Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
J Biochem Mol Toxicol ; 38(3): e23671, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38454809

ABSTRACT

Obesity is a major cause of nonalcohol fatty liver disease (NAFLD), which is characterized by hepatic fibrosis, lipotoxicity, inflammation, and apoptosis. Previous studies have shown that an imbalance in the autonomic nervous system is closely related to the pathogenesis of NAFLD. In this study, we investigated the effects of pyridostigmine (PYR), a cholinesterase (AChE) inhibitor, on HFD-induced liver injury and explored the potential mechanisms involving mitochondrial damage and oxidative stress. A murine model of HFD-induced obesity was established using the C57BL/6 mice, and PYR (3 mg/kg/d) or placebo was administered for 20 weeks. PYR reduced the body weight and liver weight of the HFD-fed mice. Additionally, the serum levels of IL-6, TNF-α, cholesterol, and triglyceride were significantly lower in the PYR-treated versus the untreated mice, corresponding to a decrease in hepatic fibrosis, lipid accumulation, and apoptosis in the former. Furthermore, the mitochondrial morphology improved significantly in the PYR-treated group. Consistently, PYR upregulated ATP production and the mRNA level of the mitochondrial dynamic factors OPA1, Drp1 and Fis1, and the mitochondrial unfolded protein response (UPRmt) factors LONP1 and HSP60. Moreover, PYR treatment activated the Keap1/Nrf2 pathway and upregulated HO-1 and NQO-1, which mitigated oxidative injury as indicated by decreased 8-OHDG, MDA and H2 O2 levels, and increased SOD activity. Finally, PYR elevated acetylcholine (ACh) levels by inhibiting AChE, and upregulated the α7nAChR and M3AChR proteins in the HFD-fed mice. PYR alleviated obesity-induced hepatic injury in mice by mitigating mitochondrial damage and oxidative stress via α7nAChR and M3AChR.


Subject(s)
Chemical and Drug Induced Liver Injury, Chronic , Non-alcoholic Fatty Liver Disease , Mice , Animals , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Pyridostigmine Bromide/pharmacology , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , Chemical and Drug Induced Liver Injury, Chronic/complications , Chemical and Drug Induced Liver Injury, Chronic/metabolism , Mice, Inbred C57BL , NF-E2-Related Factor 2/metabolism , Liver/metabolism , Oxidative Stress , Liver Cirrhosis/metabolism , Obesity/drug therapy , Obesity/metabolism , Diet , Diet, High-Fat/adverse effects
2.
Sheng Li Xue Bao ; 71(2): 216-224, 2019 Apr 25.
Article in Zh | 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
3.
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
4.
Sheng Li Xue Bao ; 69(5): 579-586, 2017 Oct 25.
Article in Zh | 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
5.
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
6.
Phytomedicine ; 126: 155441, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38394729

ABSTRACT

BACKGROUND: The mitochondrial unfolded protein response (UPRmt) is the first line of defense against mitochondrial dysfunction in several diseases. Baicalein, which is an extract of Scutellaria baicalensis Georgi roots, exerts mitoprotective effects on metabolic disorders and cardiovascular diseases. However, it remains unclear whether baicalein alleviates obesity-induced cardiac damage through the UPRmt. PURPOSE: The present research designed to clarify the role of baicalein in lipotoxicity-induced myocardial apoptosis and investigated the UPRmt-related mechanism. METHODS: In the in vitro experiment, palmitic acid (PA)-treated AC16 cardiomyocytes were established to mimic obesity-induced myocardial injury. After pretreatment of AC16 cells with baicalein, the levels of cell vitality, apoptosis, mitochondrial membrane potential, mitochondrial oxidative stress, and UPRmt-related proteins were determined. Additionally, AC16 cells were treated with ML385 or siRNA to explore the regulation of the UPRmt by NRF2 signaling. In the in vivo experiment, male db/db mice administered with baicalein for 8 weeks were used to validate the effects of baicalein on cardiac damage induced by obesity, the UPRmt, and the NRF2-related pathway. RESULTS: In AC16 cardiomyocytes, PA dose-dependently increased the expression of UPRmt markers (HSP60, LONP1, ATF4, and ATF5). This increase was accompanied by enhanced production of mitochondrial ROS, reduced mitochondrial membrane potential, and elevated the expression levels of cytochrome c, cleaved caspase-3, and Bax/Bcl2, eventually leading to cell apoptosis. Baicalein treatment reversed UPRmt activation and mitochondrial damage and impeded mitochondrial-mediated cell apoptosis. Moreover, NRF2 downregulation by its inhibitor ML385 or siRNA diminished baicalein-mediated NRF2 signaling activation and UPRmt inhibition and triggered mitochondrial dysfunction. Additionally, NRF2 deficiency more intensely activated the UPRmt, resulting in mitochondrial oxidative stress and apoptosis of PA-induced cardiomyocytes, thus indicating that NRF2 plays a vital role in mitochondrial homeostasis regulation. In the in vivo study in db/db mice, baicalein inhibited the UPRmt, enhanced the antioxidant capacity, and attenuated cardiac dysfunction through a NRF2-activated pathway. CONCLUSION: To our best knowledge, these results provide the first insight that baicalein inhibits the UPRmt to induce a protective effect against lipotoxicity-induced mitochondrial damage and cardiomyocyte apoptosis via activating NRF2 signaling and suggest a new role of NRF2 in UPRmt regulation.


Subject(s)
Flavanones , Heart Diseases , Mitochondrial Diseases , Mice , Animals , Male , NF-E2-Related Factor 2/metabolism , Unfolded Protein Response , Apoptosis , RNA, Small Interfering/pharmacology , Mitochondrial Diseases/metabolism , Oxidative Stress , Myocytes, Cardiac
7.
Cell Cycle ; 20(18): 1890-1906, 2021 09.
Article in English | MEDLINE | ID: mdl-34424820

ABSTRACT

Lipid droplets (LDs), which are neutral lipid storage organelles, are important for lipid metabolism and energy homeostasis. LD lipolysis and interactions with mitochondria are tightly coupled to cellular metabolism and may be potential targets to buffer the effects of excessive toxic lipid species levels. Acetylcholine (ACh), the major neurotransmitter of the vagus nerve, exhibits cardioprotective effects. However, limited research has focused on its effects on LD lipolysis and the LD-mitochondria association in fatty acid (FA) overload models. Here, we reveal that palmitate (PA) induces an increase in expression of the FA transport protein cluster of differentiation 36 (CD36) and LD formation; remarkably reduces the expression of lipases involved in triacylglycerol (TAG) lipolysis, such as adipose triglyceride lipase (ATGL), hormone-sensitive lipase (HSL) and monoacylglycerol lipase (MGL); impairs LD-mitochondria interaction; and decreases perilipin 5 (PLIN5) expression, resulting in LD accumulation and mitochondrial dysfunction, which ultimately lead to cardiomyocyte apoptosis. ACh significantly upregulates PLIN5 expression and improved LD lipolysis and the LD-mitochondria association. Moreover, ACh reduces CD36 expression, LD deposition and mitochondrial dysfunction, ultimately suppressing apoptosis in PA-treated neonatal rat ventricular cardiomyocytes (NRVCs). Knockdown of PLIN5, which plays a role in LD-mitochondria contact site formation, abolishes the protective effects of ACh in PA-treated NRVCs. Thus, ACh protects cardiomyocytes from PA-induced apoptosis, at least partly, by promoting LD lipolysis and activating LD-mitochondria interactions via PLIN5. These findings may aid in developing novel therapeutic approaches that target LD lipolysis and PLIN5-mediated LD-mitochondria interactions to prevent or alleviate lipotoxic cardiomyopathy.


Subject(s)
Acetylcholine/pharmacology , Apoptosis/drug effects , Lipid Droplets/metabolism , Lipolysis/drug effects , Mitochondria/metabolism , Myocytes, Cardiac/metabolism , Palmitates/adverse effects , Perilipin-5/metabolism , Signal Transduction/drug effects , Animals , Animals, Newborn , CD36 Antigens/metabolism , Cells, Cultured , Lipid Droplets/drug effects , Myocytes, Cardiac/drug effects , Rats , Rats, Sprague-Dawley , Triglycerides/metabolism
8.
Front Pharmacol ; 12: 739960, 2021.
Article in English | MEDLINE | ID: mdl-35095482

ABSTRACT

Background: Carbapenems are considered the last line of defence against bacterial infections, but their high consumption and the resulting antibacterial resistance are an increasing global concern. In this context, the Chinese health authority issued an expert consensus on the clinical applications of carbapenems. However, the long- and short-term effects of the expert consensus on carbapenem use are not clear. Methods: This study was conducted in Shaanxi, a northwest province of China. We collected all available carbapenem procurement data between January 2017 and December 2020 from the Provincial Drug Centralized Bidding Procurement System. A quasi-experimental interrupted time series analysis was used to evaluate the longitudinal effectiveness of expert consensus by measuring the change in the Defined Daily Dosesper 1,000 inhabitants per day (DID), the percentage of carbapenem expenditures to total antimicrobial expenditure, the total carbapenem expenditure, and the defined daily cost (DDDc). We used Stata SE version 15.0 for data analysis, and p < 0.05 was considered statistically significant. Results: After the distribution of the expert consensus, the level (p = 0.769) and trend (p = 0.184) of DID decreased, but the differences were not statistically significant. The percentage of carbapenem expenditures to total antimicrobial expenditure decreased abruptly (p < 0.001) after the intervention, but the long-term trend was still upward. There was no statistically significant relationship between the release of the expert consensus and carbapenem expenditure in the long term, but there was a decreasing trend (p = 0.032). However, the expert consensus had a positive impact on the economic burden of carbapenem usage in patients, as the level (p < 0.001), and trend (p = 0.003) of DDDc significantly decreased. Conclusion: The long-term effects of the distribution of the expert consensus on the use and expenditure of carbapenems in public health institutions in Shaanxi Province were not optimal. It is time to set up more administrative measures and scientific supervision to establish a specific index to limit the application of carbapenems.

9.
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
10.
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
11.
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
12.
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
13.
Sci Rep ; 7: 42553, 2017 02 22.
Article in English | MEDLINE | ID: mdl-28225018

ABSTRACT

Autonomic dysfunction and abnormal immunity lead to systemic inflammatory responses, which result in cardiovascular damage in hypertension. The aim of this report was to investigate the effects of choline on cardiovascular damage in hypertension. Eight-week-old male spontaneously hypertensive rats (SHRs) and Wistar-Kyoto rats were intraperitoneally injected with choline or vehicle (8 mg/kg/day). After 8 weeks, choline restored the cardiac function of the SHRs, as evidenced by decreased heart rate, systolic blood pressure, left ventricle systolic pressure, and ±dp/dtmax and increased ejection fraction and fractional shortening. Choline also ameliorated the cardiac hypertrophy of the SHRs, as indicated by reduced left ventricle internal dimensions and decreased cardiomyocyte cross-sectional area. Moreover, choline improved mesenteric arterial function and preserved endothelial ultrastructure in the SHRs. Notably, the protective effect of choline may be due to its anti-inflammatory effect. Choline downregulated expression of interleukin (IL)-6 and tumour necrosis factor-α and upregulated IL-10 in the mesenteric arteries of SHRs, possibly because of the inhibition of Toll-like receptor 4. Furthermore, choline restored baroreflex sensitivity and serum acetylcholine level in SHRs, thus indicating that choline improved vagal activity. This study suggests that choline elicits cardiovascular protective effects and may be useful as a potential adjunct therapeutic approach for hypertension.


Subject(s)
Cardiovascular Diseases/etiology , Cardiovascular Diseases/physiopathology , Choline/pharmacology , Vagus Nerve/drug effects , Animals , Baroreflex/drug effects , Cardiomegaly , Cardiovascular Diseases/diagnosis , Cardiovascular Diseases/drug therapy , Choline/blood , Cytokines/metabolism , Disease Models, Animal , Echocardiography , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Heart Function Tests , Hypertension/etiology , Hypertension/physiopathology , Inflammation Mediators/metabolism , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Rats , Rats, Inbred SHR , Toll-Like Receptor 4/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL