1
|
Abdullah AR, Seliem MA, Khidr EG, Sobhy AM, El-Shiekh RA, Hafeez MSAE, El-Husseiny AA. A comprehensive review on diabetic cardiomyopathy (DCM): histological spectrum, diagnosis, pathogenesis, and management with conventional treatments and natural compounds. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03980-9. [PMID: 40100371 DOI: 10.1007/s00210-025-03980-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 02/22/2025] [Indexed: 03/20/2025]
Abstract
Diabetic complications are among the most pressing health issues currently. Cardiovascular problems, particularly diabetic cardiomyopathy (DCM), are responsible for almost 80% of diabetic deaths. Because of the increasing prevalence of diabetes and the increased threat of death from its consequences, researchers are searching for new pharmaceutical targets to delay or cure it. Currently, there are a few medicines available for the treatment of DCM, some of which have serious side effects. To address this issue, researchers are focusing on natural products. Thus, in this review, we discuss the prevalence, incidence, risk factors, histological spectrum, diagnosis, pathogenic pathways of DCM, genetic and epigenetic mechanisms involved in DCM, the current treatments, and the beneficial effects of natural product-based therapeutics. Natural treatments range from single doses to continuous regimens lasting weeks or months. Flavonoids are the largest class of natural compounds reported for the treatment of DCM. Natural regimens may cover the way for new treatment strategies for DCM for being multi-target agents in the treatment of DCM, with the ability to play a variety of functions via distinct signaling pathways.
Collapse
Affiliation(s)
- Ahmed R Abdullah
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Al-Azhar University, Cairo, 11231, Egypt
| | - Mahmoud A Seliem
- Department of Biochemistry, Faculty of Pharmacy, Ahram Canadian University, 6Th of October City, Giza, Egypt
| | - Emad Gamil Khidr
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Al-Azhar University, Cairo, 11231, Egypt
| | - Ayah M Sobhy
- Pharmacognosy Department, Faculty of Pharmacy, Badr University in Assiut, Assiut, Egypt
| | - Riham A El-Shiekh
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo, 11562, Egypt.
| | - Mohamed S Abd El Hafeez
- Department of Pharmacy, Kut University College, Al Kut, Wasit, 52001, Iraq
- Department of Pharmacognosy, Faculty of Pharmacy, Egyptian Russian University, Cairo-Suez Road, Badr City, 11829, Egypt
| | - Ahmed A El-Husseiny
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Al-Azhar University, Cairo, 11231, Egypt
- Department of Biochemistry, Faculty of Pharmacy, Egyptian Russian University, Badr City, Cairo, 11829, Egypt
| |
Collapse
|
2
|
Helal SA, Gerges SH, Panahi S, Dyck JRB, El-Kadi AOS. Investigating the sexual dimorphism in isoproterenol-induced cardiac hypertrophy in Sprague Dawley rats. Drug Metab Dispos 2025; 53:100035. [PMID: 39891968 DOI: 10.1016/j.dmd.2025.100035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 12/23/2024] [Indexed: 02/03/2025] Open
Abstract
Distinct differences between sexes exist in various cardiovascular diseases. Moreover, there is a significant correlation between the pathogenesis of cardiac hypertrophy (CH) and the metabolites of arachidonic acid (AA) mediated by cytochrome P450 (CYP) enzymes. The potential link between these sex differences, the levels and the activity of CYP enzymes, and their AA-mediated metabolites remains to be elucidated. Male and female Sprague Dawley rats were injected with 1 mg/kg isoproterenol for 7 days to induce CH. Echocardiography was performed before and after the induction of CH. The hypertrophic markers and CYP enzyme levels were analyzed at the gene and protein levels using real-time polymerase chain reaction and Western blot, respectively. Heart microsomal proteins were incubated with AA, and the resulting metabolites were quantified using liquid chromatography-tandem mass spectrometry. Both sexes showed a significant degree of CH, albeit to varying extents, as the echocardiograph, heart weight/tibial length, and left ventricular parameters proved. In addition, the β/α-myosin heavy chain was 2-fold higher in male compared with female rats. Albeit the 20-hydroxyeicosatetraenoic acid (20-HETE) metabolite formation showed no increase in both sexes, the mid-chain HETEs (5- and 15-HETE) were higher in male rats, which paralleled the increase in the gene and protein levels of CYP1B1. The formation rate of the epoxyeicosatrienoic acids was almost unchanged in female-treated rats, while it was significantly decreased in male-treated rats. Our results suggest sexual dimorphism in the isoproterenol-induced CH in rats, specifically on the level of CYP enzymes and their AA-mediated metabolites. SIGNIFICANCE STATEMENT: Sexual dimorphism was observed in rats following isoproterenol-induced cardiac hypertrophy, with males showing a stronger hypertrophic response. This was linked to higher CYP1B1 gene and protein expression in males, along with sex-related differences in many cytochrome P450 enzyme activities and their mediated arachidonic acid metabolites. These findings emphasized the need for targeted, sex-specific therapeutic strategies for the management and treatment of cardiac hypertrophy and other cardiovascular disorders.
Collapse
Affiliation(s)
- Sara A Helal
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada; Faculty of Pharmacy, Department of Biochemistry, Tanta University, Tanta, Egypt
| | - Samar H Gerges
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Sareh Panahi
- Faculty of Medicine & Dentistry, Pediatrics Department, University of Alberta, Edmonton, Alberta, Canada
| | - Jason R B Dyck
- Faculty of Medicine & Dentistry, Pediatrics Department, University of Alberta, Edmonton, Alberta, Canada
| | - Ayman O S El-Kadi
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
3
|
Prajapati AK, Shah G. Exploring in vivo and in vitro models for heart failure with biomarker insights: a review. Egypt Heart J 2024; 76:141. [PMID: 39432214 PMCID: PMC11493927 DOI: 10.1186/s43044-024-00568-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 09/27/2024] [Indexed: 10/22/2024] Open
Abstract
BACKGROUND Heart failure (HF) is a condition characterized by the heart's inability to meet the body's demands, resulting in various complications. Two primary types of HF exist, namely HF with preserved left ventricular ejection fraction (LVEF) and HF reduced with LVEF. The progression of HF involves compensatory mechanisms such as cardiac hypertrophy, fibrosis, and alterations in gene expression. Pressure overload and volume overload are common etiologies of HF, with pressure overload often stemming from conditions like hypertension, leading to left ventricular hypertrophy and fibrosis. In contrast, volume overload can arise from chronic valvular regurgitant disease, also inducing left ventricular hypertrophy. MAIN BODY In vitro cell culture techniques serve as vital tools in studying HF pathophysiology, allowing researchers to investigate cellular responses and potential therapeutic targets. Additionally, biomarkers, measurable biological characteristics, play a crucial role in diagnosing and predicting HF. Some notable biomarkers include adrenomedullin, B-type natriuretic peptide, copeptin, galectin-3, interleukin-6, matrix metalloproteinases (MMPs), midregional pro-atrial natriuretic peptide, myostatin, procollagen type I C-terminal propeptide, procollagen type III N-terminal propeptide and tissue inhibitors of metalloproteinases (TIMPs). These biomarkers aid in HF diagnosis, assessing its severity, and monitoring treatment response, contributing to a deeper understanding of the disease and potentially leading to improved management strategies and outcomes. CONCLUSIONS This review provides comprehensive insights into various in vivo models of HF, commonly utilized cell lines in HF research, and pivotal biomarkers with diagnostic relevance for HF. By synthesizing this information, researchers gain valuable resources to further explore HF pathogenesis, identify novel therapeutic targets, and enhance diagnostic and prognostic approaches.
Collapse
Affiliation(s)
- Anil Kumar Prajapati
- Pharmacology Department, L. M. College of Pharmacy, Ahmedabad, Gujarat, 380009, India
- Research Scholar, Gujarat Technological University, Ahmedabad, Gujarat, 382424, India
| | - Gaurang Shah
- Pharmacology Department, L. M. College of Pharmacy, Ahmedabad, Gujarat, 380009, India.
| |
Collapse
|
4
|
Khazdair MR, Moshtagh M, Anaeigoudari A, Jafari S, Kazemi T. Protective effects of carvacrol on lipid profiles, oxidative stress, hypertension, and cardiac dysfunction - A comprehensive review. Food Sci Nutr 2024; 12:3137-3149. [PMID: 38726397 PMCID: PMC11077248 DOI: 10.1002/fsn3.4014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 01/10/2024] [Accepted: 01/24/2024] [Indexed: 05/12/2024] Open
Abstract
Cardiovascular diseases (CVDs) are a class of illnesses that affect the heart or blood vessels, leading to the most common causes of death worldwide. In 2017, CVD caused approximately 17.8 million deaths that were increased approximately to 20.5 million deaths in 2021, globally. Also, nearly 80% of worldwide CVD deaths occur in some countries. Some herbs and their constituents due to their several pharmacological activities have been used for medicinal purposes. Carvacrol is a phenolic mono-terpenoid found in the oils of aromatic herbs with several biological properties. The possible therapeutic effects of carvacrol on lipid profiles, oxidative stress, hypertension, and cardiac dysfunction were summarized in the current study. The data from this review article were obtained by searching the terms including; "Carvacrol", "Hypertension", Hypotensive, "Cardiac dysfunction", "Ischemia", "Lipid profile", and Oxidative stress in several web databases such as Web of Sciences, PubMed Central, and Google Scholar, until November 2023. The results of the reviewed studies revealed that carvacrol inhibits acetylcholinesterase (AchE) activity and alters lipid profiles, reducing heart rate as well as systolic and diastolic blood pressure (BP). Carvacrol also decreased the proinflammatory cytokine (IL-1β), while increasing secretion of anti-inflammatory cytokine (IL-10). Moreover, carvacrol improved oxidative stress and mitigated the number of apoptotic cells. The pharmacological effects of carvacrol on CVD might be through its antioxidative, anti-inflammatory, and antiapoptotic effects. The mentioned therapeutic effects of carvacrol on lipid profile, hypertension, and cardiac dysfunction indicate the possible remedy effect of carvacrol for the treatment of CVD.
Collapse
Affiliation(s)
- Mohammad Reza Khazdair
- Cardiovascular Diseases Research CenterBirjand University of Medical SciencesBirjandIran
| | - Mozhgan Moshtagh
- Social Determinants of Health Research CenterBirjand University of Medical SciencesBirjandIran
| | - Akbar Anaeigoudari
- Department of Physiology, School of MedicineJiroft University of Medical SciencesJiroftIran
| | - Shima Jafari
- Cardiovascular Diseases Research CenterBirjand University of Medical SciencesBirjandIran
- Department of Clinical Pharmacy, School of PharmacyBirjand University of Medical SciencesBirjandIran
| | - Toba Kazemi
- Cardiovascular Diseases Research CenterBirjand University of Medical SciencesBirjandIran
| |
Collapse
|
5
|
Fandl HK, Garcia VP, Treuth JW, Brewster LM, Greiner JJ, Davy KP, Stauffer BL, Desouza CA. Endothelial-derived extracellular vesicles from obese/hypertensive adults increase factors associated with hypertrophy and fibrosis in cardiomyocytes. Am J Physiol Heart Circ Physiol 2023; 324:H675-H685. [PMID: 36930654 PMCID: PMC10085555 DOI: 10.1152/ajpheart.00035.2023] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 03/08/2023] [Accepted: 03/08/2023] [Indexed: 03/18/2023]
Abstract
Obesity and hypertension, independently and combined, are associated with increased risk of heart failure and heart failure-related morbidity and mortality. Interest in circulating endothelial cell-derived microvesicles (EMVs) has intensified because of their involvement in the development and progression of endothelial dysfunction, atherosclerosis, and cardiomyopathy. The experimental aim of this study was to determine, in vitro, the effects of EMVs isolated from obese/hypertensive adults on key proteins regulating cardiomyocyte hypertrophy [cardiac troponin T (cTnT), α-actinin, nuclear factor-kB (NF-kB)] and fibrosis [transforming growth factor (TGF)-β, collagen1-α1], as well as endothelial nitric oxide synthase (eNOS) expression and nitric oxide (NO) production. EMVs (CD144+ microvesicles) were isolated from plasma by flow cytometry in 12 normal weight/normotensive [8 males/4 females; age: 56 ± 5 yr; body mass index (BMI): 23.3 ± 2.0 kg/m2; blood pressure (BP): 117/74 ± 4/5 mmHg] and 12 obese/hypertensive (8 males/4 females; 57 ± 5 yr; 31.7 ± 1.8 kg/m2; 138/83 ± 8/7 mmHg) adults. Human-induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) were cultured and treated with EMVs from either normal weight/normotensive or obese/hypertensive adults for 24 h. Expression of cTnT (64.1 ± 13.9 vs. 29.5 ± 7.8 AU), α-actinin (66.0 ± 14.7 vs. 36.2 ± 10.3 AU), NF-kB (166.3 ± 13.3 vs. 149.5 ± 8.8 AU), phosphorylated-NF-kB (226.1 ± 25.2 vs. 179.1 ± 25.5 AU), and TGF-β (62.1 ± 13.3 vs. 23.5 ± 8.8 AU) were significantly higher and eNOS activation (16.4 ± 4.3 vs. 24.8 ± 3.7 AU) and nitric oxide production (6.8 ± 1.2 vs. 9.6 ± 1.3 µmol/L) were significantly lower in iPSC-CMs treated with EMVs from obese/hypertensive compared with normal weight/normotensive adults. These data indicate that EMVs from obese/hypertensive adults induce a cardiomyocyte phenotype prone to hypertrophy, fibrosis, and reduced nitric oxide production, central factors associated with heart failure risk and development.NEW & NOTEWORTHY In the present study we determined the effect of endothelial microvesicles (EMVs) isolated from obese/hypertensive adults on mediators of cardiomyocyte hypertrophy [cardiac troponin T (cTnT), α-actinin, nuclear factor-kB (NF-kB)] and fibrosis [transforming growth factor (TGF-β), collagen1-α1] as well as endothelial nitric oxide synthase (eNOS) expression and NO production. EMVs from obese/hypertensive induced significantly higher expression of hypertrophic (cTnT, α-actinin, NF-kB) and fibrotic (TGF-β) proteins as well as significantly lower eNOS activation and NO production in cardiomyocytes than EMVs from normal weight/normotensive adults. EMVs are a potential mediating factor in the increased risk of cardiomyopathy and heart failure with obesity/hypertension.
Collapse
Affiliation(s)
- Hannah K Fandl
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado, Boulder, Colorado, United States
| | - Vinicius P Garcia
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado, Boulder, Colorado, United States
| | - John W Treuth
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado, Boulder, Colorado, United States
| | - Lillian M Brewster
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado, Boulder, Colorado, United States
| | - Jared J Greiner
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado, Boulder, Colorado, United States
| | - Kevin P Davy
- Human Integrative Physiology Laboratory, Department of Human Nutrition, Foods, and Exercise, Virginia Tech University, Blacksburg, Virginia, United States
| | - Brian L Stauffer
- Division of Cardiology, Denver Health Medical Center, Denver, Colorado, United States
- Division of Cardiology, Anschutz Medical Center, University of Colorado, Denver, Colorado, United States
| | - Christopher A Desouza
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado, Boulder, Colorado, United States
- Division of Cardiology, Anschutz Medical Center, University of Colorado, Denver, Colorado, United States
| |
Collapse
|
6
|
Liu J, Li X, Ding L, Li W, Niu X, Gao D. GRK2 participation in cardiac hypertrophy induced by isoproterenol through the regulation of Nrf2 signaling and the promotion of NLRP3 inflammasome and oxidative stress. Int Immunopharmacol 2023; 117:109957. [PMID: 37012864 DOI: 10.1016/j.intimp.2023.109957] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 02/23/2023] [Accepted: 02/26/2023] [Indexed: 03/17/2023]
Abstract
OBJECTIVE In cases of heart failure, cardiac hypertrophy may be caused by the upregulation of G-protein-coupled receptor kinase 2 (GRK2). Both NLRP3 inflammasome and oxidative stress contribute to cardiovascular disease. In this study, we clarified the effect of GRK2 on cardiac hypertrophy in H9c2 cells induced by isoproterenol (ISO) and examined the underlying mechanisms. METHODS We randomly categorized H9c2 cells into five groups: an ISO group, a paroxetine plus ISO group, a GRK2 small-interfering RNA (siRNA) plus ISO group, a GRK2 siRNA combined with ML385 plus ISO group, and a control group. To determine the effect of GRK2 on cardiac hypertrophy induced by ISO, we carried out CCK8 assays, RT-PCR, TUNEL staining, ELISA assay, DCFH-DA staining, immunofluorescence staining, and western blotting. RESULTS By using paroxetine or siRNA to inhibit GRK2, we significantly decreased cell viability; reduced the mRNA levels of ANP, BNP, and β-MHC; and limited the apoptosis rate and protein levels of cleaved caspase-3 and cytochrome c in H9c2 cells treated with ISO. We also found that oxidative stress induced by ISO could be mitigated with paroxetine or GRK2 siRNA. This result was validated by decreased activities of the antioxidant enzymes CAT, GPX, and SOD and increased MDA levels and ROS production. We observed that the protein expression of NLRP3, ASC, and caspase-1 and the intensity of NLRP3 could be inhibited by paroxetine or GRK2 siRNA. Both paroxetine and GRK2 siRNA were able to abolish the increase in GRK2 expression induced by ISO. They also could increase protein levels of HO-1, nuclear Nrf2, and Nrf2 immunofluorescence intensity; however, they could not change the protein level of cytoplasmic Nrf2. By combining treatment with ML385, we were able to reverse GRK2 inhibition on H9c2 cells treated with ISO. CONCLUSION According to the results of this study, GRK2 participated in cardiac hypertrophy induced by ISO by mitigating NLRP3 inflammasome and oxidative stress through the signaling of Nrf2 in H9c2 cells.
Collapse
Affiliation(s)
- Jing Liu
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710068, Shaanxi Province, China; Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xi'an 710038, Shaanxi Province, China
| | - Xiaoli Li
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710068, Shaanxi Province, China
| | - Lu Ding
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xi'an 710038, Shaanxi Province, China
| | - Wei Li
- Department of Cardiology, Xi'an International Medical Center Hospital, Xi'an 710100, Shaanxi Province, China
| | - Xiaolin Niu
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xi'an 710038, Shaanxi Province, China.
| | - Dengfeng Gao
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710068, Shaanxi Province, China.
| |
Collapse
|
7
|
Aminzadeh A, Darijani MH, Bashiri H. Investigating the effect of myricetin against arsenic-induced cardiac toxicity in rats. Toxicol Res (Camb) 2023; 12:117-123. [PMID: 36866219 PMCID: PMC9972817 DOI: 10.1093/toxres/tfad003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 01/04/2023] [Accepted: 01/10/2023] [Indexed: 02/05/2023] Open
Abstract
Arsenic intoxication is a serious health hazard worldwide. Its toxicity is associated with several disorders and health problems in humans. Recent studies revealed that myricetin has various biological effects, including anti-oxidation. The aim of this study is to investigate the protective effect of myricetin against arsenic-induced cardiotoxicity in rats. Rats were randomized to one of the following groups: control, myricetin (2 mg/kg), arsenic (5 mg/kg), myricetin (1 mg/kg) + arsenic, and myricetin (2 mg/kg) + arsenic. Myricetin was given intraperitoneally 30 min before arsenic administration (5 mg/kg for 10 days). After treatments, the activity of lactate dehydrogenase (LDH) and the levels of aspartate aminotransferase (AST), creatine kinase myocardial band (CK-MB), lipid peroxidation (LPO), total antioxidant capacity (TAC), and total thiol molecules (TTM) were determined in serum samples and cardiac tissues. Also, histological changes in cardiac tissue were evaluated. Myricetin pretreatment inhibited arsenic-induced increase in LDH, AST, CK-MB, and LPO levels. Pretreatment with myricetin also enhanced the decreased TAC and TTM levels. In addition, myricetin improved histopathological alterations in arsenic-treated rats. In conclusion, the results of the present study demonstrated that treatment with myricetin prevented arsenic-induced cardiac toxicity at least in part by decreasing oxidative stress and restoring the antioxidant system.
Collapse
Affiliation(s)
- Azadeh Aminzadeh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman 7616911319, Iran
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman 7616911319, Iran
| | - Mohammad Hossein Darijani
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman 7616911319, Iran
| | - Hamideh Bashiri
- Department of Physiology and Pharmacology, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman 7616914115, Iran
| |
Collapse
|
8
|
Yuan M, Jia H, Zhao B, Zhang C, Zuo X. Long noncoding RNA Mhrt alleviates angiotensin II-induced cardiac hypertrophy phenotypes by mediating the miR-765/Wnt family member 7B pathway. Open Med (Wars) 2023; 18:20230681. [PMID: 37197359 PMCID: PMC10183725 DOI: 10.1515/med-2023-0681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 02/08/2023] [Accepted: 02/28/2023] [Indexed: 05/19/2023] Open
Abstract
Long noncoding RNAs (lncRNAs) are known to participate in the pathological process of cardiac hypertrophy. This study aimed to investigate the function of the lncRNA, myosin heavy-chain associated RNA transcript (Mhrt), in cardiac hypertrophy and its possible mechanism of action. Adult mouse cardiomyocytes were treated with angiotensin II (Ang II) and transfected with Mhrt; cardiac hypertrophy was evaluated by estimating atrial natriuretic peptide, brain natriuretic peptide, and beta-myosin heavy-chain levels, and cell surface area by reverse transcription-quantitative polymerase chain reaction, western blotting, and immunofluorescence staining. The interaction between the Mhrt/Wnt family member 7B (WNT7B) and miR-765 was assessed using a luciferase reporter assay. Rescue experiments were performed by analyzing the role of the miR-765/WNT7B pathway underlying the function of Mhrt. The results indicated that Ang II induced hypertrophy of cardiomyocytes; however, overexpression of Mhrt alleviated the Ang II-induced cardiac hypertrophy. Mhrt acted as a sponge for miR-765 to regulate the expression of WNT7B. Rescue experiments revealed that the inhibitory effect of Mhrt on myocardial hypertrophy was abolished by miR-765. Additionally, the knockdown of WNT7B reversed the suppression of myocardial hypertrophy induced by downregulating miR-765. Taken together, Mhrt alleviated cardiac hypertrophy by targeting the miR-765/WNT7B axis.
Collapse
Affiliation(s)
- Manli Yuan
- Department of Ultrasound Medicine, Strategic Support Force Medical Center, Beijing, 100101, China
| | - Huaping Jia
- Department of Ultrasound Medicine, Strategic Support Force Medical Center, Beijing, 100101, China
| | - Bei Zhao
- Department of Cardiovascular Medicine, Strategic Support Force Medical Center, Beijing, China
| | - Can Zhang
- Department of Ultrasound Medicine, Strategic Support Force Medical Center, Beijing, 100101, China
| | - Xiaowen Zuo
- Department of Ultrasound Medicine, Strategic Support Force Medical Center, Beijing, 100101, China
| |
Collapse
|
9
|
Minassa VS, Aitken AV, Hott SC, de Sousa GJ, Batista TJ, Gonçalves RDCR, Coitinho JB, Paton JFR, Beijamini V, Bissoli NS, Sampaio KN. Intermittent exposure to chlorpyrifos results in cardiac hypertrophy and oxidative stress in rats. Toxicology 2022; 482:153357. [PMID: 36341877 DOI: 10.1016/j.tox.2022.153357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 09/28/2022] [Accepted: 10/24/2022] [Indexed: 11/05/2022]
Abstract
Forbidden in some countries due to its proven toxicity to humans, chlorpyrifos (CPF) still stands as an organophosphate pesticide (OP) highly used worldwide. Cardiotoxicity assessment is an unmet need in pesticide regulation and should be deeply studied through different approaches to better inform and generate an appropriate regulatory response to OP use. In the present study, we used our 4-week intermittent OP exposure model in rats to address the CPF effects on cardiac morphology allied with cardiovascular functional and biomolecular evaluation. Rats were intermittently treated with CPF at doses of 7 mg/kg and 10 mg/kg or saline (i.p.) and assessed for cardiac morphology (cardiomyocyte diameter and collagen content), cardiopulmonary Bezold-Jarisch reflex (BJR) function, cardiac autonomic tone, left ventricle (LV) contractility, cardiac expression of NADPH oxidase (Nox2), catalase (CAT), superoxide dismutase 1 (SOD1), superoxide dismutase 2 (SOD2) and cardiac levels of advanced oxidation protein products (AOPP) and thiobarbituric acid reactive substances (TBARS). Plasma butyrylcholinesterase (BuChE) and brainstem acetylcholinesterase (AChE) were also measured. Intermittent exposure to CPF induced cardiac hypertrophy, increasing cardiomyocyte diameter and collagen content. An impairment of cardioinhibitory BJR responses and an increase in cardiac vagal tone were also observed in CPF-treated animals without changes in LV contractility. CPF exposure increased cardiac Nox-2, CAT, SOD1, and TBARS levels and inhibited plasma BuChE and brainstem AChE activities. Our data showed that intermittent exposure to CPF induces cardiac hypertrophy together with cardiovascular reflex impairment, imbalance of autonomic tone and oxidative stress, which may bring significant cardiovascular risk to individuals exposed to OP compounds seasonally.
Collapse
Affiliation(s)
- Vítor Sampaio Minassa
- Postgraduate Program in Pharmaceutical Sciences, Department of Pharmaceutical Sciences, Federal University of Espírito Santo, Vitória, ES, Brazil
| | - Andrew Vieira Aitken
- Postgraduate Program in Pharmaceutical Sciences, Department of Pharmaceutical Sciences, Federal University of Espírito Santo, Vitória, ES, Brazil
| | - Sara Cristina Hott
- Postgraduate Program in Pharmaceutical Sciences, Department of Pharmaceutical Sciences, Federal University of Espírito Santo, Vitória, ES, Brazil
| | - Glauciene Januário de Sousa
- Postgraduate Program in Physiological Sciences, Department of Physiology, Federal University of Espírito Santo, Vitória, ES, Brazil
| | - Thatiany Jardim Batista
- Postgraduate Program in Physiological Sciences, Department of Physiology, Federal University of Espírito Santo, Vitória, ES, Brazil
| | - Rita de Cássia Ribeiro Gonçalves
- Postgraduate Program in Pharmaceutical Sciences, Department of Pharmaceutical Sciences, Federal University of Espírito Santo, Vitória, ES, Brazil
| | - Juliana Barbosa Coitinho
- Postgraduate Program in Pharmaceutical Sciences, Department of Pharmaceutical Sciences, Federal University of Espírito Santo, Vitória, ES, Brazil; Postgraduate Program in Biochemistry, Department of Physiology, Federal University of Espírito Santo, Vitória, ES, Brazil
| | - Julian Francis Richmond Paton
- The Centre for Heart Research - Manaaki Mānawa, Department of Physiology, Faculty of Health & Medical Sciences, University of Auckland, Grafton Campus, Auckland 1023, New Zealand
| | - Vanessa Beijamini
- Postgraduate Program in Pharmaceutical Sciences, Department of Pharmaceutical Sciences, Federal University of Espírito Santo, Vitória, ES, Brazil
| | - Nazaré Souza Bissoli
- Postgraduate Program in Physiological Sciences, Department of Physiology, Federal University of Espírito Santo, Vitória, ES, Brazil
| | - Karla Nívea Sampaio
- Postgraduate Program in Pharmaceutical Sciences, Department of Pharmaceutical Sciences, Federal University of Espírito Santo, Vitória, ES, Brazil.
| |
Collapse
|
10
|
Liu Y, Zhang Q, Yang L, Tian W, Yang Y, Xie Y, Li J, Yang L, Gao Y, Xu Y, Liu J, Wang Y, Yan J, Li G, Shen Y, Qi Z. Metformin Attenuates Cardiac Hypertrophy Via the HIF-1α/PPAR-γ Signaling Pathway in High-Fat Diet Rats. Front Pharmacol 2022; 13:919202. [PMID: 35833024 PMCID: PMC9271627 DOI: 10.3389/fphar.2022.919202] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/11/2022] [Indexed: 12/19/2022] Open
Abstract
Coronary artery disease (CAD) and cardiac hypertrophy (CH) are two main causes of ischemic heart disease. Acute CAD may lead to left ventricular hypertrophy (LVH). Long-term and sustained CH is harmful and can gradually develop into cardiac insufficiency and heart failure. It is known that metformin (Met) can alleviate CH; however, the molecular mechanism is not fully understood. Herein, we used high-fat diet (HFD) rats and H9c2 cells to induce CH and clarify the potential mechanism of Met on CH. We found that Met treatment significantly decreased the cardiomyocyte size, reduced lactate dehydrogenase (LDH) release, and downregulated the expressions of hypertrophy markers ANP, VEGF-A, and GLUT1 either in vivo or in vitro. Meanwhile, the protein levels of HIF-1α and PPAR-γ were both decreased after Met treatment, and administrations of their agonists, deferoxamine (DFO) or rosiglitazone (Ros), markedly abolished the protective effect of Met on CH. In addition, DFO treatment upregulated the expression of PPAR-γ, whereas Ros treatment did not affect the expression of HIF-1α. In conclusion, Met attenuates CH via the HIF-1α/PPAR-γ signaling pathway.
Collapse
Affiliation(s)
- Yuansheng Liu
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin, China
| | - Qian Zhang
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | - Lei Yang
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
- Tianjin Institute of Acute Abdominal Diseases of Integrated Traditional Chinese and Western Medicine, Tianjin Nankai Hospital, Tianjin, China
| | - Wencong Tian
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | - Yinan Yang
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
- Tianjin Central Hospital of Gynecology Obstetrics, Tianjin, China
| | - Yuhang Xie
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | - Jing Li
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | - Liang Yang
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | - Yang Gao
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | - Yang Xu
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | - Jie Liu
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | - Yachen Wang
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | - Jie Yan
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | - Guoxun Li
- Xinjiang Production and Construction Corps Hospital, Urumqi, China
- Tianjin Union Medical Center, Tianjin, China
- *Correspondence: Guoxun Li, ; Yanna Shen, ; Zhi Qi,
| | - Yanna Shen
- Department of Microbiology, School of Laboratory Medicine, Tianjin Medical University, Tianjin, China
- *Correspondence: Guoxun Li, ; Yanna Shen, ; Zhi Qi,
| | - Zhi Qi
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
- Xinjiang Production and Construction Corps Hospital, Urumqi, China
- Tianjin Key Laboratory of General Surgery in Construction, Tianjin Union Medical Center, Tianjin, China
- *Correspondence: Guoxun Li, ; Yanna Shen, ; Zhi Qi,
| |
Collapse
|
11
|
Role of puerarin in pathological cardiac remodeling: A review. Pharmacol Res 2022; 178:106152. [DOI: 10.1016/j.phrs.2022.106152] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 02/28/2022] [Accepted: 03/01/2022] [Indexed: 12/22/2022]
|
12
|
Scutellarein protects against cardiac hypertrophy via suppressing TRAF2/NF-κB signaling pathway. Mol Biol Rep 2022; 49:2085-2095. [PMID: 34988890 DOI: 10.1007/s11033-021-07026-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 11/26/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Scutellarein, a widely studied ingredient of scutellaria herbs, has higher bioavailability and solubility than that of scutellarin. Although the scutellarein had been reported to modulate numerous biological functions, its ability in suppressing cardiac hypertrophy remains unclear. Hence, the present study attempted to investigate whether scutellarein played critical roles in preventing phenylephrine (PE)-induced cardiac hypertrophy. METHODS AND RESULTS Immunocytochemistry (ICC) was employed for evaluating the morphology of the treated cardiomyocytes. Real-time PCR and western blot were respectively applied to assess the mRNA levels and protein expression of the relevant molecules. Bioinformatics analyses were carried out to investigate the potential mechanisms by which scutellarein modulated the PE-induced cardiac hypertrophy. The results showed that Scutellarein treatment significantly inhibited PE-induced increase in H9c2 and AC16 cardiomyocyte size. Besides, scutellarein treatment also dramatically suppressed the expression of the cardiac hypertrophic markers: ANP, BNP and β-MHC. Furthermore, the effects of scutellarein on attenuating the cardiac hypertrophy might be mediated by suppressing the activity of TRAF2/NF-κB signaling pathway. CONCLUSIONS Collectively, our data indicated that scutellarein could protect against PE-induced cardiac hypertrophy via regulating TRAF2/NF-κB signaling pathway using in vitro experiments.
Collapse
|
13
|
Pathophysiology of heart failure and an overview of therapies. Cardiovasc Pathol 2022. [DOI: 10.1016/b978-0-12-822224-9.00025-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
14
|
Sari N, Katanasaka Y, Sugiyama Y, Sunagawa Y, Miyazaki Y, Funamoto M, Shimizu S, Shimizu K, Murakami A, Mori K, Wada H, Hasegawa K, Morimoto T. Zerumbone prevents pressure overload-induced left ventricular systolic dysfunction by inhibiting cardiac hypertrophy and fibrosis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 92:153744. [PMID: 34563985 DOI: 10.1016/j.phymed.2021.153744] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 08/25/2021] [Accepted: 09/07/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Cardiac hypertrophy and fibrosis are hallmarks of cardiac remodeling and are involved functionally in the development of heart failure (HF). However, it is unknown whether Zerumbone (Zer) prevents left ventricular (LV) systolic dysfunction by inhibiting cardiac hypertrophy and fibrosis. PURPOSE This study investigated the effect of Zer on cardiac hypertrophy and fibrosis in vitro and in vivo. STUDY DESIGN/METHODS In primary cultured cardiac cells from neonatal rats, the effect of Zer on phenylephrine (PE)-induced hypertrophic responses and transforming growth factor beta (TGF-β)-induced fibrotic responses was observed. To determine whether Zer prevents the development of pressure overload-induced HF in vivo, a transverse aortic constriction (TAC) mouse model was utilized. Cardiac function was evaluated by echocardiography. The changes of cardiomyocyte surface area were observed using immunofluorescence staining and histological analysis (HE and WGA staining). Collagen synthesis and fibrosis formation were measured by scintillation counter and picrosirius staining, respectively. The total mRNA levels of genes associated with hypertrophy (ANF and BNP) and fibrosis (Postn and α-SMA) were measured by qRT-PCR. The protein expressions (Akt and α-SMA) were assessed by western blotting. RESULTS Zer significantly suppressed PE-induced increase in cell size, mRNA levels of ANF and BNP, and Akt phosphorylation in cardiomyocytes. The TGF-β-induced increase in proline incorporation, mRNA levels of Postn and α-SMA, and protein expression of α-SMA were decreased by Zer in cultured cardiac fibroblasts. In the TAC male C57BL/6 mice, echocardiography results demonstrated that Zer improved cardiac function by increasing LV fractional shortening and reducing LV wall thickness compared with the vehicle group. ZER significantly reduced the level of phosphorylated Akt both in cultured cardiomyocytes treated with PE and in the hearts of TAC. Finally, Zer inhibited the pressure overload-induced cardiac hypertrophy and cardiac fibrosis. CONCLUSION Zer ameliorates pressure overload-induced LV dysfunction, at least in part by suppressing both cardiac hypertrophy and fibrosis.
Collapse
Affiliation(s)
- Nurmila Sari
- Division of Molecular Medicine, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, 422-8526, Japan
| | - Yasufumi Katanasaka
- Division of Molecular Medicine, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, 422-8526, Japan; Clinical Research Institute, Division of Translational Research, National Hospital Organization Kyoto Medical Center, Kyoto, 612-8555, Japan; Shizuoka General Hospital, Shizuoka, Japan
| | - Yuga Sugiyama
- Division of Molecular Medicine, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, 422-8526, Japan
| | - Yoichi Sunagawa
- Division of Molecular Medicine, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, 422-8526, Japan; Clinical Research Institute, Division of Translational Research, National Hospital Organization Kyoto Medical Center, Kyoto, 612-8555, Japan; Shizuoka General Hospital, Shizuoka, Japan
| | - Yusuke Miyazaki
- Division of Molecular Medicine, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, 422-8526, Japan
| | - Masafumi Funamoto
- Division of Molecular Medicine, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, 422-8526, Japan
| | - Satoshi Shimizu
- Division of Molecular Medicine, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, 422-8526, Japan
| | - Kana Shimizu
- Division of Molecular Medicine, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, 422-8526, Japan
| | - Akira Murakami
- Department of Food Science and Nutrition, School of Human Science and Environment, University of Hyogo, Hyogo, Japan
| | - Kiyoshi Mori
- Division of Molecular Medicine, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, 422-8526, Japan
| | - Hiromichi Wada
- Clinical Research Institute, Division of Translational Research, National Hospital Organization Kyoto Medical Center, Kyoto, 612-8555, Japan
| | - Koji Hasegawa
- Clinical Research Institute, Division of Translational Research, National Hospital Organization Kyoto Medical Center, Kyoto, 612-8555, Japan
| | - Tatsuya Morimoto
- Division of Molecular Medicine, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, 422-8526, Japan; Clinical Research Institute, Division of Translational Research, National Hospital Organization Kyoto Medical Center, Kyoto, 612-8555, Japan; Shizuoka General Hospital, Shizuoka, Japan.
| |
Collapse
|
15
|
Lu J, Ma X, Gao WC, Zhang X, Fu Y, Liu Q, Tian L, Qin XD, Yang W, Zheng HY, Zheng CB. Gastrodin Exerts Cardioprotective Action via Inhibition of Insulin-Like Growth Factor Type 2/Insulin-Like Growth Factor Type 2 Receptor Expression in Cardiac Hypertrophy. ACS OMEGA 2021; 6:16763-16774. [PMID: 34250336 PMCID: PMC8264851 DOI: 10.1021/acsomega.1c00797] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 06/10/2021] [Indexed: 05/05/2023]
Abstract
Pathological cardiac hypertrophy is commonly associated with an upregulation of fetal genes, fibrosis, cardiac dysfunction, and heart failure. Previous studies have demonstrated that gastrodin (GAS) exerts cardioprotective action in the treatment of cardiac hypertrophy. However, the mechanism by which GAS protects against cardiac hypertrophy is yet to be elucidated. A mouse model of myocardial hypertrophy was established using an angiotensin II (Ang II) induction. GAS (5 or 50 mg/kg/d) was orally administered every day starting 7 days prior to the Ang II infusion combined with sham-operated controls. Heart samples from each group were collected for RNA sequencing. Using bioinformatics analysis, the key differentially expressed genes (DEGs) that are involved in reversing cardiac function were identified. Through bioinformatics analysis, the key DEGs that are involved in GAS's inhibition of Ang II-induced abnormal gene expression within the heart were identified. This was further validated using quantitative real-time PCR and Western blotting in neonatal rat cardiomyocytes (NRCMs). Oral administration of GAS significantly suppressed the Ang II-induced increase in heart size and heart weight to body weight. Furthermore, pretreatment of the NRCMs with GAS led to a dose-dependent inhibition of Ang II-induced increases in Nppb mRNA expression. We identified 620 upregulated and 87 downregulated Ang II-induced DEGs II, among which the expression patterns of 58 and 146 genes were inverted by low-dose and high-dose GAS, respectively. These inverted DEGs were found to be mainly enriched in the biological processes of regulation of Ras protein signal transduction, heart contraction, covalent chromatin modification, glucose metabolism, and positive regulation of cell cycle. Among them, the insulin-like growth factor type 2 (Igf2) gene, which was found to be highly reversed and downregulated by GAS, served as a core gene linking energy metabolism, immune regulation, and systemic development. Subsequent functional verification demonstrated that IGF2, and its receptor IGF2R, is one of the targets of GAS that helps protect against cardiac hypertrophy. Taken together, we have identified, for the first time, IGF2/IGF2R as a potential target influenced by GAS in the prevention of cardiac hypertrophy.
Collapse
Affiliation(s)
- Jun Lu
- Department
of Pharmacology, Guilin Medical University, Guilin 541199, China
| | - Xin Ma
- School
of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology
for Natural Products, Kunming Medical University, Kunming 650500, China
- Key
Laboratory of Animal Models and Human Diseases Mechanisms of Chinese
Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
| | - Wen-Cong Gao
- School
of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology
for Natural Products, Kunming Medical University, Kunming 650500, China
- Key
Laboratory of Animal Models and Human Diseases Mechanisms of Chinese
Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
| | - Xin Zhang
- School
of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology
for Natural Products, Kunming Medical University, Kunming 650500, China
| | - Yuanling Fu
- School
of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology
for Natural Products, Kunming Medical University, Kunming 650500, China
| | - Qian Liu
- School
of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology
for Natural Products, Kunming Medical University, Kunming 650500, China
- Key
Laboratory of Animal Models and Human Diseases Mechanisms of Chinese
Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
| | - Lixiang Tian
- School
of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology
for Natural Products, Kunming Medical University, Kunming 650500, China
| | - Xiao-Dan Qin
- Department
of Pharmacology, Guilin Medical University, Guilin 541199, China
| | - Weimin Yang
- School
of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology
for Natural Products, Kunming Medical University, Kunming 650500, China
| | - Hong-Yi Zheng
- Key
Laboratory of Animal Models and Human Diseases Mechanisms of Chinese
Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
| | - Chang-Bo Zheng
- School
of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology
for Natural Products, Kunming Medical University, Kunming 650500, China
| |
Collapse
|
16
|
Zheng CB, Gao WC, Xie M, Li Z, Ma X, Song W, Luo D, Huang Y, Yang J, Zhang P, Huang Y, Yang W, Yao X. Ang II Promotes Cardiac Autophagy and Hypertrophy via Orai1/STIM1. Front Pharmacol 2021; 12:622774. [PMID: 34079454 PMCID: PMC8165566 DOI: 10.3389/fphar.2021.622774] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 03/16/2021] [Indexed: 01/07/2023] Open
Abstract
The pathophysiology of cardiac hypertrophy is complex and multifactorial. Both the store-operated Ca2+ entry (SOCE) and excessive autophagy are the major causative factors for pathological cardiac hypertrophy. However, it is unclear whether these two causative factors are interdependent. In this study, we examined the functional role of SOCE and Orai1 in angiotensin II (Ang II)-induced autophagy and hypertrophy using in vitro neonatal rat cardiomyocytes (NRCMs) and in vivo mouse model, respectively. We show that YM-58483 or SKF-96365 mediated pharmacological inhibition of SOCE, or silencing of Orai1 with Orail-siRNA inhibited Ang II-induced cardiomyocyte autophagy both in vitro and in vivo. Also, the knockdown of Orai1 attenuated Ang II-induced pathological cardiac hypertrophy. Together, these data suggest that Ang II promotes excessive cardiomyocyte autophagy through SOCE/Orai1 which can be the prime contributing factors in cardiac hypertrophy.
Collapse
Affiliation(s)
- Chang-Bo Zheng
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, China
| | - Wen-Cong Gao
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, China
| | - Mingxu Xie
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, China
| | - Zhichao Li
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, China
| | - Xin Ma
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, China
| | - Wencong Song
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, China
| | - Dan Luo
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, China
| | - Yongxiang Huang
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, China
| | - Jichen Yang
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, China
| | - Peng Zhang
- Longgang E.N.T. Hospital and Shenzhen Key Laboratory of E.N.T., Shenzhen, China
| | - Yu Huang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, China
| | - Weimin Yang
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, China
| | - Xiaoqiang Yao
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, China
| |
Collapse
|
17
|
Zhu L, Li YL, Qian ZQ, Hua L, Yue Y, Yang DL. Osthole improves pulmonary artery hypertension by inducing apoptosis in pulmonary artery smooth muscle cells. J Pharm Pharmacol 2021; 73:1109-1117. [PMID: 33988241 DOI: 10.1093/jpp/rgab068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 04/22/2021] [Indexed: 11/13/2022]
Abstract
OBJECTIVES The objectives of this study were to explore the effect of Osthole (Ost) on apoptosis in pulmonary artery smooth muscle cells (PASMCs) and investigate the potential mechanism of this effect. METHODS Rats were injected subcutaneously with monocrotaline (MCT) to establish a PAH model, and Ost were intragastrically administrated from day 1 to day 35. After 35 days administration, the mean pulmonary artery pressure and lung weight index were measured. HE and TUNEL staining were used to observe the morphology of pulmonary artery and the apoptosis of PASMCs. In addition, the apoptosis of PASMCs were detected by flow cytometry in cultured PASMCs. The proteins of Bax and Bcl-2, and the levels of p-ASK1 and cleaved caspase 3 were measured by Western blot. KEY FINDINGS Ost decreased the mean pulmonary artery pressure and lung weight index in MCT-induced rats, and promoted apoptosis in PASMCs in MCT-induced rats and PDGF-BB stimulated PASMCs. Ost increased the ratio of Bax/Bcl-2 and the levels of p-ASK1, cleaved caspase 3 in MCT-induced rats and PDGF-BB stimulated PASMCs. CONCLUSION Ost promoted apoptosis in PASMCs in vivo and in vitro, and the mechanism may be associated with upregulation of ASK1 and the Bax/Bcl-2-caspase 3 signalling pathway.
Collapse
Affiliation(s)
- Ling Zhu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Key Laboratory of Basic Pharmacology of Guizhou Province, Department of Pharmacology, School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China.,School of Nursing, Qiannan Medical College for Nationalities, Anshun, Guizhou, China
| | - Ye-Li Li
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Key Laboratory of Basic Pharmacology of Guizhou Province, Department of Pharmacology, School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China
| | - Zhi-Qiang Qian
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Key Laboratory of Basic Pharmacology of Guizhou Province, Department of Pharmacology, School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China
| | - Liang Hua
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Key Laboratory of Basic Pharmacology of Guizhou Province, Department of Pharmacology, School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China
| | - Yun Yue
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Key Laboratory of Basic Pharmacology of Guizhou Province, Department of Pharmacology, School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China
| | - Dan-Li Yang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Key Laboratory of Basic Pharmacology of Guizhou Province, Department of Pharmacology, School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China
| |
Collapse
|
18
|
Wu X, Huang L, Liu J. Relationship between oxidative stress and nuclear factor-erythroid-2-related factor 2 signaling in diabetic cardiomyopathy (Review). Exp Ther Med 2021; 22:678. [PMID: 33986843 PMCID: PMC8111863 DOI: 10.3892/etm.2021.10110] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 08/26/2020] [Indexed: 12/12/2022] Open
Abstract
Diabetic cardiomyopathy (DCM) is the leading cause of death worldwide, and oxidative stress was discovered to serve an important role in the pathophysiology of the condition. An imbalance between free radicals and antioxidant defenses is known to be associated with cellular dysfunction, leading to the development of various types of cardiac disease. Nuclear factor-erythroid-2-related factor 2 (NRF2) is a transcription factor that controls the basal and inducible expression levels of various antioxidant genes and other cytoprotective phase II detoxifying enzymes, which are ubiquitously expressed in the cardiac system. Kelch-like ECH-associated protein 1 (Keap1) serves as the main intracellular regulator of NRF2. Emerging evidence has revealed that NRF2 is a critical regulator of cardiac homeostasis via the suppression of oxidative stress. The activation of NRF2 was discovered to enhance specific endogenous antioxidant defense factors, one of which is antioxidant response element (ARE), which was subsequently illustrated to detoxify and counteract oxidative stress-associated DCM. The NRF2 signaling pathway is closely associated with the development of various types of cardiac disease, including ischemic heart disease, heart failure, myocardial infarction, atrial fibrillation and myocarditis. Therefore, it is hypothesized that drugs targeting this pathway may be developed to inhibit the activation of NRF2 signaling, thereby preventing the occurrence of DCM and effectively treating the disease.
Collapse
Affiliation(s)
- Xia Wu
- Department of Pharmacy, Nanchang University, Nanchang, Jiangxi 330006, P.R. China.,Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Leitao Huang
- Department of Orthopedics, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210031, P.R. China
| | - Jichun Liu
- Department of Pharmacy, Nanchang University, Nanchang, Jiangxi 330006, P.R. China.,Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
19
|
Xu X, Xie X, Zhang H, Wang P, Li G, Chen J, Chen G, Cao X, Xiong L, Peng F, Peng C. Water-soluble alkaloids extracted from Aconiti Radix lateralis praeparata protect against chronic heart failure in rats via a calcium signaling pathway. Biomed Pharmacother 2021; 135:111184. [PMID: 33418305 DOI: 10.1016/j.biopha.2020.111184] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 12/15/2020] [Accepted: 12/26/2020] [Indexed: 11/16/2022] Open
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Many studies have shown the beneficial effects of aconite water-soluble alkaloid extract (AWA) in experimental models of heart disease, which have been ascribed to the presence of aconine, hypaconine, talatisamine, fuziline, neoline, and songorine. This study evaluated the effects of a chemically characterized AWA by chemical content, evaluated its effects in suprarenal abdominal aortic coarctation surgery (AAC)-induced chronic heart failure (CHF) in rats, and revealed the underlying mechanisms of action by proteomics. METHODS Rats were distributed into different groups: sham, model, and AWA-treated groups (10, 20, and 40 mg/kg/day). Sham rats received surgery without AAC, whereas model rats an AWA-treated groups underwent AAC surgery. after 8 weeks, the treatment group was fed AWA for 4 weeks, and body weight was assessed weekly. At the end of the treatment, heart function was tested by echocardiography. AAC-induced chronic heart failure, including myocardial fibrosis, cardiomyocyte hypertrophy, and apoptosis, was evaluated in heart tissue and plasma by RT-qPCR, ELISA, hematoxylin and eosin (H&E) staining, Masson's trichrome staining, TUNEL staining, and immunofluorescence staining of α-SMA, Col Ⅰ, and Col Ⅲ. Then, a proteomics approach was used to explore the underlying mechanisms of action of AWA in chronic heart failure. RESULTS AWA administration reduced body weight gain, myocardial fibrosis, cardiomyocyte hypertrophy, and apoptosis, and rats showed improvement in cardiac function compared to model group. The extract significantly ameliorated the AAC-induced altered expression of heart failure markers such as ANP, NT-proBNP, and β-MHC, as well as fibrosis, hypertrophy markers MMP-2 and MMP-9, and other heart failure-related factors including plasma levels of TNF-α and IL-6. Furthermore, the extract reduced the protein expression of α-SMA, Col Ⅰ, and Col Ⅲ in the left ventricular (LV), thus inhibiting the LV remodeling associated with CHF. In addition, proteomics characterization of differentially expressed proteins showed that AWA administration inhibited left ventricular remodeling in CHF rats via a calcium signaling pathway, and reversed the expression of RyR2 and SERCA2a. CONCLUSIONS AWA extract exerts beneficial effects in an AAC-induced CHF model in rats, which was associated with an improvement in LV function, hypertrophy, fibrosis, and apoptotic status. These effects may be related to the regulation of calcium signaling by the altered expression of RyR2 and SERCA2a.
Collapse
MESH Headings
- Aconitum/chemistry
- Animals
- Apoptosis/drug effects
- Calcium Signaling/drug effects
- Cardiovascular Agents/isolation & purification
- Cardiovascular Agents/pharmacology
- Chronic Disease
- Disease Models, Animal
- Fibrosis
- Heart Failure/drug therapy
- Heart Failure/metabolism
- Heart Failure/pathology
- Heart Failure/physiopathology
- Hypertrophy, Left Ventricular/drug therapy
- Hypertrophy, Left Ventricular/metabolism
- Hypertrophy, Left Ventricular/pathology
- Hypertrophy, Left Ventricular/physiopathology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Plant Extracts/isolation & purification
- Plant Extracts/pharmacology
- Rats, Sprague-Dawley
- Ryanodine Receptor Calcium Release Channel/metabolism
- Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
- Solubility
- Solvents/chemistry
- Ventricular Dysfunction, Left/drug therapy
- Ventricular Dysfunction, Left/metabolism
- Ventricular Dysfunction, Left/pathology
- Ventricular Dysfunction, Left/physiopathology
- Ventricular Function, Left/drug effects
- Ventricular Remodeling/drug effects
- Water/chemistry
- Rats
Collapse
Affiliation(s)
- Xin Xu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Chengdu 611137, China; Key Laboratory of the Ministry of Education for Standardization of Chinese Medicine Co-founded by Sichuan Province and MOST, Chengdu 611137, China
| | - Xiaofang Xie
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Chengdu 611137, China; Key Laboratory of the Ministry of Education for Standardization of Chinese Medicine Co-founded by Sichuan Province and MOST, Chengdu 611137, China
| | - Huiqiong Zhang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Chengdu 611137, China; Key Laboratory of the Ministry of Education for Standardization of Chinese Medicine Co-founded by Sichuan Province and MOST, Chengdu 611137, China
| | - Pei Wang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Gangmin Li
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Chengdu 611137, China; Key Laboratory of the Ministry of Education for Standardization of Chinese Medicine Co-founded by Sichuan Province and MOST, Chengdu 611137, China
| | - Junren Chen
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Chengdu 611137, China; Key Laboratory of the Ministry of Education for Standardization of Chinese Medicine Co-founded by Sichuan Province and MOST, Chengdu 611137, China
| | - Guanru Chen
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Chengdu 611137, China; Key Laboratory of the Ministry of Education for Standardization of Chinese Medicine Co-founded by Sichuan Province and MOST, Chengdu 611137, China
| | - Xiaoyu Cao
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Liang Xiong
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Chengdu 611137, China; Key Laboratory of the Ministry of Education for Standardization of Chinese Medicine Co-founded by Sichuan Province and MOST, Chengdu 611137, China
| | - Fu Peng
- West China School of Pharmacy, Sichuan University, Chengdu 611137, China.
| | - Cheng Peng
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Chengdu 611137, China; Key Laboratory of the Ministry of Education for Standardization of Chinese Medicine Co-founded by Sichuan Province and MOST, Chengdu 611137, China.
| |
Collapse
|
20
|
Jain A, Behera M, Mahapatra C, Sundaresan NR, Chatterjee K. Nanostructured polymer scaffold decorated with cerium oxide nanoparticles toward engineering an antioxidant and anti-hypertrophic cardiac patch. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 118:111416. [DOI: 10.1016/j.msec.2020.111416] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 08/07/2020] [Accepted: 08/13/2020] [Indexed: 02/06/2023]
|
21
|
Li W, Yang J, Lyu Q, Wu G, Lin S, Yang Q, Hu J. Taurine attenuates isoproterenol-induced H9c2 cardiomyocytes hypertrophy by improving antioxidative ability and inhibiting calpain-1-mediated apoptosis. Mol Cell Biochem 2020; 469:119-132. [PMID: 32304004 DOI: 10.1007/s11010-020-03733-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 04/08/2020] [Indexed: 12/18/2022]
Abstract
Pathological cardiac hypertrophy is ultimately accompanied by cardiomyocyte apoptosis. Apoptosis mainly related to calpain-1-mediated apoptotic pathways. Studies had proved that taurine can maintain heart health through antioxidation and antiapoptotic functions, but the effect of taurine on cardiac hypertrophy is still unclear. This study aimed to determine whether taurine could inhibit calpain-1-mediated mitochondria-dependent apoptotic pathways in isoproterenol (ISO)-induced hypertrophic cardiomyocytes. We found that taurine could inhibit the increase in cell surface area and reduce the protein expression levels of the hypertrophic markers atrial natriuretic peptide, brain natriuretic polypeptide, and β-myosin heavy chain. Taurine also reduced ROS, intracellular Ca2+ overload and mitochondrial membrane potential. Moreover, taurine inhibited cardiomyocyte apoptosis by decreasing the protein expression of calpain-1, Bax, t-Bid, cytosolic cytochrome c, Apaf-1, cleaved caspase-9 and cleaved caspase-3 and by enhancing calpastatin and Bcl-2 protein expression. Calpain-1 small interfering RNA transfection results showed similar antiapoptotic effects as the taurine prevention group. However, compared with the two treatments, taurine inhibited the expression of cleaved caspase-9 more significantly. Therefore, we believe that taurine prevents ISO-induced H9c2 cardiomyocyte hypertrophy by inhibiting oxidative stress, intracellular Ca2+ overload, the calpain-1-mediated mitochondria-dependent apoptotic pathway and cleaved caspase-9 levels.
Collapse
Affiliation(s)
- Weiwei Li
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, Liaoning, People's Republic of China
| | - Jiancheng Yang
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, Liaoning, People's Republic of China
| | - Qiufeng Lyu
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, Liaoning, People's Republic of China
| | - Gaofeng Wu
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, Liaoning, People's Republic of China
| | - Shumei Lin
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, Liaoning, People's Republic of China
| | - Qunhui Yang
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, Liaoning, People's Republic of China.
| | - Jianmin Hu
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, Liaoning, People's Republic of China.
| |
Collapse
|
22
|
Ahmadi F, Ghanbar Zadeh M, Habibi A, Karimi F. Effect of resistance training with Spirulina platensis on PI3K/Akt/mTOR/p70S6k signaling pathway in cardiac muscle. Sci Sports 2020. [DOI: 10.1016/j.scispo.2019.06.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
23
|
Exosomal Nrf2: From anti-oxidant and anti-inflammation response to wound healing and tissue regeneration in aged-related diseases. Biochimie 2020; 171-172:103-109. [PMID: 32109502 DOI: 10.1016/j.biochi.2020.02.011] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 02/19/2020] [Indexed: 12/18/2022]
Abstract
Accumulation of oxidative stress in cells is an essential feature of cellular senescence and aging. This phenomenon is involved in different age-related diseases through dysregulation of homeostasis and impairing repair and regeneration (wound healing) capacity, which can suppress antioxidant responses such as the activity of antioxidant enzymes and damaged protein clearance system. Nuclear factor erythroid 2-related factor 2 (Nrf2) is a transcription factor which regulates basal and inducible expression pattern of specific genes (antioxidants and detoxifications) through antioxidant element response (ARE) sites in the stress condition, specifically in chronic and age-related stresses. Nrf2 maintains cellular redox hemostasis and promotes rejuvenation. Exosomes are nanoscale vesicles that are released by various cells to actively regulate the complex cellular signaling networks. Exosomal-Nrf2 and exosomal-Nrf2-mediated products can modulate oxidative hemostasis in target cells to induce tissue repairing with therapeutic proposes, and regeneration capability. In this study, we summarized the role of exosomal-Nrf2 in different age-related diseases, including diabetic foot ulcers, atherosclerosis, chronic heart failure, reproductive cell failures, and neurodegenerative diseases. In addition, we briefly explained the crosstalk between plant exosomes and mammalian cell metabolism in the benefit of cellular stress suppression.
Collapse
|
24
|
Abstract
Arsenic trioxide (ATO) is among the first-line chemotherapeutic drugs used in oncological practice. It has shown substantial efficacy in treating patients with relapsed or refractory acute promyelocytic leukaemia. The clinical use of ATO is hampered due to cardiotoxicity and hence many patients are precluded from receiving this highly effective treatment. An alternative to this would be to use any drug that can ameliorate the cardiotoxic effects and allow exploiting the full therapeutic potential of ATO, with considerable impact on cancer therapy. Generation of reactive oxygen species is involved in a wide range of human diseases, including cancer, cardiovascular, pulmonary and neurological disorders. Hence, agents with the ability to protect against these reactive species may be therapeutically useful. The present review focuses on the beneficial as well as harmful effects of arsenic and ATO, the mechanisms underlying ATO toxicity and the possible ways that can be adopted to circumvent ATO-induced toxicity.
Collapse
|
25
|
Saucerman JJ, Tan PM, Buchholz KS, McCulloch AD, Omens JH. Mechanical regulation of gene expression in cardiac myocytes and fibroblasts. Nat Rev Cardiol 2019; 16:361-378. [PMID: 30683889 PMCID: PMC6525041 DOI: 10.1038/s41569-019-0155-8] [Citation(s) in RCA: 133] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The intact heart undergoes complex and multiscale remodelling processes in response to altered mechanical cues. Remodelling of the myocardium is regulated by a combination of myocyte and non-myocyte responses to mechanosensitive pathways, which can alter gene expression and therefore function in these cells. Cellular mechanotransduction and its downstream effects on gene expression are initially compensatory mechanisms during adaptations to the altered mechanical environment, but under prolonged and abnormal loading conditions, they can become maladaptive, leading to impaired function and cardiac pathologies. In this Review, we summarize mechanoregulated pathways in cardiac myocytes and fibroblasts that lead to altered gene expression and cell remodelling under physiological and pathophysiological conditions. Developments in systems modelling of the networks that regulate gene expression in response to mechanical stimuli should improve integrative understanding of their roles in vivo and help to discover new combinations of drugs and device therapies targeting mechanosignalling in heart disease.
Collapse
Affiliation(s)
- Jeffrey J Saucerman
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Philip M Tan
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Kyle S Buchholz
- Departments of Bioengineering and Medicine, University of California San Diego, La Jolla, CA, USA
| | - Andrew D McCulloch
- Departments of Bioengineering and Medicine, University of California San Diego, La Jolla, CA, USA.
| | - Jeffrey H Omens
- Departments of Bioengineering and Medicine, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
26
|
Soares DDS, Pinto GH, Lopes A, Caetano DSL, Nascimento TG, Andrades ME, Clausell N, Rohde LEP, Leitão SAT, Biolo A. Cardiac hypertrophy in mice submitted to a swimming protocol: influence of training volume and intensity on myocardial renin-angiotensin system. Am J Physiol Regul Integr Comp Physiol 2019; 316:R776-R782. [DOI: 10.1152/ajpregu.00205.2018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Exercise promotes physiological cardiac hypertrophy and activates the renin-angiotensin system (RAS), which plays an important role in cardiac physiology, both through the classical axis [angiotensin II type 1 receptor (AT1R) activated by angiotensin II (ANG II)] and the alternative axis [proto-oncogene Mas receptor (MASR) activated by angiotensin-(1–7)]. However, very intense exercise could have deleterious effects on the cardiovascular system. We aimed to analyze the cardiac hypertrophy phenotype and the classical and alternative RAS axes in the myocardium of mice submitted to swimming exercises of varying volume and intensity for the development of cardiac hypertrophy. Male Balb/c mice were divided into three groups, sedentary, swimming twice a day without overload (T2), and swimming three times a day with a 2% body weight overload (T3), totaling 6 wk of training. Both training groups developed similar cardiac hypertrophy, but only T3 mice improved their oxidative capacity. We observed that T2 had increased levels of MASR, which was followed by the activation of its main downstream protein AKT; meanwhile, AT1R and its main downstream protein ERK remained unchanged. Furthermore, no change was observed regarding the levels of angiotensin peptides, in either group. In addition, we observed no change in the ratio of expression of the myosin heavy chain β-isoform to that of the α-isoform. Fibrosis was not observed in any of the groups. In conclusion, our results suggest that increasing exercise volume and intensity did not induce a pathological hypertrophy phenotype, but instead improved the oxidative capacity, and this process might have the participation of the RAS alternative axis.
Collapse
Affiliation(s)
- Douglas dos Santos Soares
- Experimental and Molecular Cardiovascular Laboratory and Heart Failure and Cardiac Transplant Unit, Cardiology Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Postgraduate Program in Cardiology and Cardiovascular Science, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Graziela Hünning Pinto
- Experimental and Molecular Cardiovascular Laboratory and Heart Failure and Cardiac Transplant Unit, Cardiology Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Postgraduate Program in Cardiology and Cardiovascular Science, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Amanda Lopes
- Experimental and Molecular Cardiovascular Laboratory and Heart Failure and Cardiac Transplant Unit, Cardiology Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Daniel Sturza Lucas Caetano
- Experimental and Molecular Cardiovascular Laboratory and Heart Failure and Cardiac Transplant Unit, Cardiology Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Thaiane Gomes Nascimento
- Experimental and Molecular Cardiovascular Laboratory and Heart Failure and Cardiac Transplant Unit, Cardiology Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Michael E. Andrades
- Experimental and Molecular Cardiovascular Laboratory and Heart Failure and Cardiac Transplant Unit, Cardiology Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Postgraduate Program in Cardiology and Cardiovascular Science, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Nadine Clausell
- Experimental and Molecular Cardiovascular Laboratory and Heart Failure and Cardiac Transplant Unit, Cardiology Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Postgraduate Program in Cardiology and Cardiovascular Science, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Luis E. Paim Rohde
- Experimental and Molecular Cardiovascular Laboratory and Heart Failure and Cardiac Transplant Unit, Cardiology Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Postgraduate Program in Cardiology and Cardiovascular Science, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Santiago Alonso Tobar Leitão
- Experimental and Molecular Cardiovascular Laboratory and Heart Failure and Cardiac Transplant Unit, Cardiology Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Postgraduate Program in Cardiology and Cardiovascular Science, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Andreia Biolo
- Experimental and Molecular Cardiovascular Laboratory and Heart Failure and Cardiac Transplant Unit, Cardiology Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Postgraduate Program in Cardiology and Cardiovascular Science, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
27
|
Molecular targets of fenofibrate in the cardiovascular-renal axis: A unifying perspective of its pleiotropic benefits. Pharmacol Res 2019; 144:132-141. [PMID: 30970278 DOI: 10.1016/j.phrs.2019.03.025] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 03/28/2019] [Accepted: 03/29/2019] [Indexed: 12/17/2022]
|
28
|
Boureima Oumarou D, Ji H, Xu J, Li S, Ruan W, Xiao F, Yu F. Involvement of microRNA-23b-5p in the promotion of cardiac hypertrophy and dysfunction via the HMGB2 signaling pathway. Biomed Pharmacother 2019; 116:108977. [PMID: 31103821 DOI: 10.1016/j.biopha.2019.108977] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 05/03/2019] [Accepted: 05/08/2019] [Indexed: 01/08/2023] Open
Abstract
The processes involved in the progression of myocardial cells towards hypertrophy and its gradual transition to heart failure represent a multifactorial health disorder. The aim of this study was to identify the molecular mechanism(s) underlying the abnormal overexpression of miR-23b-5p and its involvement in the promotion of cardiac hypertrophy and dysfunction via HMGB2. A type 9 recombinant adeno-associated virus (rAAV9) was employed to manipulate miR-23b-5p expression under conditions of thoracic aortic constriction (TAC)-/angiotensin-II (Ang-II)-induced cardiac dysfunction. Cardiac structures and functions were assessed by echocardiography and invasive pressure-volume analysis. HMGB2 expression under conditions of cardiac hypertrophy was detected by western blotting. The biochemical relationship between miR-23b-5p and HMGB2 was verified using a luciferase reporter vector, lentiviral construct comprising the miR-23b-5p mimic sequence, and microRNA inhibitor (miR-inhibitor). The expression levels of miR-23b-5p were increased in the hearts under conditions of both Ang-II- and TAC-induced cardiac hypertrophy. The results of the luciferase activity analysis showed that HMGB2 is a supposed target of miR-23b-5p. miR-23b-5p overexpression in vivo aggravated pressure overload-induced cardiac hypertrophy and dysfunction, whereas the miR-inhibitor increased HMGB2 expression and reversed these effects. In the present study, we observed that miR-23b-5p mediates and is involved in the aggravation of cardiac hypertrophy and dysfunction via the HMGB2 signaling pathway.
Collapse
Affiliation(s)
- Diafara Boureima Oumarou
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China
| | - Heyu Ji
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China
| | - Junmei Xu
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China
| | - Suobei Li
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China
| | - Wei Ruan
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China
| | - Feng Xiao
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China.
| | - Fei Yu
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China; Department of Anesthesiology, Affiliated Hospital of Binzhou Medical University, Binzhou, Shandong 256603, PR China.
| |
Collapse
|
29
|
Jahanifar F, Astani A, Shekarforoosh S, Jamhiri M, Safari F, Zarei F, Safari F. 1.25 Dihydroxyvitamin D3 Attenuates Hypertrophy Markers in Cardiomyoblast H9c2 Cells: Evaluation of Sirtuin3 mRNA and Protein Level. INT J VITAM NUTR RES 2019; 89:144-151. [PMID: 30856082 DOI: 10.1024/0300-9831/a000469] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Background: The cellular and molecular mechanisms of cardioprotective effects of Vitamin D are poorly understood. Given the essential role of sirtuin-3 (SIRT3) as an endogenous negative regulator of cardiac hypertrophy, this study was designed to investigate the effect of 1, 25-dihydroxyvitamin D3 (calcitriol) on hypertrophy markers and SIRT3 mRNA and protein levels following angiotensin II induced - hypertrophy in cardiomyoblast H9c2 cells. Methods: Rat cardiomyoblast H9c2 cells were treated for 48 hr with angiotensin II alone (Ang group) or in combination with 1, 10 and 100 nM of calcitriol (C + Ang groups). Intact cells served as control (Ctl). The cell area was measured using methylene blue staining. Atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP) and SIRT3 transcription levels were measured by real time RT-PCR. SIRT3 protein expression was evaluated using western blot technique. Results: The results showed that in Ang group cell size was increase by 128.4 ± 15% (P < 0.001 vs. Ctl) whereas in C100 + Ang group it was increased by 21.3 ± 6% (P < 0.001 vs. Ang group). Calcitriol pretreatment decreased ANP mRNA level significantly (P < 0.05) in comparison with Ang group (Ang: 75.5 ± 15%, C100 + Ang: 19.2 ± 9%). There were no significant differences between Ang group and cells pretreated with 1 and 10 nM of calcitriol. SIRT3 at mRNA and protein levels did not change significantly among the experimental groups. Conclusions: In conclusion, pretreatment with calcitriol (100 nM) prevents Ang II-induced hypertrophy in cardiomyoblast H9c2 cells. This probably occurs through other pathways except SIRT3 upregulation.
Collapse
Affiliation(s)
- Fatemeh Jahanifar
- Department of Physiology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Akram Astani
- Department of Microbiology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | | | - Mohabbat Jamhiri
- Department of Physiology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | | | - Farideh Zarei
- Premature Neonates Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fatemeh Safari
- Department of Physiology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
30
|
Gupta I, Varshney NK, Khan S. Emergence of Members of TRAF and DUB of Ubiquitin Proteasome System in the Regulation of Hypertrophic Cardiomyopathy. Front Genet 2018; 9:336. [PMID: 30186311 PMCID: PMC6110912 DOI: 10.3389/fgene.2018.00336] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 08/03/2018] [Indexed: 01/10/2023] Open
Abstract
The ubiquitin proteasome system (UPS) plays an imperative role in many critical cellular processes, frequently by mediating the selective degradation of misfolded and damaged proteins and also by playing a non-degradative role especially important as in many signaling pathways. Over the last three decades, accumulated evidence indicated that UPS proteins are primal modulators of cell cycle progression, DNA replication, and repair, transcription, immune responses, and apoptosis. Comparatively, latest studies have demonstrated a substantial complexity by the UPS regulation in the heart. In addition, various UPS proteins especially ubiquitin ligases and proteasome have been identified to play a significant role in the cardiac development and dynamic physiology of cardiac pathologies such as ischemia/reperfusion injury, hypertrophy, and heart failure. However, our understanding of the contribution of UPS dysfunction in the plausible development of cardiac pathophysiology and the complete list of UPS proteins regulating these afflictions is still in infancy. The recent emergence of the roles of TNF receptor-associated factor (TRAFs) and deubiquitinating enzymes (DUBs) superfamily in hypertrophic cardiomyopathy has enhanced our knowledge. In this review, we have mainly compiled the TRAF superfamily of E3 ligases and few DUBs proteins with other well-documented E3 ligases such as MDM2, MuRF-1, Atrogin-I, and TRIM 32 that are specific to myocardial hypertrophy. In this review, we also aim to highlight their expression profile following physiological and pathological stimulation leading to the onset of hypertrophic phenotype in the heart that can serve as biomarkers and the opportunity for the development of novel therapies.
Collapse
Affiliation(s)
- Ishita Gupta
- Structural Immunology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India.,Drug Discovery Research Center, Translational Health Science and Technology Institute, Faridabad, India
| | - Nishant K Varshney
- Drug Discovery Research Center, Translational Health Science and Technology Institute, Faridabad, India
| | - Sameena Khan
- Drug Discovery Research Center, Translational Health Science and Technology Institute, Faridabad, India
| |
Collapse
|
31
|
Over-expression of a cardiac-specific human dopamine D5 receptor mutation in mice causes a dilated cardiomyopathy through ROS over-generation by NADPH oxidase activation and Nrf2 degradation. Redox Biol 2018; 19:134-146. [PMID: 30153650 PMCID: PMC6111036 DOI: 10.1016/j.redox.2018.07.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 07/10/2018] [Accepted: 07/12/2018] [Indexed: 12/26/2022] Open
Abstract
Dilated cardiomyopathy (DCM) is a severe disorder caused by medications or genetic mutations. D5 dopamine receptor (D5R) gene knockout (D5-/-) mice have cardiac hypertrophy and high blood pressure. To investigate the role and mechanism by which the D5R regulates cardiac function, we generated cardiac-specific human D5R F173L(hD5F173L-TG) and cardiac-specific human D5R wild-type (hD5WT-TG) transgenic mice, and H9c2 cells stably expressing hD5F173L and hD5WT. We found that cardiac-specific hD5F173L-TG mice, relative to hD5WT-TG mice, presented with DCM and increased cardiac expression of cardiac injury markers, NADPH oxidase activity, Nrf2 degradation, and activated ERK1/2/JNK pathway. H9c2-hD5F173L cells also had an increase in NADPH oxidase activity, Nrf2 degradation, and phospho-JNK (p-JNK) expression. A Nrf2 inhibitor also increased p-JNK expression in H9c2-hD5F173L cells but not in H9c2-hD5WT cells. We suggest that the D5R may play an important role in the preservation of normal heart function by inhibiting the production of reactive oxygen species, via inhibition of NADPH oxidase, Nrf2 degradation, and ERK1/2/JNK pathways.
Collapse
|
32
|
Shibu MA, Kuo CH, Chen BC, Ju DT, Chen RJ, Lai CH, Huang PJ, Viswanadha VP, Kuo WW, Huang CY. Oolong tea prevents cardiomyocyte loss against hypoxia by attenuating p-JNK mediated hypertrophy and enhancing P-IGF1R, p-akt, and p-Bad ser136 activity and by fortifying NRF2 antioxidation system. ENVIRONMENTAL TOXICOLOGY 2018; 33:220-233. [PMID: 29139225 DOI: 10.1002/tox.22510] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 10/24/2017] [Accepted: 10/25/2017] [Indexed: 06/07/2023]
Abstract
Tea, the most widely consumed natural beverage has been associated with reduced mortality risk from cardiovascular disease. Oolong tea is a partially fermented tea containing high levels of catechins, their degree of oxidation varies between 20%-80% causing differences in their active metabolites. In this study we examined the effect of oolong tea extract (OTE) obtained by oxidation at low-temperature for short-time against hypoxic injury and found that oolong tea provides cyto-protective effects by suppressing the JNK mediated hypertrophic effects and by enhancing the innate antioxidant mechanisms in neonatal cardiomyocytes and in H9c2 cells. OTE effectively attenuates 24 h hypoxia-triggered cardiomyocyte loss by suppressing caspase-3-cleavage and apoptosis in a dose-dependent manner. OTE also enhances the IGFIR/p-Akt associated survival-mechanism involving the elevation of p-Badser136 in a dose-dependent manner to aid cellular adaptations against hypoxic challenge. The results show the effects and mechanism of Oolong tea to provide cardio-protective benefits during hypoxic conditions.
Collapse
Affiliation(s)
| | - Chia-Hua Kuo
- Department of Sports Sciences, University of Taipei, Taipei, Taiwan
| | - Bih-Cheng Chen
- School of Post-Baccalaureate Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Da-Tong Ju
- Department of Neurological Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Ray-Jade Chen
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chao-Hung Lai
- Division of Cardiology, Department of Internal Medicine, Armed Force Taichung General Hospital, Taichung, Taiwan
| | - Pei-Jane Huang
- Department of Biotechnology, Asia University, Taichung, Taiwan
| | | | - Wei-Wen Kuo
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Chih-Yang Huang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
- Department of Biotechnology, Asia University, Taichung, Taiwan
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
| |
Collapse
|
33
|
Tian C, Gao L, Zimmerman MC, Zucker IH. Myocardial infarction-induced microRNA-enriched exosomes contribute to cardiac Nrf2 dysregulation in chronic heart failure. Am J Physiol Heart Circ Physiol 2018; 314:H928-H939. [PMID: 29373037 DOI: 10.1152/ajpheart.00602.2017] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The imbalance between the synthesis of reactive oxygen species and their elimination by antioxidant defense systems results in macromolecular damage and disruption of cellular redox signaling, affecting cardiac structure and function, thus contributing to contractile dysfunction, myocardial hypertrophy, and fibrosis in chronic heart failure [chronic heart failure (CHF)]. The Kelch-like ECH-associated protein 1-nuclear factor erythroid 2-related factor 2 (Nrf2) pathway is an important antioxidant defense mechanism and is closely associated with oxidative stress-mediated cardiac remodeling in CHF. In the present study, we investigated the regulation of myocardial Nrf2 in the postmyocardial infarction (post-MI) state. Six weeks post-MI, Nrf2 protein was downregulated in the heart, resulting in a decrease of Nrf2-targeted antioxidant enzymes, whereas paradoxically the transcription of Nrf2 was increased, suggesting that translational inhibition of Nrf2 may contribute to the dysregulation in CHF. We therefore hypothesized that microRNAs may be involved in the translational repression of Nrf2 mRNA in the setting of CHF. Using quantitative real-time PCR analysis, we found that three microRNAs, including microRNA-27a, microRNA-28-3p, and microRNA-34a, were highly expressed in the left ventricle of infarcted hearts compared with other organs. Furthermore, in vitro analysis revealed that cultured cardiac myocytes and fibroblasts expressed these three microRNAs in response to TNF-α stimulation. These microRNAs were preferentially incorporated into exosomes and secreted into the extracellular space in which microRNA-enriched exosomes mediated intercellular communication and Nrf2 dysregulation. Taken together, these results suggest that increased local microRNAs induced by MI may contribute to oxidative stress by the inhibition of Nrf2 translation in CHF. NEW & NOTEWORTHY The results of this work provide a novel mechanism mediated by microRNA-enriched exosomes, contributing to the nuclear factor erythroid 2-related factor 2 dysregulation and subsequent oxidative stress. Importantly, these new findings will provide a promising strategy to improve the therapeutic efficacy through targeting nuclear factor erythroid 2-related factor 2-related microRNAs in the chronic heart failure state, which show potentially clinical applications.
Collapse
Affiliation(s)
- Changhai Tian
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center , Omaha, Nebraska
| | - Lie Gao
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center , Omaha, Nebraska
| | - Matthew C Zimmerman
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center , Omaha, Nebraska
| | - Irving H Zucker
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center , Omaha, Nebraska
| |
Collapse
|
34
|
Fei F, Qu J, Li C, Wang X, Li Y, Zhang S. Role of metastasis-induced protein S100A4 in human non-tumor pathophysiologies. Cell Biosci 2017; 7:64. [PMID: 29204268 PMCID: PMC5702147 DOI: 10.1186/s13578-017-0191-1] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2017] [Accepted: 11/20/2017] [Indexed: 12/24/2022] Open
Abstract
S100A4, an important member of the S100 family of proteins, is best known for its significant role in promoting cancer progression and metastasis. In addition to its expression in tumors, upregulation of S100A4 expression has been associated with various non-tumor pathophysiology processes. However, the mechanisms underlying the role of S100A4 remain unclear. Activated “host” cells (fibroblasts, immunocytes, vascular cells, among others) secrete S100A4 into the extracellular space in various non-tumor human disorders, where it executes its biological functions by interacting with intracellular target proteins. However, the exact molecular mechanisms underlying these interactions in different non-tumor pathophysiologies vary, and S100A4 is likely one of the cross-linking factors that acts as common intrinsic constituents of biological mechanisms. Numerous studies have indicated that the S100A4-mediated epithelial–mesenchymal transition plays a vital role in the occurrence and development of various non-tumor pathophysiologies. Epithelial–mesenchymal transition can be categorized into three general subtypes based on the phenotype and function of the output cells. S100A4 regulates tissue fibrosis associated with the type II epithelial–mesenchymal transition via various signaling pathways. Additionally, S100A4 stimulates fibroblasts to secrete fibronectin and collagen, thus forming the structural components of the extracellular matrix (ECM) and stimulating their deposition in tissues, contributing to the formation of a pro-inflammatory niche. Simultaneously, S100A4 enhances the motility of macrophages, neutrophils, and leukocytes and promotes the recruitment and chemotaxis of these inflammatory cells to regulate inflammation and immune functions. S100A4 also exerts a neuroprotective pro-survival effect on neurons by rescuing them from brain injury and participates in angiogenesis by interacting with other target molecules. In this review, we summarize the role of S100A4 in fibrosis, inflammation, immune response, neuroprotection, angiogenesis, and some common non-tumor diseases as well as its possible involvement in molecular pathways and potential clinical value.
Collapse
Affiliation(s)
- Fei Fei
- Nankai University School of Medicine, Nankai University, Tianjin, 300071 People's Republic of China.,Departments of Pathology, Tianjin Union Medical Center, Tianjin, 300121 People's Republic of China
| | - Jie Qu
- Nankai University School of Medicine, Nankai University, Tianjin, 300071 People's Republic of China.,Departments of Pathology, Tianjin Union Medical Center, Tianjin, 300121 People's Republic of China
| | - Chunyuan Li
- Nankai University School of Medicine, Nankai University, Tianjin, 300071 People's Republic of China.,Departments of Pathology, Tianjin Union Medical Center, Tianjin, 300121 People's Republic of China
| | - Xinlu Wang
- Departments of Pathology, Tianjin Union Medical Center, Tianjin, 300121 People's Republic of China.,Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193 People's Republic of China
| | - Yuwei Li
- Departments of Colorectal Surgery, Tianjin Union Medical Center, Tianjin, 300121 People's Republic of China
| | - Shiwu Zhang
- Departments of Pathology, Tianjin Union Medical Center, Tianjin, 300121 People's Republic of China
| |
Collapse
|
35
|
Li Y, Li Y, Shi F, Wang L, Li L, Yang D. Osthole attenuates right ventricular remodeling via decreased myocardial apoptosis and inflammation in monocrotaline-induced rats. Eur J Pharmacol 2017; 818:525-533. [PMID: 29146527 DOI: 10.1016/j.ejphar.2017.11.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 09/08/2017] [Accepted: 11/03/2017] [Indexed: 11/29/2022]
Abstract
Osthole (Ost) is a coumarin that exhibits wide pharmacological effects in the cardiovascular system. However, whether Ost can inhibit apoptosis and inflammation in right ventricle (RV) cardiomyocytes and prevent RV remodeling is not clear. This study was designed to investigate the effect of Ost on RV remodeling and the underlying mechanism. By applying a monocrotaline (MCT)-induced rat model, the effect of Ost on RV remodeling was investigated. Rats were given a single dose of MCT (50mg/kg) subcutaneously (s.c.) to establish the RV remodeling model, followed by treatment with 10 or 20mg/kg Ost via daily gavage for 28 days. The RV pressure was measured, and a histological analysis was performed. The results suggested that Ost remarkably decreased RV pressure and improved myocardial hypertrophy and mitochondrial swelling, vacuolization, and sarcoplasmic reticulum enlargement when compared with the model group. To further investigate the roles of apoptosis and inflammation in the effects of Ost on MCT-induced RV remodeling, apoptosis-related factors and inflammatory-associated factors were examined by western blot. Ost was found to inhibit myocardial apoptosis and inflammation in the RV. Overall, the present results indicate that Ost suppresses the RV remodeling process induced by MCT in rats, which may be at least partially mediated through the reduction of myocardial apoptosis and inflammation.
Collapse
Affiliation(s)
- Yeli Li
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou 563003, China
| | - Yiqi Li
- Department of Pharmacology, Zunyi Medical University, Zhuhai Campus, Zhuhai, Guangdong 519041, China
| | - Fuguo Shi
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou 563003, China
| | - Lina Wang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou 563003, China
| | - Lisheng Li
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou 563003, China
| | - Danli Yang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou 563003, China.
| |
Collapse
|
36
|
Cai L, Fan G, Wang F, Liu S, Li T, Cong X, Chun J, Chen X. Protective Role for LPA 3 in Cardiac Hypertrophy Induced by Myocardial Infarction but Not by Isoproterenol. Front Physiol 2017; 8:356. [PMID: 28611684 PMCID: PMC5447740 DOI: 10.3389/fphys.2017.00356] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 05/15/2017] [Indexed: 01/04/2023] Open
Abstract
Background: We previously reported that lysophosphatidic acid (LPA) promoted cardiomyocyte hypertrophy in vitro via one of its G protein-coupled receptor subtypes, LPA3. In this study, we examined the role of LPA3 in cardiac hypertrophy induced by isoproterenol (ISO) and myocardial infarction. Methods:In vitro, neonatal rat cardiomyocytes (NRCMs) were subjected to LPA3 knocked-down, or pretreated with a β-adrenergic receptor (β-AR) antagonist (propranolol) before LPA/ISO treatment. Cardiomyocyte size and hypertrophic gene (ANP, BNP) mRNA levels were determined. In vivo, [Formula: see text] and wild-type mice were implanted subcutaneously with an osmotic mini-pump containing ISO or vehicle for 2 weeks; echocardiography was performed to determine the heart weight/body weight ratio, cardiomyocyte cross-sectional area, and level of ANP mRNA expression. [Formula: see text] and wild-type mice were subjected to permanent coronary artery ligation or sham surgery for 4 weeks; cardiac function, including the degree of hypertrophy and infarction size, was determined. Results:In vitro, we found that knocked-down LPA3 in NRCMs did not attenuate ISO-induced hypertrophy, and propranolol was unable to abolish LPA-induced hypertrophy. In vivo, chronic ISO infusion caused cardiac hypertrophy in wild-type mice, while hypertrophic responses to ISO infusion were not attenuated in [Formula: see text] mice. However, in a myocardial infarction (MI) model, [Formula: see text] mice exhibited reduced cardiac hypertrophy compared to wild-type mice at 4 weeks post-MI, which was associated with reduced cardiac function and increased infarct size. Conclusions: Our data show that LPA3 appears to play a protective role in myocardial hypertrophy post-MI, but does not appear to be involved in the hypertrophy that occurs in response to β-AR stimulation in vivo and in vitro. These results implicate LPA-LPA3 lipid signaling in cardiac hypertrophy occurring after pathological insults like MI, which presents a new variable in β-AR-independent hypertrophy. Thus, modulation of LPA3 signaling might represent a new strategy for preventing the stressed myocardium from ischemia injury.
Collapse
Affiliation(s)
- Lin Cai
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing, China
| | - Guangpu Fan
- Cardiovascular Surgery Department, FuWai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing, China
| | - Fang Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing, China
| | - Si Liu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing, China
| | - Tiewei Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing, China
| | - Xiangfeng Cong
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing, China
| | - Jerold Chun
- Sanford Burnham Prebys Medical Discovery InstituteLa Jolla, CA, United States
| | - Xi Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing, China
| |
Collapse
|
37
|
Zheng C, Lo CY, Meng Z, Li Z, Zhong M, Zhang P, Lu J, Yang Z, Yan F, Zhang Y, Huang Y, Yao X. Gastrodin Inhibits Store-Operated Ca 2+ Entry and Alleviates Cardiac Hypertrophy. Front Pharmacol 2017; 8:222. [PMID: 28487655 PMCID: PMC5404510 DOI: 10.3389/fphar.2017.00222] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 04/10/2017] [Indexed: 11/13/2022] Open
Abstract
Cardiac hypertrophy is a major risk factor for heart failure, which are among the leading causes of human death. Gastrodin is a small molecule that has been used clinically to treat neurological and vascular diseases for many years without safety issues. In the present study, we examined protective effect of gastrodin against cardiac hypertrophy and explored the underlying mechanism. Phenylephrine and angiotensin II were used to induce cardiac hypertrophy in a mouse model and a cultured cardiomyocyte model. Gastrodin was found to alleviate the cardiac hypertrophy in both models. Mechanistically, gastrodin attenuated the store-operated Ca2+ entry (SOCE) by reducing the expression of STIM1 and Orai1, two key proteins in SOCE, in animal models as well as in cultured cardiomyocyte model. Furthermore, suppressing SOCE by RO2959, Orai1-siRNAs or STIM1-siRNAs markedly attenuated the phenylephrine-induced hypertrophy in cultured cardiomyocyte model. Together, these results showed that gastrodin inhibited cardiac hypertrophy and it also reduced the SOCE via its action on the expression of STIM1 and Orai1. Furthermore, suppression of SOCE could reduce the phenylephrine-induced cardiomyocyte hypertrophy, suggesting that SOCE-STIM1-Orai1 is located upstream of hypertrophy.
Collapse
Affiliation(s)
- Changbo Zheng
- Li Ka Shing Institute of Health Sciences and School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong KongHong Kong, China.,Shenzhen Research Institute, The Chinese University of Hong KongShenzhen, China
| | - Chun-Yin Lo
- Li Ka Shing Institute of Health Sciences and School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong KongHong Kong, China.,Shenzhen Research Institute, The Chinese University of Hong KongShenzhen, China
| | - Zhaoyue Meng
- Li Ka Shing Institute of Health Sciences and School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong KongHong Kong, China.,Shenzhen Research Institute, The Chinese University of Hong KongShenzhen, China.,School of Life Sciences, The Chinese University of Hong KongHong Kong, China
| | - Zhichao Li
- Li Ka Shing Institute of Health Sciences and School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong KongHong Kong, China.,Shenzhen Research Institute, The Chinese University of Hong KongShenzhen, China
| | - Mingkui Zhong
- Li Ka Shing Institute of Health Sciences and School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong KongHong Kong, China
| | - Peng Zhang
- Li Ka Shing Institute of Health Sciences and School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong KongHong Kong, China
| | - Jun Lu
- Li Ka Shing Institute of Health Sciences and School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong KongHong Kong, China.,Shenzhen Research Institute, The Chinese University of Hong KongShenzhen, China
| | - Zhaoxiang Yang
- Institute for Drug Research and Development, Kunming Pharmaceutical CorporationKunming, China
| | - Fuman Yan
- Li Ka Shing Institute of Health Sciences and School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong KongHong Kong, China
| | - Yunting Zhang
- Li Ka Shing Institute of Health Sciences and School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong KongHong Kong, China
| | - Yu Huang
- Li Ka Shing Institute of Health Sciences and School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong KongHong Kong, China
| | - Xiaoqiang Yao
- Li Ka Shing Institute of Health Sciences and School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong KongHong Kong, China.,Shenzhen Research Institute, The Chinese University of Hong KongShenzhen, China
| |
Collapse
|
38
|
Nemska S, Monassier L, Gassmann M, Frossard N, Tavakoli R. Kinetic mRNA Profiling in a Rat Model of Left-Ventricular Hypertrophy Reveals Early Expression of Chemokines and Their Receptors. PLoS One 2016; 11:e0161273. [PMID: 27525724 PMCID: PMC4985150 DOI: 10.1371/journal.pone.0161273] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 07/28/2016] [Indexed: 11/29/2022] Open
Abstract
Left-ventricular hypertrophy (LVH), a risk factor for heart failure and death, is characterized by cardiomyocyte hypertrophy, interstitial cell proliferation, and leukocyte infiltration. Chemokines interacting with G protein-coupled chemokine receptors may play a role in LVH development by promoting recruitment of activated leukocytes or modulating left-ventricular remodeling. Using a pressure overload-induced kinetic model of LVH in rats, we examined during 14 days the expression over time of chemokine and chemokine receptor mRNAs in left ventricles from aortic-banded vs sham-operated animals. Two phases were clearly distinguished: an inflammatory phase (D3-D5) with overexpression of inflammatory genes such as il-1ß, tnfa, nlrp3, and the rela subunit of nf-kb, and a hypertrophic phase (D7-D14) where anp overexpression was accompanied by a heart weight/body weight ratio that increased by more than 20% at D14. No cardiac dysfunction was detectable by echocardiography at the latter time point. Of the 36 chemokines and 20 chemokine receptors analyzed by a Taqman Low Density Array panel, we identified at D3 (the early inflammatory phase) overexpression of mRNAs for the monocyte chemotactic proteins CCL2 (12-fold increase), CCL7 (7-fold increase), and CCL12 (3-fold increase), for the macrophage inflammatory proteins CCL3 (4-fold increase), CCL4 (2-fold increase), and CCL9 (2-fold increase), for their receptors CCR2 (4-fold increase), CCR1 (3-fold increase), and CCR5 (3-fold increase), and for CXCL1 (8-fold increase) and CXCL16 (2-fold increase). During the hypertrophic phase mRNA expression of chemokines and receptors returned to the baseline levels observed at D0. Hence, this first exhaustive study of chemokine and chemokine receptor mRNA expression kinetics reports early expression of monocyte/macrophage-related chemokines and their receptors during the development of LVH in rats, followed by regulation of inflammation as LVH progresses.
Collapse
Affiliation(s)
- Simona Nemska
- Institute of Veterinary Physiology and Zurich Center of Integrative Human Physiology, University of Zurich, Zurich, Switzerland
- Laboratoire d’Innovation Thérapeutique, UMR7200, Université de Strasbourg—CNRS, Strasbourg, France
| | - Laurent Monassier
- Laboratoire de Neurobiologie et Pharmacologie Cardiovasculaire EA7296, Fédération de Médecine Translationnelle, Université de Strasbourg, Strasbourg, France
| | - Max Gassmann
- Institute of Veterinary Physiology and Zurich Center of Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Nelly Frossard
- Laboratoire d’Innovation Thérapeutique, UMR7200, Université de Strasbourg—CNRS, Strasbourg, France
- * E-mail: (RT); (NF)
| | - Reza Tavakoli
- Institute of Veterinary Physiology and Zurich Center of Integrative Human Physiology, University of Zurich, Zurich, Switzerland
- Department of Cardiac Surgery, Canton Hospital Lucerne, Lucerne, Switzerland
- * E-mail: (RT); (NF)
| |
Collapse
|
39
|
Huang J, Chen L, Yao Y, Tang C, Ding J, Fu C, Li H, Ma G. Pivotal Role of Regulator of G-protein Signaling 12 in Pathological Cardiac Hypertrophy. Hypertension 2016; 67:1228-36. [PMID: 27091895 DOI: 10.1161/hypertensionaha.115.06877] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Accepted: 03/19/2016] [Indexed: 11/16/2022]
Abstract
Cardiac hypertrophy is a major predictor of heart failure and is regulated by diverse signaling pathways. As a typical multi-domain member of the regulator of G-protein signaling (RGS) family, RGS12 plays a regulatory role in various signaling pathways. However, the precise effect of RGS12 on cardiac hypertrophy remains largely unknown. In this study, we observed increased expression of RGS12 in the development of pathological cardiac hypertrophy and heart failure. We then generated genetically engineered mice and neonatal rat cardiomyocytes to investigate the effects of RGS12 during this pathological process. Four weeks after aortic banding, RGS12-deficient hearts showed decreased cardiomyocyte cross area (374.7±43.2 μm(2) versus 487.1±47.9 μm(2) in controls; P<0.05) with preserved fractional shortening (43.0±3.4% versus 28.4±2.2% in controls; P<0.05), whereas RGS12-overexpressing hearts exhibited increased cardiomyocyte cross area (582.4±46.7 μm(2) versus 474.8±40.0 μm(2) in controls; P<0.05) and reduced fractional shortening (20.8±4.1% versus 28.6±3.2% in controls; P<0.05). RGS12 also contributed to angiotensin II-induced hypertrophy in isolated cardiomyocytes. Mechanistically, our data indicated that the activation of MEK1/2-ERK1/2 signaling may be responsible for the prohypertrophic action of RGS12. In addition, the requirement of the MEK1/2-ERK1/2 signaling for RGS12-mediated cardiac hypertrophy was confirmed in rescue experiments using the MEK1/2-specific inhibitor U0126. In conclusion, our findings provide a novel diagnostic and therapeutic target for pathological cardiac hypertrophy and heart failure.
Collapse
Affiliation(s)
- Jia Huang
- From the Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, Nanjing, Jiangsu, PR China (J.H., L.C., Y.Y., C.T., J.D., C.F., G.M.); Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China (H.L.); and Cardiovascular Research Institute of Wuhan University, Wuhan, PR China (H.L.)
| | - Lijuan Chen
- From the Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, Nanjing, Jiangsu, PR China (J.H., L.C., Y.Y., C.T., J.D., C.F., G.M.); Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China (H.L.); and Cardiovascular Research Institute of Wuhan University, Wuhan, PR China (H.L.)
| | - Yuyu Yao
- From the Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, Nanjing, Jiangsu, PR China (J.H., L.C., Y.Y., C.T., J.D., C.F., G.M.); Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China (H.L.); and Cardiovascular Research Institute of Wuhan University, Wuhan, PR China (H.L.)
| | - Chengchun Tang
- From the Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, Nanjing, Jiangsu, PR China (J.H., L.C., Y.Y., C.T., J.D., C.F., G.M.); Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China (H.L.); and Cardiovascular Research Institute of Wuhan University, Wuhan, PR China (H.L.)
| | - Jiandong Ding
- From the Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, Nanjing, Jiangsu, PR China (J.H., L.C., Y.Y., C.T., J.D., C.F., G.M.); Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China (H.L.); and Cardiovascular Research Institute of Wuhan University, Wuhan, PR China (H.L.)
| | - Cong Fu
- From the Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, Nanjing, Jiangsu, PR China (J.H., L.C., Y.Y., C.T., J.D., C.F., G.M.); Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China (H.L.); and Cardiovascular Research Institute of Wuhan University, Wuhan, PR China (H.L.)
| | - Hongliang Li
- From the Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, Nanjing, Jiangsu, PR China (J.H., L.C., Y.Y., C.T., J.D., C.F., G.M.); Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China (H.L.); and Cardiovascular Research Institute of Wuhan University, Wuhan, PR China (H.L.)
| | - Genshan Ma
- From the Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, Nanjing, Jiangsu, PR China (J.H., L.C., Y.Y., C.T., J.D., C.F., G.M.); Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China (H.L.); and Cardiovascular Research Institute of Wuhan University, Wuhan, PR China (H.L.).
| |
Collapse
|
40
|
Yang H, Schmidt LP, Wang Z, Yang X, Shao Y, Borg TK, Markwald R, Runyan R, Gao BZ. Dynamic Myofibrillar Remodeling in Live Cardiomyocytes under Static Stretch. Sci Rep 2016; 6:20674. [PMID: 26861590 PMCID: PMC4748238 DOI: 10.1038/srep20674] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 01/11/2016] [Indexed: 12/21/2022] Open
Abstract
An increase in mechanical load in the heart causes cardiac hypertrophy, either physiologically (heart development, exercise and pregnancy) or pathologically (high blood pressure and heart-valve regurgitation). Understanding cardiac hypertrophy is critical to comprehending the mechanisms of heart development and treatment of heart disease. However, the major molecular event that occurs during physiological or pathological hypertrophy is the dynamic process of sarcomeric addition, and it has not been observed. In this study, a custom-built second harmonic generation (SHG) confocal microscope was used to study dynamic sarcomeric addition in single neonatal CMs in a 3D culture system under acute, uniaxial, static, sustained stretch. Here we report, for the first time, live-cell observations of various modes of dynamic sarcomeric addition (and how these real-time images compare to static images from hypertrophic hearts reported in the literature): 1) Insertion in the mid-region or addition at the end of a myofibril; 2) Sequential addition with an existing myofibril as a template; and 3) Longitudinal splitting of an existing myofibril. The 3D cell culture system developed on a deformable substrate affixed to a stretcher and the SHG live-cell imaging technique are unique tools for real-time analysis of cultured models of hypertrophy.
Collapse
Affiliation(s)
- Huaxiao Yang
- Department of Bioengineering, Clemson University, Clemson, SC, USA
| | - Lucas P Schmidt
- Department of Bioengineering, Clemson University, Clemson, SC, USA
| | - Zhonghai Wang
- Department of Bioengineering, Clemson University, Clemson, SC, USA
| | - Xiaoqi Yang
- Department of Bioengineering, Clemson University, Clemson, SC, USA
| | - Yonghong Shao
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen, China
| | - Thomas K Borg
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Roger Markwald
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Raymond Runyan
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
| | - Bruce Z Gao
- Department of Bioengineering, Clemson University, Clemson, SC, USA
| |
Collapse
|
41
|
High concentrations of H2O2 trigger hypertrophic cascade and phosphatase and tensin homologue (PTEN) glutathionylation in H9c2 cardiomyocytes. Exp Mol Pathol 2016; 100:199-206. [DOI: 10.1016/j.yexmp.2016.01.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 11/17/2015] [Accepted: 01/06/2016] [Indexed: 11/20/2022]
|
42
|
Berthiaume J, Kirk J, Ranek M, Lyon R, Sheikh F, Jensen B, Hoit B, Butany J, Tolend M, Rao V, Willis M. Pathophysiology of Heart Failure and an Overview of Therapies. Cardiovasc Pathol 2016. [DOI: 10.1016/b978-0-12-420219-1.00008-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
43
|
Cominacini L, Mozzini C, Garbin U, Pasini A, Stranieri C, Solani E, Vallerio P, Tinelli IA, Fratta Pasini A. Endoplasmic reticulum stress and Nrf2 signaling in cardiovascular diseases. Free Radic Biol Med 2015; 88:233-242. [PMID: 26051167 DOI: 10.1016/j.freeradbiomed.2015.05.027] [Citation(s) in RCA: 139] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 05/14/2015] [Accepted: 05/17/2015] [Indexed: 12/30/2022]
Abstract
Various cellular perturbations implicated in the pathophysiology of human diseases, including cardiovascular and neurodegenerative diseases, diabetes mellitus, obesity, and liver diseases, can alter endoplasmic reticulum (ER) function and lead to the abnormal accumulation of misfolded proteins. This situation configures the so-called ER stress, a form of intracellular stress that occurs whenever the protein-folding capacity of the ER is overwhelmed. Reduction in blood flow as a result of atherosclerotic coronary artery disease causes tissue hypoxia, a condition that induces protein misfolding and ER stress. In addition, ER stress has an important role in cardiac hypertrophy mainly in the transition to heart failure (HF). ER transmembrane sensors detect the accumulation of unfolded proteins and activate transcriptional and translational pathways that deal with unfolded and misfolded proteins, known as the unfolded protein response (UPR). Once the UPR fails to control the level of unfolded and misfolded proteins in the ER, ER-initiated apoptotic signaling is induced. Furthermore, there is considerable evidence that implicates the presence of oxidative stress and subsequent related cellular damage as an initial cause of injury to the myocardium after ischemia/reperfusion (I/R) and in cardiac hypertrophy secondary to pressure overload. Oxidative stress is counterbalanced by complex antioxidant defense systems regulated by a series of multiple pathways, including the UPR, to ensure that the response to oxidants is adequate. Nuclear factor-E2-related factor (Nrf2) is an emerging regulator of cellular resistance to oxidants; Nrf2 is strictly interrelated with the UPR sensor called pancreatic endoplasmic reticulum kinase. A series of studies has shown that interventions against ER stress and Nrf2 activation reduce myocardial infarct size and cardiac hypertrophy in the transition to HF in animals exposed to I/R injury and pressure overload, respectively. Finally, recent data showed that Nrf2/antioxidant-response element pathway activation may be of importance also in ischemic preconditioning, a phenomenon in which the heart is subjected to one or more episodes of nonlethal myocardial I/R before the sustained coronary artery occlusion.
Collapse
Affiliation(s)
- Luciano Cominacini
- Section of Internal Medicine, Department of Medicine, University of Verona, 37134 Verona, Italy.
| | - Chiara Mozzini
- Section of Internal Medicine, Department of Medicine, University of Verona, 37134 Verona, Italy
| | - Ulisse Garbin
- Section of Internal Medicine, Department of Medicine, University of Verona, 37134 Verona, Italy
| | - Andrea Pasini
- Section of Internal Medicine, Department of Medicine, University of Verona, 37134 Verona, Italy
| | - Chiara Stranieri
- Section of Internal Medicine, Department of Medicine, University of Verona, 37134 Verona, Italy
| | - Erika Solani
- Section of Internal Medicine, Department of Medicine, University of Verona, 37134 Verona, Italy
| | - Paola Vallerio
- Section of Internal Medicine, Department of Medicine, University of Verona, 37134 Verona, Italy
| | | | - Anna Fratta Pasini
- Section of Internal Medicine, Department of Medicine, University of Verona, 37134 Verona, Italy
| |
Collapse
|
44
|
Nehra S, Bhardwaj V, Ganju L, Saraswat D. Nanocurcumin Prevents Hypoxia Induced Stress in Primary Human Ventricular Cardiomyocytes by Maintaining Mitochondrial Homeostasis. PLoS One 2015; 10:e0139121. [PMID: 26406246 PMCID: PMC4583454 DOI: 10.1371/journal.pone.0139121] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 09/09/2015] [Indexed: 01/01/2023] Open
Abstract
Hypoxia induced oxidative stress incurs pathophysiological changes in hypertrophied cardiomyocytes by promoting translocation of p53 to mitochondria. Here, we investigate the cardio-protective efficacy of nanocurcumin in protecting primary human ventricular cardiomyocytes (HVCM) from hypoxia induced damages. Hypoxia induced hypertrophy was confirmed by FITC-phenylalanine uptake assay, atrial natriuretic factor (ANF) levels and cell size measurements. Hypoxia induced translocation of p53 was investigated by using mitochondrial membrane permeability transition pore blocker cyclosporin A (blocks entry of p53 to mitochondria) and confirmed by western blot and immunofluorescence. Mitochondrial damage in hypertrophied HVCM cells was evaluated by analysing bio-energetic, anti-oxidant and metabolic function and substrate switching form lipids to glucose. Nanocurcumin prevented translocation of p53 to mitochondria by stabilizing mitochondrial membrane potential and de-stressed hypertrophied HVCM cells by significant restoration in lactate, acetyl-coenzyme A, pyruvate and glucose content along with lactate dehydrogenase (LDH) and 5' adenosine monophosphate-activated protein kinase (AMPKα) activity. Significant restoration in glucose and modulation of GLUT-1 and GLUT-4 levels confirmed that nanocurcumin mediated prevention of substrate switching. Nanocurcumin prevented of mitochondrial stress as confirmed by c-fos/c-jun/p53 signalling. The data indicates decrease in p-300 histone acetyl transferase (HAT) mediated histone acetylation and GATA-4 activation as pharmacological targets of nanocurcumin in preventing hypoxia induced hypertrophy. The study provides an insight into propitious therapeutic effects of nanocurcumin in cardio-protection and usability in clinical applications.
Collapse
Affiliation(s)
- Sarita Nehra
- Experimental Biology Division, Department of Experimental Biology, Defence Institute of Physiology and Allied Science, Defence Research and Development Organization, Timarpur, New Delhi, India
| | - Varun Bhardwaj
- Experimental Biology Division, Department of Experimental Biology, Defence Institute of Physiology and Allied Science, Defence Research and Development Organization, Timarpur, New Delhi, India
| | - Lilly Ganju
- Experimental Biology Division, Department of Experimental Biology, Defence Institute of Physiology and Allied Science, Defence Research and Development Organization, Timarpur, New Delhi, India
| | - Deepika Saraswat
- Experimental Biology Division, Department of Experimental Biology, Defence Institute of Physiology and Allied Science, Defence Research and Development Organization, Timarpur, New Delhi, India
| |
Collapse
|
45
|
Wang Y, Li ZC, Zhang P, Poon E, Kong CW, Boheler KR, Huang Y, Li RA, Yao X. Nitric Oxide-cGMP-PKG Pathway Acts on Orai1 to Inhibit the Hypertrophy of Human Embryonic Stem Cell-Derived Cardiomyocytes. Stem Cells 2015; 33:2973-84. [PMID: 26269433 DOI: 10.1002/stem.2118] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 07/15/2015] [Indexed: 11/08/2022]
Abstract
Cardiac hypertrophy is an abnormal enlargement of heart muscle. It frequently results in congestive heart failure, which is a leading cause of human death. Previous studies demonstrated that the nitric oxide (NO), cyclic GMP (cGMP), and protein kinase G (PKG) signaling pathway can inhibit cardiac hypertrophy and thus improve cardiac function. However, the underlying mechanisms are not fully understood. Here, based on the human embryonic stem cell-derived cardiomyocyte (hESC-CM) model system, we showed that Orai1, the pore-forming subunit of store-operated Ca(2+) entry (SOCE), is the downstream effector of PKG. Treatment of hESC-CMs with an α-adrenoceptor agonist phenylephrine (PE) caused a marked hypertrophy, which was accompanied by an upregulation of Orai1. Moreover, suppression of Orai1 expression/activity using Orai1-siRNAs or a dominant-negative construct Orai1(G98A) inhibited the hypertrophy, suggesting that Orai1-mediated SOCE is indispensable for the PE-induced hypertrophy of hESC-CMs. In addition, the hypertrophy was inhibited by NO and cGMP via activating PKG. Importantly, substitution of Ala for Ser(34) in Orai1 abolished the antihypertrophic effects of NO, cGMP, and PKG. Furthermore, PKG could directly phosphorylate Orai1 at Ser(34) and thus prevent Orai1-mediated SOCE. Together, we conclude that NO, cGMP, and PKG inhibit the hypertrophy of hESC-CMs via PKG-mediated phosphorylation on Orai1-Ser-34. These results provide novel mechanistic insights into the action of cGMP-PKG-related antihypertrophic agents, such as NO donors and sildenafil.
Collapse
Affiliation(s)
- Y Wang
- Li Ka Shing Institute of Health Sciences and School of Biomedical Sciences, Faculty of Medicine, the Chinese University of Hong Kong, Hong Kong, People's Republic of China.,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, People's Republic of China.,Department of Hematology, The 3rd Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Z C Li
- Li Ka Shing Institute of Health Sciences and School of Biomedical Sciences, Faculty of Medicine, the Chinese University of Hong Kong, Hong Kong, People's Republic of China.,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, People's Republic of China
| | - P Zhang
- Li Ka Shing Institute of Health Sciences and School of Biomedical Sciences, Faculty of Medicine, the Chinese University of Hong Kong, Hong Kong, People's Republic of China.,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, People's Republic of China
| | - E Poon
- Stem Cell and Regenerative Medicine Consortium, The University of Hong Kong, Hong Kong, People's Republic of China.,The Department of Physiology, The University of Hong Kong, Hong Kong, People's Republic of China
| | - C W Kong
- Stem Cell and Regenerative Medicine Consortium, The University of Hong Kong, Hong Kong, People's Republic of China
| | - K R Boheler
- Stem Cell and Regenerative Medicine Consortium, The University of Hong Kong, Hong Kong, People's Republic of China.,The Department of Physiology, The University of Hong Kong, Hong Kong, People's Republic of China
| | - Y Huang
- Li Ka Shing Institute of Health Sciences and School of Biomedical Sciences, Faculty of Medicine, the Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - R A Li
- Stem Cell and Regenerative Medicine Consortium, The University of Hong Kong, Hong Kong, People's Republic of China
| | - X Yao
- Li Ka Shing Institute of Health Sciences and School of Biomedical Sciences, Faculty of Medicine, the Chinese University of Hong Kong, Hong Kong, People's Republic of China.,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, People's Republic of China
| |
Collapse
|
46
|
Xia J, Zhang Y, Xin L, Kong S, Chen Y, Yang S, Li K. Global Transcriptomic Profiling of Cardiac Hypertrophy and Fatty Heart Induced by Long-Term High-Energy Diet in Bama Miniature Pigs. PLoS One 2015; 10:e0132420. [PMID: 26161779 PMCID: PMC4498776 DOI: 10.1371/journal.pone.0132420] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 06/13/2015] [Indexed: 12/31/2022] Open
Abstract
A long-term high-energy diet affects human health and leads to obesity and metabolic syndrome in addition to cardiac steatosis and hypertrophy. Ectopic fat accumulation in the heart has been demonstrated to be a risk factor for heart disorders, but the molecular mechanism of heart disease remains largely unknown. Bama miniature pigs were fed a high-fat, high-sucrose diet (HFHSD) for 23 months. These pigs developed symptoms of metabolic syndrome and showed cardiac steatosis and hypertrophy with a greatly increased body weight (2.73-fold, P<0.01), insulin level (4.60-fold, P<0.01), heart weight (1.82-fold, P<0.05) and heart volume (1.60-fold, P<0.05) compared with the control pigs. To understand the molecular mechanisms of cardiac steatosis and hypertrophy, nine pig heart cRNA samples were hybridized to porcine GeneChips. Microarray analyses revealed that 1,022 genes were significantly differentially expressed (P<0.05, ≥1.5-fold change), including 591 up-regulated and 431 down-regulated genes in the HFHSD group relative to the control group. KEGG analysis indicated that the observed heart disorder involved the signal transduction-related MAPK, cytokine, and PPAR signaling pathways, energy metabolism-related fatty acid and oxidative phosphorylation signaling pathways, heart function signaling-related focal adhesion, axon guidance, hypertrophic cardiomyopathy and actin cytoskeleton signaling pathways, inflammation and apoptosis pathways, and others. Quantitative RT-PCR assays identified several important differentially expressed heart-related genes, including STAT3, ACSL4, ATF4, FADD, PPP3CA, CD74, SLA-8, VCL, ACTN2 and FGFR1, which may be targets of further research. This study shows that a long-term, high-energy diet induces obesity, cardiac steatosis, and hypertrophy and provides insights into the molecular mechanisms of hypertrophy and fatty heart to facilitate further research.
Collapse
Affiliation(s)
- Jihan Xia
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, P. R. China
| | - Yuanyuan Zhang
- College of Veterinary Medicine, China Agricultural University, Beijing, P. R. China
| | - Leilei Xin
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, P. R. China
| | - Siyuan Kong
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, P. R. China
| | - Yaoxing Chen
- College of Veterinary Medicine, China Agricultural University, Beijing, P. R. China
| | - Shulin Yang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, P. R. China
- * E-mail:
| | - Kui Li
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, P. R. China
- Agricultural Genomes Institute at Shenzhen, CAAS, Shenzhen, 518120, P.R. China
| |
Collapse
|
47
|
Abdelhamid G, El-Kadi AOS. Buthionine sulfoximine, an inhibitor of glutathione biosynthesis, induces expression of soluble epoxide hydrolase and markers of cellular hypertrophy in a rat cardiomyoblast cell line: roles of the NF-κB and MAPK signaling pathways. Free Radic Biol Med 2015; 82:1-12. [PMID: 25614461 DOI: 10.1016/j.freeradbiomed.2015.01.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 01/09/2015] [Accepted: 01/10/2015] [Indexed: 01/04/2023]
Abstract
Evidence suggests that upregulation of soluble epoxide hydrolase (sEH) is associated with the development of myocardial infarction, dilated cardiomyopathy, cardiac hypertrophy, and heart failure. However, the upregulation mechanism is still unknown. In this study, we treated H9C2 cells with buthionine sulfoximine (BSO) to explore whether oxidative stress upregulates sEH gene expression and to identify the molecular and cellular mechanisms behind this upregulatory response. Real-time PCR and Western blot analyses were used to measure mRNA and protein expression, respectively. We demonstrated that BSO significantly upregulated sEH at mRNA levels in a concentration- and time-dependent manner, leading to a significant increase in the cellular hypertrophic markers, atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP). Furthermore, BSO significantly increased the cytosolic phosphorylated IκB-α and translocation of NF-κB p50 subunits, as measured by Western blot analysis. This level of translocation was paralleled by an increase in the DNA-binding activity of NF-κB P50 subunits. Moreover, our results demonstrated that pretreatment with the NF-κB inhibitor PDTC significantly inhibited BSO-mediated induction of sEH and cellular hypertrophic marker gene expression in a dose-dependent manner. Additionally, mitogen-activated protein kinases (MAPKs) were transiently phosphorylated by BSO treatment. To understand further the role of MAPKs pathway in BSO-mediated induction of sEH mRNA, we examined the role of extracellular signal-regulated kinase (ERK), c-JunN-terminal kinase (JNK), and p38 MAPK. Indeed, treatment with the MEK/ERK signal transduction inhibitor, PD98059, partially blocked the activation of IκB-α and translocation of NF-κB p50 subunits induced by BSO. Moreover, pretreatment with MEK/ERK signal transduction inhibitors, PD98059 and U0126, significantly inhibited BSO-mediated induction of sEH and cellular hypertrophic marker gene expression. These results clearly demonstrated that the NF-κB signaling pathway is involved in BSO-mediated induction of sEH gene expression, and appears to be associated with the activation of the MAPK pathway. Furthermore, our findings provide a strong link between sEH-induced cardiac dysfunction and involvement of NF-κB in the development of cellular hypertrophy.
Collapse
Affiliation(s)
- Ghada Abdelhamid
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada T6G 2N8
| | - Ayman O S El-Kadi
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada T6G 2N8.
| |
Collapse
|
48
|
Nehra S, Bhardwaj V, Kalra N, Ganju L, Bansal A, Saxena S, Saraswat D. Nanocurcumin protects cardiomyoblasts H9c2 from hypoxia-induced hypertrophy and apoptosis by improving oxidative balance. J Physiol Biochem 2015; 71:239-51. [PMID: 25846484 DOI: 10.1007/s13105-015-0405-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 03/19/2015] [Indexed: 12/15/2022]
Abstract
Hypoxia-induced cardiomyocyte hypertrophy is evident; however, the distinct molecular mechanism underlying the oxidative stress-mediated damages to cardiomyocytes remains unknown. Curcumin (diferuloylmethane) is known for anti-hypertrophic effects, but low bioavailability makes it unsuitable to exploit its pharmacological properties. We assessed the efficacy of nanotized curcumin, i.e. nanocurcumin, in ameliorating hypoxia-induced hypertrophy and apoptosis in H9c2 cardiomyoblasts and compared it to curcumin. H9c2 cardiomyoblasts were challenged with 0.5 % oxygen, for 24 h to assess hypoxia-induced oxidative damage, hypertrophy and consequent apoptosis. The molecular mechanism underlying the protective efficacy of nanocurcumin was evaluated in regulating Raf-1/Erk-1/2 apoptosis by caspase-3/-7 pathway and oxidative stress. Nanocurcumin ameliorated hypoxia-induced hypertrophy and apoptosis in H9c2 cells significantly (p ≤ 0.01), by downregulating atrial natriuretic factor expression, caspase-3/-7 activation, oxidative stress and stabilizing hypoxia-inducible factor-1α (HIF-1α) better than curcumin. Nanocurcumin provides insight into its use as a potential candidate in curing hypoxia-induced cardiac pathologies by restoring oxidative balance.
Collapse
Affiliation(s)
- Sarita Nehra
- Experimental Biology Division, Department of Experimental Biology, Defence Institute of Physiology and Allied Science, Defence Research and Development Organization, Lucknow Road, Timarpur, New Delhi, India
| | | | | | | | | | | | | |
Collapse
|
49
|
Guo H, Liu B, Hou L, The E, Li G, Wang D, Jie Q, Che W, Wei Y. The role of mAKAPβ in the process of cardiomyocyte hypertrophy induced by angiotensin II. Int J Mol Med 2015; 35:1159-68. [PMID: 25739102 PMCID: PMC4380120 DOI: 10.3892/ijmm.2015.2119] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 02/04/2015] [Indexed: 12/16/2022] Open
Abstract
Angiotensin II (AngII) is the central product of the renin-angiotensin system (RAS) and this octapeptide contributes to the pathophysiology of cardiac hypertrophy and remodeling. mAKAPβ is an A-kinase anchoring protein (AKAP) that has the function of binding to the regulatory subunit of protein kinase A (PKA) and confining the holoenzyme to discrete locations within the cell. In this study, we aimed to investigate the role of mAKAPβ in AngII‑induced cardiomyocyte hypertrophy and the possible mechanisms involved. Cultured cardiomyocytes from neonatal rats were treated with AngII. Subsequently, the morphology of the cardiomyocytes was observed and the expression of mAKAPβ and cardiomyocyte hypertrophic markers was measured. mAKAPβ‑shRNA was constructed for RNA interference; the expression of mAKAPβ and hypertrophic markers, the cell surface area and the [3H]Leucine incorporation rate in the AngII‑treated rat cardiomyocytes were detected following RNA interference. Simultaneously, changes in the expression levels of phosphorylated extracellular signal-regulated kinase (p-ERK)2 in the cardiomyocytes were assessed. The cell size of the AngII-treated cardiaomyocytes was significantly larger than that of the untreated cardiomyocytes. The expression of hypertrophic markers and p-ERK2, the cell surface area and the [3H]Leucine incorporation rate were all significantly increased in the AngII‑treated cells. However, the expression of mAKAPβ remained unaltered in this process. RNA interference simultaneously inhibited the protein expression of mAKAPβ and p‑ERK2, and the hypertrophy of the cardiomyocytes induced by AngII was attenuated. These results demonstrate that AngII induces hypertrophy in cardiomyocytes, and mAKAPβ is possibly involved in this process. The effects of mAKAPβ on AngII‑induced cardiomyocyte hypertrophy may be associated with p-ERK2 expression.
Collapse
Affiliation(s)
- Huixin Guo
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Baoxin Liu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Lei Hou
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Erlinda The
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Gang Li
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Dongzhi Wang
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Qiqiang Jie
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Wenliang Che
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Yidong Wei
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| |
Collapse
|
50
|
Different effects of prolonged β-adrenergic stimulation on heart and cerebral artery. Integr Med Res 2014; 3:204-210. [PMID: 28664099 PMCID: PMC5481746 DOI: 10.1016/j.imr.2014.10.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Revised: 10/01/2014] [Accepted: 10/01/2014] [Indexed: 01/19/2023] Open
Abstract
The aim of this review was to understand the effects of β-adrenergic stimulation on oxidative stress, structural remodeling, and functional alterations in the heart and cerebral artery. Diverse stimuli activate the sympathetic nervous system, leading to increased levels of catecholamines. Long-term overstimulation of the β-adrenergic receptor (βAR) in response to catecholamines causes cardiovascular diseases, including cardiac hypertrophy, stroke, coronary artery disease, and heart failure. Although catecholamines have identical sites of action in the heart and cerebral artery, the structural and functional modifications differentially activate intracellular signaling cascades. βAR-stimulation can increase oxidative stress in the heart and cerebral artery, but has also been shown to induce different cytoskeletal and functional modifications by modulating various components of the βAR signal transduction pathways. Stimulation of βAR leads to cardiac dysfunction due to an overload of intracellular Ca2+ in cardiomyocytes. However, this stimulation induces vascular dysfunction through disruption of actin cytoskeleton in vascular smooth muscle cells. Many studies have shown that excessive concentrations of catecholamines during stressful conditions can produce coronary spasms or arrhythmias by inducing Ca2+-handling abnormalities and impairing energy production in mitochondria, In this article, we highlight the different fates caused by excessive oxidative stress and disruptions in the cytoskeletal proteome network in the heart and the cerebral artery in responsed to prolonged βAR-stimulation.
Collapse
|