnetic ordering with dominant interfacial Ti3+ character is due to the coexistence of LaAlO3 surface oxygen vacancies and interfacial (TiAl-AlTi) antisite defects. On the basis of this new understanding, we revisit the origin of the weak magnetism in LaAlO3/SrTiO3 heterostructures prepared at low oxygen partial pressures. Our calculations show that LaAlO3 surface oxygen vacancies are responsible for the weak magnetism at the interface. Our result provides direct evidence on the presence of room-temperature stable magnetism and a novel perspective to understand magnetic and electronic reconstructions at such strategic oxide interfaces.

9.
Biochim Biophys Acta Mol Basis Dis ; 1864(4 Pt A): 1037-1050, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29309922

ABSTRACT

Obesity, a major contributor to the development of cardiovascular diseases, is associated with an autonomic imbalance characterized by sympathetic hyperactivity and diminished vagal activity. Vagal activation plays important roles in weight loss and improvement of cardiac function. Pyridostigmine is a reversible acetylcholinesterase inhibitor, but whether it ameliorates cardiac lipid accumulation and cardiac remodeling in rats fed a high-fat diet has not been determined. This study investigated the effects of pyridostigmine on high-fat diet-induced cardiac dysfunction and explored the potential mechanisms. Rats were fed a normal or high-fat diet and treated with pyridostigmine. Vagal discharge was evaluated using the BL-420S system, and cardiac function by echocardiograms. Lipid deposition and cardiac remodeling were determined histologically. Lipid utility was assessed by qPCR. A high-fat diet led to a significant reduction in vagal discharge and lipid utility and a marked increase in lipid accumulation, cardiac remodeling, and cardiac dysfunction. Pyridostigmine improved vagal activity and lipid metabolism disorder and cardiac remodeling, accompanied by an improvement of cardiac function in high-fat diet-fed rats. An increase in the browning of white adipose tissue in pyridostigmine-treated rats was also observed and linked to the expression of UCP-1 and CIDEA. Additionally, pyridostigmine facilitated activation of brown adipose tissue via activation of the SIRT-1/AMPK/PGC-1α pathway. In conclusion, a high-fat diet resulted in cardiac lipid accumulation, cardiac remodeling, and a significant decrease in vagal discharge. Pyridostigmine ameliorated cardiomyopathy, an effect related to reduced cardiac lipid accumulation, and facilitated the browning of white adipose tissue while activating brown adipose tissue.


Subject(s)
Adipose Tissue/metabolism , Cardiomyopathies/prevention & control , Dietary Fats/adverse effects , Pyridostigmine Bromide/pharmacology , Vagus Nerve/physiopathology , Adipose Tissue/pathology , Adipose Tissue/physiopathology , Animals , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Cardiomyopathies/physiopathology , Dietary Fats/pharmacology , Lipid Metabolism/drug effects , Male , Muscle Proteins/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Vagus Nerve/metabolism , Vagus Nerve/pathology
10.
Sheng Li Xue Bao ; 69(5): 579-586, 2017 Oct 25.
Article in Chinese | MEDLINE | ID: mdl-29063106

ABSTRACT

Ischemic heart disease (IHD) is the life-threatening cardiovascular disease. Mitochondria have emerged as key participants and regulators of cellular energy demands and signal transduction. Mitochondrial quality is controlled by a number of coordinated mechanisms including mitochondrial fission, fusion and mitophagy, which plays an important role in maintaining healthy mitochondria and cardiac function. Recently, dysfunction of each process in mitochondrial quality control has been observed in the ischemic hearts. This review describes the mechanism of mitochondrial dynamics and mitophagy as well as its performance linked to myocardial ischemia. Moreover, in combination with our study, we will discuss the effect of vagal nerve on mitochondria in cardio-protection.


Subject(s)
Mitochondria/physiology , Myocardial Ischemia/physiopathology , Vagus Nerve/physiology , Animals , Mitochondrial Dynamics , Mitophagy , Signal Transduction
11.
Clin Exp Pharmacol Physiol ; 44(12): 1192-1200, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28732106

ABSTRACT

It is well-accepted that inflammation plays an important role in the development of cardiac remodelling and that therapeutic approaches targeting inflammation can inhibit cardiac remodelling. Although a large amount of evidence indicates that activation of α7 nicotinic acetylcholine receptor (α7nAChR) causes an anti-inflammatory effect, the role of α7nAChR in cardiac remodelling and the underlying mechanism have not been established. To investigate the effect of the specific α7nAChR agonist, PNU282987, on cardiac remodelling induced by isoproterenol (ISO 60 mg/kg per day) in mice, the cardiomyocyte cross-sectional area (CSA) and collagen volume fraction were evaluated by hematoxylin and eosin (HE) and Masson staining, respectively. Cardiac function and ventricular wall thickness were measured by echocardiography. The protein expressions of collagen I, matrix metalloproteinase 9 (MMP-9), transforming growth factor ß1 (TGF-ß1), and Smad3 were analyzed by Western blot. ISO-induced cardiac hypertrophy, characterized by an increase in the heart weight/body weight ratio, CSA and ventricular wall thickness. Moreover, cardiac fibrosis indices, such as collagen volume fraction, MMP-9 and collagen I protein expression, were also increased by ISO. PNU282987 not only attenuated cardiac hypertrophy but also decreased the cardiac fibrosis induced by ISO. Furthermore, PNU282987 suppressed TGF-ß1 protein expression and the phosphorylation of Smad3 induced by ISO. In conclusion, PNU282987 ameliorated the cardiac remodelling induced by ISO, which may be related to the TGF-ß1/Smad3 pathway. These data imply that the α7nAChR may represent a novel therapeutic target for cardiac remodelling in many cardiovascular diseases.


Subject(s)
Benzamides/therapeutic use , Bridged Bicyclo Compounds/therapeutic use , Cardiomegaly/drug therapy , Nicotinic Agonists/therapeutic use , Smad3 Protein/metabolism , Transforming Growth Factor beta1/metabolism , Ventricular Remodeling/drug effects , alpha7 Nicotinic Acetylcholine Receptor/agonists , Animals , Benzamides/administration & dosage , Bridged Bicyclo Compounds/administration & dosage , Cardiomegaly/metabolism , Cardiomegaly/pathology , Isoproterenol/pharmacology , Male , Mice, Inbred BALB C , Myocardium/metabolism , Myocardium/pathology , Nicotinic Agonists/administration & dosage , Signal Transduction
12.
J Mol Cell Cardiol ; 107: 1-12, 2017 06.
Article in English | MEDLINE | ID: mdl-28395930

ABSTRACT

The endoplasmic reticulum (ER) forms discrete junctions with the plasma membrane (PM) that play a critical role in the regulation of Ca2+ signaling during cellular bioenergetics, apoptosis and autophagy. We have previously confirmed that acetylcholine can inhibit ER stress and apoptosis after inflammatory injury. However, limited research has focused on the effects of acetylcholine on ER-PM junctions. In this work, we evaluated the structure and function of the supramolecular sodium-calcium exchanger 1 (NCX1)-transient receptor potential canonical 3 (TRPC3)-inositol 1,4,5-trisphosphate receptor 1 (IP3R1) complex, which is involved in regulating Ca2+ homeostasis during inflammatory injury. The width of the ER-PM junctions of human umbilical vein endothelial cells (HUVECs) was measured in nanometres using transmission electron microscopy and a fluorescent probe for Ca2+. Protein-protein interactions were assessed by immunoprecipitation. Ca2+ concentration was measured using a confocal microscope. An siRNA assay was employed to silence specific proteins. Our results demonstrated that the peripheral ER was translocated to PM junction sites when induced by tumour necrosis factor-alpha (TNF-α) and that NCX1-TRPC3-IP3R1 complexes formed at these sites. After down-regulating the protein expression of NCX1 or IP3R1, we found that the NCX1-mediated inflow of Ca2+ and the release of intracellular Ca2+ stores were reduced in TNF-α-treated cells. We also observed that acetylcholine attenuated the formation of NCX1-TRPC3-IP3R1 complexes and maintained calcium homeostasis in cells treated with TNF-α. Interestingly, the positive effects of acetylcholine were abolished by the selective M3AChR antagonist darifenacin and by AMPK siRNAs. These results indicate that acetylcholine protects endothelial cells from TNF-alpha-induced injury, [Ca2+]cyt overload and ER-PM interactions, which depend on the muscarinic 3 receptor/AMPK pathway, and that acetylcholine may be a new inhibitor for suppressing [Ca2+]cyt overload.


Subject(s)
Inflammation/genetics , Inositol 1,4,5-Trisphosphate Receptors/genetics , Sodium-Calcium Exchanger/genetics , TRPC Cation Channels/genetics , Tumor Necrosis Factor-alpha/metabolism , Acetylcholine/metabolism , Apoptosis/genetics , Calcium/metabolism , Calcium Signaling/genetics , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress , Homeostasis/genetics , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Inflammation/metabolism , Inflammation/pathology , Inositol 1,4,5-Trisphosphate Receptors/chemistry , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , RNA, Small Interfering/genetics , Sodium-Calcium Exchanger/chemistry , TRPC Cation Channels/chemistry
13.
J Cell Mol Med ; 21(9): 2106-2116, 2017 09.
Article in English | MEDLINE | ID: mdl-28296184

ABSTRACT

Cardiac hypertrophy is associated with autonomic imbalance, characterized by enhanced sympathetic activity and withdrawal of parasympathetic control. Increased parasympathetic function improves ventricular performance. However, whether pyridostigmine, a reversible acetylcholinesterase inhibitor, can offset cardiac hypertrophy induced by pressure overload remains unclear. Hence, this study aimed to determine whether pyridostigmine can ameliorate pressure overload-induced cardiac hypertrophy and identify the underlying mechanisms. Rats were subjected to either sham or constriction of abdominal aorta surgery and treated with or without pyridostigmine for 8 weeks. Vagal activity and cardiac function were determined using PowerLab. Cardiac hypertrophy was evaluated using various histological stains. Protein markers for cardiac hypertrophy were quantitated by Western blot and immunoprecipitation. Pressure overload resulted in a marked reduction in vagal discharge and a profound increase in cardiac hypertrophy index and cardiac dysfunction. Pyridostigmine increased the acetylcholine levels by inhibiting acetylcholinesterase in rats with pressure overload. Pyridostigmine significantly attenuated cardiac hypertrophy based on reduction in left ventricular weight/body weight, suppression of the levels of atrial natriuretic peptide, brain natriuretic peptide and ß-myosin heavy chain, and a reduction in cardiac fibrosis. These effects were accompanied by marked improvement of cardiac function. Additionally, pyridostigmine inhibited the CaN/NFAT3/GATA4 pathway and suppressed Orai1/STIM1 complex formation. In conclusion, pressure overload resulted in cardiac hypertrophy, cardiac dysfunction and a significant reduction in vagal discharge. Pyridostigmine attenuated cardiac hypertrophy and improved cardiac function, which was related to improved cholinergic transmission efficiency (decreased acetylcholinesterase and increased acetylcholine), inhibition of the CaN/NFAT3/GATA4 pathway and suppression of the interaction of Orai1/STIM1.


Subject(s)
Calcineurin/metabolism , Cardiomegaly/drug therapy , Cardiomegaly/metabolism , Pressure , Pyridostigmine Bromide/administration & dosage , Pyridostigmine Bromide/therapeutic use , Signal Transduction , Animals , Cardiomegaly/diagnostic imaging , Cardiomegaly/physiopathology , GATA4 Transcription Factor/metabolism , Heart Function Tests , Hemodynamics/drug effects , Male , NFATC Transcription Factors/metabolism , ORAI1 Protein/metabolism , Protein Binding/drug effects , Rats, Sprague-Dawley , Stromal Interaction Molecule 1/metabolism , Time Factors , Vagus Nerve/drug effects , Vagus Nerve/pathology
14.
J Cell Mol Med ; 21(1): 58-71, 2017 01.
Article in English | MEDLINE | ID: mdl-27491814

ABSTRACT

Mitochondrial dynamics-fission and fusion-are associated with ischaemic heart disease (IHD). This study explored the protective effect of vagal nerve stimulation (VNS) against isoproterenol (ISO)-induced myocardial ischaemia in a rat model and tested whether VNS plays a role in preventing disorders of mitochondrial dynamics and function. Isoproterenol not only caused cardiac injury but also increased the expression of mitochondrial fission proteins [dynamin-related peptide1 (Drp1) and mitochondrial fission protein1 (Fis-1)) and decreased the expression of fusion proteins (optic atrophy-1 (OPA1) and mitofusins1/2 (Mfn1/2)], thereby disrupting mitochondrial dynamics and leading to increase in mitochondrial fragments. Interestingly, VNS restored mitochondrial dynamics through regulation of Drp1, Fis-1, OPA1 and Mfn1/2; enhanced ATP content and mitochondrial membrane potential; reduced mitochondrial permeability transition pore (MPTP) opening; and improved mitochondrial ultrastructure and size. Furthermore, VNS reduced the size of the myocardial infarction and ameliorated cardiomyocyte apoptosis and cardiac dysfunction induced by ISO. Moreover, VNS activated AMP-activated protein kinase (AMPK), which was accompanied by phosphorylation of Ca2+ /calmodulin-dependent protein kinase kinase ß (CaMKKß) during myocardial ischaemia. Treatment with subtype-3 of muscarinic acetylcholine receptor (M3 R) antagonist 4-diphenylacetoxy-N-methylpiperidine methiodide or AMPK inhibitor Compound C abolished the protective effects of VNS on mitochondrial dynamics and function, suggesting that M3 R/CaMKKß/AMPK signalling are involved in mediating beneficial effects of VNS. This study demonstrates that VNS modulates mitochondrial dynamics and improves mitochondrial function, possibly through the M3 R/CaMKKß/AMPK pathway, to attenuate ISO-induced cardiac damage in rats. Targeting mitochondrial dynamics may provide a novel therapeutic strategy in IHD.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Kinase/metabolism , Isoproterenol/pharmacology , Mitochondrial Dynamics/physiology , Myocardial Ischemia/chemically induced , Myocardial Ischemia/metabolism , Receptor, Muscarinic M3/metabolism , Animals , Apoptosis/physiology , Male , Membrane Potential, Mitochondrial/physiology , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Mitochondrial Dynamics/drug effects , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Myocardial Infarction/chemically induced , Myocardial Infarction/metabolism , Myocardial Reperfusion Injury/chemically induced , Myocardial Reperfusion Injury/metabolism , Phosphorylation/physiology , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology , Vagus Nerve Stimulation/methods
15.
Sheng Li Xue Bao ; 68(4): 517-24, 2016 Aug 25.
Article in Chinese | MEDLINE | ID: mdl-27546511

ABSTRACT

Calcium overload is one of the important mechanisms of cardiovascular disease. Endoplasmic reticulum is an important organelle which regulates intracellular calcium homeostasis by uptake, storage and mobilization of calcium. So it plays a critical role in regulation of intracellular calcium homeostasis. Endoplasmic reticulum, which is widely distributed in cytoplasm, has a large number of membrane junction sites. Recent studies have reported that these junction sites are distributed on plasma membrane and organelle membranes (mitochondria, lysosomes, Golgi apparatus, etc.), separately. They could form complexes to regulate calcium transport. In this review, we briefly outlined the recent research progresses of endoplasmic reticulum-plasma membrane junctions in intracellular calcium homeostasis and cardiovascular disease, which may offer a new strategy for prevention and treatment of cardiovascular disease.


Subject(s)
Cardiovascular Diseases , Cell Membrane , Endoplasmic Reticulum , Homeostasis , Calcium , Humans
16.
J Cell Physiol ; 231(5): 1171-81, 2016 May.
Article in English | MEDLINE | ID: mdl-26465230

ABSTRACT

Acetylcholine (ACh) protected against cardiac injury via promoting autophagy and mitochondrial biogenesis, however, the involvement of mitophagy in ACh-elicited cardioprotection remains unknown. In the present study, H9c2 cardiomyocytes were subjected to hypoxia/reoxygenation (H/R) and ACh treatment during reoxygenation. Mitophagy markers PTEN-induced kinase 1 (PINK1) and Parkin translocation were examined using western blot and confocal fluorescence microscopy. Mitochondrial membrane potential and reactive oxygen species (ROS) were detected with fluorescence staining. We found that H/R-treated cells exhibited reduced levels of PINK1 and Parkin in mitochondria, accompanied with decreased autophagy flux (reduced LC3-II/LC3-I and increased p62). Conversely, ACh increased PINK1 and Parkin translocation to mitochondria and enhanced autophagy proteins. Confocal imaging of Parkin and MitoTracker Green-labeled mitochondria further confirmed ACh-induced mitochondrial translocation of Parkin, which was reversed by M2 receptor antagonist methoctramine and M2 receptor siRNA, suggesting ACh could induce mitophagy by M2 receptor after H/R. Mitophagy inhibitor 3-methaladenine abolished ACh-induced mitoprotection, manifesting as aggravated mitochondrial morphology disruption, ATP and membrane potential depletion, increased ROS overproduction, and apoptosis. Furthermore, PINK1/Parkin siRNA attenuated the protective effects of ACh against ATP loss and oxidative stress due to mitochondrial-dependent injury. Taken together, ACh promoted mitochondrial translocation of PINK1/Parkin to stimulate cytoprotective mitophagy via M2 receptor, which may provide beneficial targets in the preservation of cardiac homeostasis against H/R injury.


Subject(s)
Acetylcholine/pharmacology , Mitophagy/drug effects , Oxygen/pharmacology , Protein Kinases/metabolism , Signal Transduction/drug effects , Ubiquitin-Protein Ligases/metabolism , Animals , Cell Hypoxia/drug effects , Cell Line , Cytoprotection/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , RNA, Small Interfering/metabolism , Rats , Receptor, Muscarinic M2/metabolism
17.
Sheng Li Ke Xue Jin Zhan ; 47(4): 255-9, 2016 Aug.
Article in Chinese | MEDLINE | ID: mdl-29888895

ABSTRACT

Hypertension, which can cause a variety of cardiovascular and cerebrovascular complications, is a serious threat to human health. Currently, it is found that hypertension is related to immunoregulatory abnormality, which could lead to chronic inflammation. Then the chronic inflammation may impair vascular endothelial function and activate renin-angiotensin system, which cause vascular remodeling, angiosclerosis, dysfunctional vasoconstriction and vasodilatation, and exacerbate hypertension. The immunoregulatory abnormality of hypertension involves macrophage infiltration of the organization, the dendritic cell of antigen presentation and natural killer cells of activation in nonspecific immunity and activation of T cells in specific immune. The key of immunity mechanism of hypertension is the Toll like receptor to activate immune system and lead to inflammation. Autonomic nervous system is also closely related to the development and progression of hypertension. Autonomic imbalance in hypertension leads to immunoregulatory abnormality, cardiovascular injury, and dysfunctional vasoconstriction and vasodilatation, which ultimately results in exacerbation of hypertension. Therefore, research on neuro-immune regulation will help to provide novel strategies for therapy of hypertension. In this review, we will provide an overview of the research progress of the immunity mechanism of hypertension and the regulation of immune system by the autonomic nervous system.


Subject(s)
Hypertension/etiology , Renin-Angiotensin System , Animals , Cardiovascular Diseases , Humans , Hypertension/physiopathology , Immunity, Innate , Inflammation , T-Lymphocytes , Toll-Like Receptors/physiology , Vascular Remodeling , Vasoconstriction
18.
Sci Rep ; 5: 17108, 2015 Nov 24.
Article in English | MEDLINE | ID: mdl-26596640

ABSTRACT

Cardiac remodeling is characterized by overactivity of the renin-angiotensin system (RAS) and withdrawal of vagal activity. We hypothesized that improving vagal activity could attenuate cardiac fibrosis induced by angiotensin II (Ang II) in vivo and in vitro. Rats were subjected to abdominal aorta constriction (AAC) with or without pyridostigmine (PYR) (31 mg/kg/d). After 8 weeks, PYR significantly decreased Ang II level, AT1 protein expression, and collagen deposition in cardiac tissue and improved heart rate variability, baroreflex sensitivity and cardiac function, which were abolished by atropine. In vitro, treatment of cardiac fibroblasts (CFs) with Ang II (10(-7) M) increased cell proliferation, migration, transformation, and secretory properties, which were significantly diminished by acetylcholine (ACh, 10(-6) M). Subsequently, Ang II significantly increased collagen type I expression as well as metalloproteinase (MMP)-2 expression and activity. Transforming growth factor (TGF)-ß1 expression and Smad3 phosphorylation presented a similar trend. Notably, the knockdown of the acetylcholine M2 receptor by siRNA could abolish ACh anti-fibrotic action. These data implicated cholinesterase inhibitor can increase vagal activity and reduce local Ang II level, and ACh inhibit Ang II pro-fibrotic effects. Our findings suggested that the parasympathetic nervous system can serve as a promising target for cardiac remodeling treatment.


Subject(s)
Cholinesterase Inhibitors/pharmacology , Heart Diseases/drug therapy , Pyridostigmine Bromide/pharmacology , Vagus Nerve/physiopathology , Angiotensin II/metabolism , Animals , Cell Movement , Cell Proliferation , Cell Survival , Cells, Cultured , Cholinesterase Inhibitors/therapeutic use , Collagen/biosynthesis , Constriction, Pathologic/drug therapy , Drug Evaluation, Preclinical , Fibroblasts/physiology , Fibrosis , Gene Knockdown Techniques , Heart Diseases/physiopathology , Male , Pyridostigmine Bromide/therapeutic use , Rats, Sprague-Dawley , Receptor, Muscarinic M2/genetics , Receptor, Muscarinic M2/metabolism , Vagus Nerve/drug effects
19.
Cell Physiol Biochem ; 36(5): 2025-38, 2015.
Article in English | MEDLINE | ID: mdl-26202362

ABSTRACT

BACKGROUND: Excessive activation of matrix metalloproteinase 9 (MMP-9) has been found in several inflammatory diseases. Previous studies have shown that acetylcholine (ACh) reduced the levels of pro-inflammatory cytokines and decreased tissue damage. Therefore, this study was designed to explore the potential effects and mechanisms of ACh on MMP-9 production and cell migration in response to lipopolysaccharide (LPS) stimulation in RAW264.7 cells. METHODS: MMP-9 expression and activity were induced by LPS in RAW264.7 cells, and examined by real-time PCR, western blotting and gelatin zymography, respectively. ELISA was used to determine the changes in MMP-9 secretion among the groups. Macrophage migration was evaluated using transwell migration assay. Knockdown of α7 nicotinic acetylcholine receptor (α7 nAChR) expression was performed using siRNA transfection. RESULTS: Pre-treatment with ACh inhibited LPS-induced MMP-9 production and macrophage migration in RAW264.7 cells. These effects were abolished by the α7 nAChR antagonist methyllycaconitine (MLA) and α7 nAChR siRNA. The α7 nAChR agonist PNU282987 was found to have an effect similar to that of ACh. Moreover, ACh enhanced the expression of JAK2 and STAT3, and the JAK2 inhibitor AG490 and the STAT3 inhibitor static restored the effect of ACh. Meanwhile, ACh decreased the phosphorylation and nuclear translocation of NF-κB, and this effect was abrogated in the presence of MLA. In addition, the JAK2 and STAT3 inhibitor abolished the inhibitory effects of ACh on phosphorylation of NF-κB. CONCLUSIONS: Activation of α7 nAChR by ACh inhibited LPS-induced MMP-9 production and macrophage migration through the JAK2/STAT3 signaling pathway. These results provide novel insights into the anti-inflammatory effects and mechanisms of ACh.


Subject(s)
Acetylcholine/pharmacology , Cell Movement/drug effects , Janus Kinase 2/metabolism , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Matrix Metalloproteinase 9/biosynthesis , STAT3 Transcription Factor/metabolism , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Animals , Cell Line , Macrophages/enzymology , Macrophages/metabolism , Mice
20.
Br J Pharmacol ; 172(23): 5619-33, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25953628

ABSTRACT

BACKGROUND AND PURPOSE: The activation of M3 cholinoceptors (M3 receptors) by choline reduces cardiovascular risk, but it is unclear whether these receptors can regulate ischaemia/reperfusion (I/R)-induced vascular injury. Thus, the primary goal of the present study was to explore the effects of choline on the function of mesenteric arteries following I/R, with a major focus on Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) regulation. EXPERIMENTAL APPROACH: Rats were given choline (10 mg · kg(-1), i.v.) and then the superior mesenteric artery was occluded for 60 min (ischaemia), followed by 90 min of reperfusion. The M3 receptor antagonist, 4-diphenylacetoxy-N-methylpiperidine methiodide (4-DAMP), was injected (0.12 µg · kg(-1), i.v.) 5 min prior to choline treatment. Vascular function was examined in rings of mesenteric arteries isolated after the reperfusion procedure. Vascular superoxide anion production, CaMKII and the levels of Ca(2+)-cycling proteins were also assessed. KEY RESULTS: Choline treatment attenuated I/R-induced vascular dysfunction, blocked elevations in the levels of reactive oxygen species (ROS) and decreased the up-regulated expression of oxidised CaMKII and phosphorylated CaMKII. In addition, choline reversed the abnormal expression of Ca(2+)-cycling proteins, including Na(+)Ca(2+) exchanger, inositol 1,4,5-trisphosphate receptor, sarcoplasmic reticulum Ca(2+)-ATPase and phospholamban. All of these cholinergic effects of choline were abolished by 4-DAMP. CONCLUSIONS AND IMPLICATIONS: Our data suggest that inhibition of the ROS-mediated CaMKII pathway and modulation of Ca(2+)-cycling proteins may be novel mechanisms underlying choline-induced vascular protection. These results represent a significant addition to the understanding of the pharmacological roles of M3 receptors in the vasculature, providing a new therapeutic strategy for I/R-induced vascular injury.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Cholinergic Agonists/pharmacology , Ischemia/complications , Receptor, Muscarinic M3/metabolism , Reperfusion Injury/complications , Vascular System Injuries/drug therapy , Administration, Intravenous , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Choline/administration & dosage , Choline/pharmacology , Choline/therapeutic use , Cholinergic Agonists/administration & dosage , Cholinergic Agonists/therapeutic use , Dose-Response Relationship, Drug , Male , Piperidines/administration & dosage , Piperidines/pharmacology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Structure-Activity Relationship , Vascular System Injuries/chemically induced , Vascular System Injuries/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...